101
|
Reactive Astrocytes as Drug Target in Alzheimer's Disease. BIOMED RESEARCH INTERNATIONAL 2018; 2018:4160247. [PMID: 29888263 PMCID: PMC5977027 DOI: 10.1155/2018/4160247] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/20/2018] [Accepted: 04/10/2018] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease is a neurodegenerative disease characterized by deposition of extracellular amyloid-β, intracellular neurofibrillary tangles, and loss of cortical neurons. However, the mechanism underlying neurodegeneration in Alzheimer's disease (AD) remains to be explored. Many of the researches on AD have been primarily focused on neuronal changes. Current research, however, broadens to give emphasis on the importance of nonneuronal cells, such as astrocytes. Astrocytes play fundamental roles in several cerebral functions and their dysfunctions promote neurodegeneration and, eventually, retraction of neuronal synapses, which leads to cognitive deficits found in AD. Astrocytes become reactive as a result of deposition of Aβ, which in turn have detrimental consequences, including decreased glutamate uptake due to reduced expression of uptake transporters, altered energy metabolism, altered ion homeostasis (K+ and Ca+), increased tonic inhibition, and increased release of cytokines and inflammatory mediators. In this review, recent insights on the involvement of, tonic inhibition, astrocytic glutamate transporters and aquaporin in the pathogenesis of Alzheimer's disease are provided. Compounds which increase expression of GLT1 have showed efficacy for AD in preclinical studies. Tonic inhibition mediated by GABA could also be a promising target and drugs that block the GABA synthesizing enzyme, MAO-B, have shown efficacy. However, there are contradictory evidences on the role of AQP4 in AD.
Collapse
|
102
|
Potential Use of 18F-THK5351 PET to Identify Wallerian Degeneration of the Pyramidal Tract Caused by Cerebral Infarction. Clin Nucl Med 2018; 42:e523-e524. [PMID: 29076904 DOI: 10.1097/rlu.0000000000001868] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
A 41-year-old man underwent F-THK5351 PET 2 years after a right middle cerebral artery infarction. F-THK5351 PET imaging revealed intense radioligand uptake along the ipsilateral pyramidal tract from the corona radiata to the medulla; intense uptake changed from the right side to the left side with descending axial sections at the level of the pyramidal decussation. F-THK5351 reportedly binds to monoamine oxidase B, which is highly expressed in astrocytes, suggesting that F-THK5351 concentrates in the lesion where gliosis occurs. Hence, in this case, F-THK5351 uptake may represent Wallerian degeneration accompanied with gliosis in the ipsilateral pyramidal tract.
Collapse
|
103
|
Kim D, Beck S, Jung J. Monitoring of Monoamine Oxidases as Biomarkers of Disease and Disorder. B KOREAN CHEM SOC 2018. [DOI: 10.1002/bkcs.11397] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Dokyoung Kim
- Department of Anatomy and Neurobiology; College of Medicine, Kyung Hee University; Seoul 02447 Republic of Korea
- Center for Converging Humanities; Kyung Hee University; Seoul 02447 Republic of Korea
| | - Sungjun Beck
- Skaggs School of Pharmacy and Pharmaceutical Sciences; University of California, San Diego (UCSD); La Jolla CA 92093 USA
| | - Junyang Jung
- Department of Anatomy and Neurobiology; College of Medicine, Kyung Hee University; Seoul 02447 Republic of Korea
| |
Collapse
|
104
|
Madeo F, Eisenberg T, Pietrocola F, Kroemer G. Spermidine in health and disease. Science 2018; 359:359/6374/eaan2788. [DOI: 10.1126/science.aan2788] [Citation(s) in RCA: 438] [Impact Index Per Article: 62.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
105
|
Tripathi AC, Upadhyay S, Paliwal S, Saraf SK. Privileged scaffolds as MAO inhibitors: Retrospect and prospects. Eur J Med Chem 2018; 145:445-497. [PMID: 29335210 DOI: 10.1016/j.ejmech.2018.01.003] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Revised: 12/01/2017] [Accepted: 01/01/2018] [Indexed: 12/24/2022]
Abstract
This review aims to be a comprehensive, authoritative, critical, and readable review of general interest to the medicinal chemistry community because it focuses on the pharmacological, chemical, structural and computational aspects of diverse chemical categories as monoamine oxidase inhibitors (MAOIs). Monoamine oxidases (MAOs), namely MAO-A and MAO-B represent an enormously valuable class of neuronal enzymes embodying neurobiological origin and functions, serving as potential therapeutic target in neuronal pharmacotherapy, and hence we have coined the term "Neurozymes" which is being introduced for the first time ever. Nowadays, therapeutic attention on MAOIs engrosses two imperative categories; MAO-A inhibitors, in certain mental disorders such as depression and anxiety, and MAO-B inhibitors, in neurodegenerative disorders like Alzheimer's disease (AD) and Parkinson's disease (PD). The use of MAOIs declined due to some potential side effects, food and drug interactions, and introduction of other classes of drugs. However, curiosity in MAOIs is reviving and the recent developments of new generation of highly selective and reversible MAOIs, have renewed the therapeutic prospective of these compounds. The initial section of the review emphasizes on the detailed classification, structural and binding characteristics, therapeutic potential, current status and future challenges of the privileged pharmacophores. However, the chemical prospective of privileged scaffolds such as; aliphatic and aromatic amines, amides, hydrazines, azoles, diazoles, tetrazoles, indoles, azines, diazines, xanthenes, tricyclics, benzopyrones, and more interestingly natural products, along with their conclusive SARs have been discussed in the later segment of review. The last segment of the article encompasses some patents granted in the field of MAOIs, in a simplistic way.
Collapse
Affiliation(s)
- Avinash C Tripathi
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India
| | - Savita Upadhyay
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India
| | - Sarvesh Paliwal
- Pharmacy Department, Banasthali Vidyapith, Banasthali, Tonk 304022, Rajasthan, India
| | - Shailendra K Saraf
- Division of Pharmaceutical Chemistry, Faculty of Pharmacy, Babu Banarasi Das Northern India Institute of Technology, Lucknow 226028, UP, India.
| |
Collapse
|
106
|
Rodriguez-Vieitez E, Nordberg A. Imaging Neuroinflammation: Quantification of Astrocytosis in a Multitracer PET Approach. Methods Mol Biol 2018; 1750:231-251. [PMID: 29512077 DOI: 10.1007/978-1-4939-7704-8_16] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
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 (NFT) of hyperphosphorylated tau proteins, accompanied by the activation of glial cells-astrocytes and microglia-and neuroinflammatory responses, leading to neurodegeneration and cognitive dysfunction. An increasing diversity of positron emission tomography (PET) imaging radiotracers are 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 also a great interest to develop PET tracers to image glial activation and neuroinflammation. While most research to date has focused on imaging microgliosis, recent studies using 11C-deuterium-L-deprenyl (11C-DED) PET imaging suggest that astrocytosis may be present from very 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, and glucose metabolism in patients 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 responses in neurodegenerative diseases. Progress in the field will contribute to the incorporation of PET imaging of glial activation 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 Translational Alzheimer Neurobiology, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden.
| | - Agneta Nordberg
- Division of Translational Alzheimer Neurobiology, Center for Alzheimer Research, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| |
Collapse
|
107
|
Yurt KK, Kaplan S. As a painkiller: a review of pre- and postnatal non-steroidal anti-inflammatory drug exposure effects on the nervous systems. Inflammopharmacology 2017; 26:15-28. [DOI: 10.1007/s10787-017-0434-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2017] [Accepted: 12/10/2017] [Indexed: 11/29/2022]
|
108
|
Vermeiren C, Motte P, Viot D, Mairet-Coello G, Courade JP, Citron M, Mercier J, Hannestad J, Gillard M. The tau positron-emission tomography tracer AV-1451 binds with similar affinities to tau fibrils and monoamine oxidases. Mov Disord 2017; 33:273-281. [DOI: 10.1002/mds.27271] [Citation(s) in RCA: 96] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Revised: 11/01/2017] [Accepted: 11/22/2017] [Indexed: 12/25/2022] Open
Affiliation(s)
| | - Philippe Motte
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | - Delphine Viot
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | | | | | - Martin Citron
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | - Joël Mercier
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| | - Jonas Hannestad
- Denali Therapeutics Inc.; South San Francisco California USA
| | - Michel Gillard
- UCB BioPharma sprl, Chemin du Foriest; Braine l'Alleud Belgium
| |
Collapse
|
109
|
Lesniak A, Aarnio M, Diwakarla S, Norberg T, Nyberg F, Gordh T. Characterization of the binding site for d-deprenyl in human inflamed synovial membrane. Life Sci 2017; 194:26-33. [PMID: 29221756 DOI: 10.1016/j.lfs.2017.12.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 11/28/2017] [Accepted: 12/03/2017] [Indexed: 01/23/2023]
Abstract
AIMS d-Deprenyl when used as a positron emission tomography tracer visualizes peripheral inflammation. The major aim of the current study was to identify and investigate the properties of the binding target for d-deprenyl in synovial membrane explants from arthritic patients. MAIN METHODS Thirty patients diagnosed with arthritis or osteoarthritis were enrolled into the study. Homologous and competitive radioligand binding assays utilizing [3H]d-deprenyl were performed to investigate the biochemical characteristics of the binding site and assess differences in the binding profile in synovial membranes exhibiting varying levels of inflammation. KEY FINDINGS The [3H]d-deprenyl binding assay confirmed the existence of a single, saturable population of membrane-bound protein binding sites in synovial membrane homogenates. The macroscopically determined level of inflammation correlated with an increase in [3H]d-deprenyl binding affinity, without significant alterations in binding site density. Selective monoamine oxidase B inhibitor, selegiline competed for the same site as [3H]d-deprenyl, but failed to differentiate the samples with regard to their inflammation grade. A monoamine oxidase A inhibitor, pirlindole mesylate showed only weak displacement of [3H]d-deprenyl binding. No significant alterations in monoamine oxidase B expression was detected, thus it was not confirmed whether it could serve as a marker for ongoing inflammation. SIGNIFICANCE Our study was the first to show the biochemical characteristics of the [3H]d-deprenyl binding site in inflamed human synovium. We confirmed that d-deprenyl could differentiate between patients with varying severity of synovitis in the knee joint by binding to a protein target distinct from monoamine oxidase B.
Collapse
Affiliation(s)
- Anna Lesniak
- Uppsala University, Department of Pharmaceutical Biosciences, SE 751 24 Uppsala, Sweden; Medical University of Warsaw, Department of Pharmacodynamics, Centre for Preclinical Research and Technology, 02-097 Warsaw, Poland.
| | - Mikko Aarnio
- Uppsala University Hospital, Department of Surgical Sciences, Anaesthesiology and Intensive Care, SE 751 85 Uppsala, Sweden
| | - Shanti Diwakarla
- Uppsala University, Department of Pharmaceutical Biosciences, SE 751 24 Uppsala, Sweden
| | - Thomas Norberg
- Uppsala University, Department of Physical Organic Chemistry, SE 751 23 Uppsala, Sweden
| | - Fred Nyberg
- Uppsala University, Department of Pharmaceutical Biosciences, SE 751 24 Uppsala, Sweden
| | - Torsten Gordh
- Uppsala University Hospital, Department of Surgical Sciences, Anaesthesiology and Intensive Care, SE 751 85 Uppsala, Sweden
| |
Collapse
|
110
|
Kreisl WC, Henter ID, Innis RB. Imaging Translocator Protein as a Biomarker of Neuroinflammation in Dementia. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2017; 82:163-185. [PMID: 29413519 PMCID: PMC6190574 DOI: 10.1016/bs.apha.2017.08.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Neuroinflammation has long been considered a potential contributor to neurodegenerative disorders that result in dementia. Accumulation of abnormal protein aggregates in Alzheimer's disease, frontotemporal dementia, and dementia with Lewy bodies is associated with the activation of microglia and astrocytes into proinflammatory states, and chronic low-level activation of glial cells likely contributes to the pathological changes observed in these and other neurodegenerative diseases. The 18kDa translocator protein (TSPO) is a key biomarker for measuring inflammation in the brain via positron emission tomography (PET). Increased TSPO density has been observed in brain tissue from patients with neurodegenerative diseases and colocalizes to activated microglia and reactive astrocytes. Several radioligands have been developed to measure TSPO density in vivo with PET, and these have been used in clinical studies of different dementia syndromes. However, TSPO radioligands have limitations, including low specific-to-nonspecific signal and differential affinity to a polymorphism on the TSPO gene, which must be taken into consideration in designing and interpreting human PET studies. Nonetheless, most PET studies have shown that increased TSPO binding is associated with various dementias, suggesting that TSPO has potential as a biomarker to further explore the role of neuroinflammation in dementia pathogenesis and may prove useful in monitoring disease progression.
Collapse
Affiliation(s)
- William C Kreisl
- Taub Institute, Columbia University Medical Center, New York, NY, United States.
| | - Ioline D Henter
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| | - Robert B Innis
- Molecular Imaging Branch, National Institute of Mental Health, Bethesda, MD, United States
| |
Collapse
|
111
|
Tripathi RKP, M Sasi V, Gupta SK, Krishnamurthy S, Ayyannan SR. Design, synthesis, and pharmacological evaluation of 2-amino-5-nitrothiazole derived semicarbazones as dual inhibitors of monoamine oxidase and cholinesterase: effect of the size of aryl binding site. J Enzyme Inhib Med Chem 2017; 33:37-57. [PMID: 29098902 PMCID: PMC6009888 DOI: 10.1080/14756366.2017.1389920] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
A series of 2-amino-5-nitrothiazole derived semicarbazones were designed, synthesised and investigated for MAO and ChE inhibition properties. Most of the compounds showed preferential inhibition towards MAO-B. Compound 4, (1-(1-(4-Bromophenyl)ethylidene)-4-(5-nitrothiazol-2-yl)semicarbazide) emerged as lead candidate (IC50 = 0.212 µM, SI = 331.04) against MAO-B; whereas compounds 21 1-(5-Bromo-2-oxoindolin-3-ylidene)-4-(5-nitrothiazol-2-yl)semicarbazide (IC50 = 0.264 µM) and 17 1-((4-Chlorophenyl) (phenyl)methylene)-4-(5-nitrothiazol-2-yl)semicarbazide (IC50 = 0.024 µM) emerged as lead AChE and BuChE inhibitors respectively; with activity of compound 21 almost equivalent to tacrine. Kinetic studies indicated that compound 4 exhibited competitive and reversible MAO-B inhibition while compounds 21 and 17 showed mixed-type of AChE and BuChE inhibition respectively. Docking studies revealed that these compounds were well-accommodated within MAO-B and ChE active sites through stable hydrogen bonding and/or hydrophobic interactions. This study revealed the requirement of small heteroaryl ring at amino terminal of semicarbazone template for preferential inhibition and selectivity towards MAO-B. Our results suggest that 5-nitrothiazole derived semicarbazones could be further exploited for its multi-targeted role in development of anti-neurodegenerative agents. [Formula: see text] A library of 2-amino-5-nitrothiazole derived semicarbazones (4-21) was designed, synthesised and evaluated for in vitro MAO and ChE inhibitory activity. Compounds 4, 21 and 17 (shown) have emerged as lead MAO-B (IC50:0.212 µM, competitive and reversible), AChE (IC50:0.264 µM, mixed and reversible) and BuChE (IC50:0.024 µM, mixed and reversible) inhibitor respectively. SAR studies disclosed several structural aspects significant for potency and selectivity and indicated the role of size of aryl binding site in potency and selectivity towards MAO-B. Antioxidant activity and neurotoxicity screening results further suggested their multifunctional potential for the therapy of neurodegenerative diseases.
Collapse
Affiliation(s)
- Rati K P Tripathi
- a Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi , India
| | - Vishnu M Sasi
- a Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi , India
| | - Sukesh K Gupta
- b Neurotherapeutics Research Laboratory, Department of Pharmaceutical Engineering & Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi , India
| | - Sairam Krishnamurthy
- b Neurotherapeutics Research Laboratory, Department of Pharmaceutical Engineering & Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi , India
| | - Senthil R Ayyannan
- a Pharmaceutical Chemistry Research Laboratory, Department of Pharmaceutical Engineering & Technology , Indian Institute of Technology (Banaras Hindu University) , Varanasi , India
| |
Collapse
|
112
|
Tong J, Rathitharan G, Meyer JH, Furukawa Y, Ang LC, Boileau I, Guttman M, Hornykiewicz O, Kish SJ. Brain monoamine oxidase B and A in human parkinsonian dopamine deficiency disorders. Brain 2017; 140:2460-2474. [PMID: 29050386 DOI: 10.1093/brain/awx172] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Accepted: 05/30/2017] [Indexed: 11/13/2022] Open
Abstract
See Jellinger (doi:10.1093/awx190) for a scientific commentary on this article. The enzyme monoamine oxidases (B and A subtypes, encoded by MAOB and MAOA, respectively) are drug targets in the treatment of Parkinson's disease. Inhibitors of MAOB are used clinically in Parkinson's disease for symptomatic purposes whereas the potential disease-modifying effect of monoamine oxidase inhibitors is debated. As astroglial cells express high levels of MAOB, the enzyme has been proposed as a brain imaging marker of astrogliosis, a cellular process possibly involved in Parkinson's disease pathogenesis as elevation of MAOB in astrocytes might be harmful. Since brain monoamine oxidase status in Parkinson's disease is uncertain, our objective was to measure, by quantitative immunoblotting in autopsied brain homogenates, protein levels of both monoamine oxidases in three different degenerative parkinsonian disorders: Parkinson's disease (n = 11), multiple system atrophy (n = 11), and progressive supranuclear palsy (n = 16) and in matched controls (n = 16). We hypothesized that if MAOB is 'substantially' localized to astroglial cells, MAOB levels should be generally associated with standard astroglial protein measures (e.g. glial fibrillary acidic protein). MAOB levels were increased in degenerating putamen (+83%) and substantia nigra (+10%, non-significant) in multiple system atrophy; in caudate (+26%), putamen (+27%), frontal cortex (+31%) and substantia nigra (+23%) of progressive supranuclear palsy; and in frontal cortex (+33%), but not in substantia nigra of Parkinson's disease, a region we previously reported no increase in astrocyte protein markers. Although the magnitude of MAOB increase was less than those of standard astrocytic markers, significant positive correlations were observed amongst the astrocyte proteins and MAOB. Despite suggestions that MAOA (versus MAOB) is primarily responsible for metabolism of dopamine in dopamine neurons, there was no loss of the enzyme in the parkinsonian substantia nigra; instead, increased nigral levels of a MAOA fragment and 'turnover' of the enzyme were observed in the conditions. Our findings provide support that MAOB might serve as a biochemical imaging marker, albeit not entirely specific, for astrocyte activation in human brain. The observation that MAOB protein concentration is generally increased in degenerating brain areas in multiple system atrophy (especially putamen) and in progressive supranuclear palsy, but not in the nigra in Parkinson's disease, also distinguishes astrocyte behaviour in Parkinson's disease from that in the two 'Parkinson-plus' conditions. The question remains whether suppression of either MAOB in astrocytes or MAOA in dopamine neurons might influence progression of the parkinsonian disorders.
Collapse
Affiliation(s)
- Junchao Tong
- Preclinical Imaging Unit, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada.,Human Brain Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Gausiha Rathitharan
- Human Brain Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Jeffrey H Meyer
- Research Imaging Centre and Campbell Family Mental Health Research Institute, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Yoshiaki Furukawa
- Department of Neurology, Juntendo Tokyo Koto Geriatric Medical Center, and Faculty of Medicine, University and Post Graduate University of Juntendo, Tokyo, Japan
| | - Lee-Cyn Ang
- Division of Neuropathology, London Health Science Centre, University of Western Ontario, London, Ontario, Canada
| | - Isabelle Boileau
- Addiction Imaging Research Group, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| | - Mark Guttman
- Centre for Movement Disorders, Markham, Ontario, Canada
| | - Oleh Hornykiewicz
- Centre for Brain Research, Medical University of Vienna, Spitalgasse 4, A-1090 Vienna, Austria
| | - Stephen J Kish
- Human Brain Laboratory, Research Imaging Centre, Centre for Addiction and Mental Health, Toronto, Ontario, Canada
| |
Collapse
|
113
|
Effect of fluorinated tetrahydrocarbazole derivatives on the enzymes of oxidative deamination of biogenic amines and on the process of lipid peroxidation. Russ Chem Bull 2017. [DOI: 10.1007/s11172-017-1821-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
114
|
Harada R, Ishiki A, Kai H, Sato N, Furukawa K, Furumoto S, Tago T, Tomita N, Watanuki S, Hiraoka K, Ishikawa Y, Funaki Y, Nakamura T, Yoshikawa T, Iwata R, Tashiro M, Sasano H, Kitamoto T, Yanai K, Arai H, Kudo Y, Okamura N. Correlations of 18F-THK5351 PET with Postmortem Burden of Tau and Astrogliosis in Alzheimer Disease. J Nucl Med 2017; 59:671-674. [PMID: 28864633 DOI: 10.2967/jnumed.117.197426] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2017] [Accepted: 07/25/2017] [Indexed: 01/03/2023] Open
Abstract
Clinical PET studies using 18F-THK5351 have demonstrated significant tracer retention in sites susceptible to tau burden in Alzheimer disease (AD). However, the in vivo PET signal to reflect tau aggregates remains controversial. Methods: We examined the spatial pattern of tracer binding, amyloid-β, tau, and gliosis in an autopsy-confirmed AD patient who underwent 18F-THK5351 and 11C-Pittsburgh compound B PET before death. Results: Regional in vivo 18F-THK5351 retention was significantly correlated with the density of tau aggregates in the neocortex and monoamine oxidase-B in the whole brain, but not correlated with that of insoluble amyloid-β. Furthermore, significant association was observed between the density of tau aggregates, monoamine oxidase-B, and glial fibrillary acidic protein, suggesting that neocortical tau would strongly influence the formation of reactive astrocytes. Conclusion:18F-THK5351 PET may have limited utility as a biomarker of tau pathology in AD; however, it could be used to monitor the neuroinflammatory processes in the living brain.
Collapse
Affiliation(s)
- Ryuichi Harada
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan .,Department of Gerontology and Geriatrics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Aiko Ishiki
- Department of Gerontology and Geriatrics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Hideaki Kai
- Department of Neurological Science, Tohoku University School of Medicine, Sendai, Japan
| | - Naomi Sato
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | - Katsutoshi Furukawa
- Department of Gerontology and Geriatrics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan.,Division of Community Medicine, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Shozo Furumoto
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Tetsuro Tago
- Research Team for Neuroimaging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan; and
| | - Naoki Tomita
- Department of Gerontology and Geriatrics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Shoichi Watanuki
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Kotaro Hiraoka
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yoichi Ishikawa
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Yoshihito Funaki
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Tadaho Nakamura
- Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Takeo Yoshikawa
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan
| | - Ren Iwata
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Manabu Tashiro
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Hironobu Sasano
- Department of Pathology, Tohoku University School of Medicine, Sendai, Japan
| | - Tetsuyuki Kitamoto
- Department of Neurological Science, Tohoku University School of Medicine, Sendai, Japan
| | - Kazuhiko Yanai
- Department of Pharmacology, Tohoku University School of Medicine, Sendai, Japan.,Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan
| | - Hiroyuki Arai
- Department of Gerontology and Geriatrics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Yukitsuka Kudo
- Department of Gerontology and Geriatrics, Institute of Development, Aging and Cancer, Tohoku University, Sendai, Japan
| | - Nobuyuki Okamura
- Cyclotron and Radioisotope Center, Tohoku University, Sendai, Japan.,Division of Pharmacology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
115
|
Borroni E, Bohrmann B, Grueninger F, Prinssen E, Nave S, Loetscher H, Chinta SJ, Rajagopalan S, Rane A, Siddiqui A, Ellenbroek B, Messer J, Pähler A, Andersen JK, Wyler R, Cesura AM. Sembragiline: A Novel, Selective Monoamine Oxidase Type B Inhibitor for the Treatment of Alzheimer's Disease. J Pharmacol Exp Ther 2017; 362:413-423. [PMID: 28642233 DOI: 10.1124/jpet.117.241653] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2017] [Accepted: 05/23/2017] [Indexed: 12/26/2022] Open
Abstract
Monoamine oxidase B (MAO-B) has been implicated in the pathogenesis of Alzheimer's disease (AD) and other neurodegenerative disorders. Increased MAO-B expression in astroglia has been observed adjacent to amyloid plaques in AD patient brains. This phenomenon is hypothesized to lead to increased production of hydrogen peroxide and reactive oxygen species (ROS), thereby contributing to AD pathology. Therefore, reduction of ROS-induced oxidative stress via inhibition of MAO-B activity may delay the progression of the disease. In the present study we report the pharmacological properties of sembragiline, a novel selective MAO-B inhibitor specifically developed for the treatment of AD, and on its effect on ROS-mediated neuronal injury and astrogliosis in MAO-B transgenic animals. Sembragiline showed potent and long-lasting MAO-B-selective inhibition and did not inhibit MAO-A at doses where full inhibition of MAO-B was observed. Such selectivity should translate into a favorable clinical safety profile. Indeed, sembragiline neither induced the serotonin syndrome when administered together with the serotonin precursor l-5-hydroxytryptophan in combination with antidepressants such as fluoxetine, nor potentiated the pressor effect of tyramine. Additionally, in experiments using a transgenic animal model conditionally overexpressing MAO-B in astroglia, sembragiline protected against neuronal loss and reduced both ROS formation and reactive astrogliosis. Taken together, these findings warrant further investigation of the potential therapeutic benefit of MAO-B inhibitors in patients with AD and other neurologic disorders.
Collapse
Affiliation(s)
- Edilio Borroni
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Bernd Bohrmann
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Fiona Grueninger
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Eric Prinssen
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Stephane Nave
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Hansruedi Loetscher
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Shankar J Chinta
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Subramanian Rajagopalan
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Anand Rane
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Almas Siddiqui
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Bart Ellenbroek
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Juerg Messer
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Axel Pähler
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Julie K Andersen
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Rene Wyler
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| | - Andrea M Cesura
- Roche Innovation Center Basel, Pharma Research and Early Development, F. Hoffmann-La Roche Ltd., Basel, Switzerland (E.B., B.B., F.G., E.P., S.N., H.L., J.M., A.P., and R.W.); Buck Institute for Research on Aging, Novato, California (S.C., S.R., A.R., A.S., and J.A.); and Evotec International GmbH, Hamburg, Germany (A.M.C. and B.E.)
| |
Collapse
|
116
|
Deng YH, Wang NN, Zou ZX, Zhang L, Xu KP, Chen AF, Cao DS, Tan GS. Multi-Target Screening and Experimental Validation of Natural Products from Selaginella Plants against Alzheimer's Disease. Front Pharmacol 2017; 8:539. [PMID: 28890698 PMCID: PMC5574911 DOI: 10.3389/fphar.2017.00539] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2017] [Accepted: 08/03/2017] [Indexed: 11/30/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disorder which is considered to be the most common cause of dementia. It has a greater impact not only on the learning and memory disturbances but also on social and economy. Currently, there are mainly single-target drugs for AD treatment but the complexity and multiple etiologies of AD make them difficult to obtain desirable therapeutic effects. Therefore, the choice of multi-target drugs will be a potential effective strategy inAD treatment. To find multi-target active ingredients for AD treatment from Selaginella plants, we firstly explored the behaviors effects on AD mice of total extracts (TE) from Selaginella doederleinii on by Morris water maze test and found that TE has a remarkable improvement on learning and memory function for AD mice. And then, multi-target SAR models associated with AD-related proteins were built based on Random Forest (RF) and different descriptors to preliminarily screen potential active ingredients from Selaginella. Considering the prediction outputs and the quantity of existing compounds in our laboratory, 13 compounds were chosen to carry out the in vitro enzyme inhibitory experiments and 4 compounds with BACE1/MAO-B dual inhibitory activity were determined. Finally, the molecular docking was applied to verify the prediction results and enzyme inhibitory experiments. Based on these study and validation processes, we explored a new strategy to improve the efficiency of active ingredients screening based on trace amount of natural product and numbers of targets and found some multi-target compounds with biological activity for the development of novel drugs for AD treatment.
Collapse
Affiliation(s)
- Yin-Hua Deng
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China
| | - Ning-Ning Wang
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China
| | - Zhen-Xing Zou
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China.,Pharmacy Department, Xiangya Hospital, Central South UniversityChangsha, China
| | - Lin Zhang
- College of Food Science and Technology, Central South University of Forestry and TechnologyChangsha, China
| | - Kang-Ping Xu
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China
| | - Alex F Chen
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China.,Center for Vascular Disease and Translational Medicine, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Dong-Sheng Cao
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China.,Center for Vascular Disease and Translational Medicine, Third Xiangya Hospital, Central South UniversityChangsha, China
| | - Gui-Shan Tan
- Xiangya School of Pharmaceutical Sciences, Central South UniversityChangsha, China.,Pharmacy Department, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
117
|
Schedin-Weiss S, Inoue M, Hromadkova L, Teranishi Y, Yamamoto NG, Wiehager B, Bogdanovic N, Winblad B, Sandebring-Matton A, Frykman S, Tjernberg LO. Monoamine oxidase B is elevated in Alzheimer disease neurons, is associated with γ-secretase and regulates neuronal amyloid β-peptide levels. ALZHEIMERS RESEARCH & THERAPY 2017; 9:57. [PMID: 28764767 PMCID: PMC5540560 DOI: 10.1186/s13195-017-0279-1] [Citation(s) in RCA: 157] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Accepted: 06/21/2017] [Indexed: 01/03/2023]
Abstract
Background Increased levels of the pathogenic amyloid β-peptide (Aβ), released from its precursor by the transmembrane protease γ-secretase, are found in Alzheimer disease (AD) brains. Interestingly, monoamine oxidase B (MAO-B) activity is also increased in AD brain, but its role in AD pathogenesis is not known. Recent neuroimaging studies have shown that the increased MAO-B expression in AD brain starts several years before the onset of the disease. Here, we show a potential connection between MAO-B, γ-secretase and Aβ in neurons. Methods MAO-B immunohistochemistry was performed on postmortem human brain. Affinity purification of γ-secretase followed by mass spectrometry was used for unbiased identification of γ-secretase-associated proteins. The association of MAO-B with γ-secretase was studied by coimmunoprecipitation from brain homogenate, and by in-situ proximity ligation assay (PLA) in neurons as well as mouse and human brain sections. The effect of MAO-B on Aβ production and Notch processing in cell cultures was analyzed by siRNA silencing or overexpression experiments followed by ELISA, western blot or FRET analysis. Methodology for measuring relative intraneuronal MAO-B and Aβ42 levels in single cells was developed by combining immunocytochemistry and confocal microscopy with quantitative image analysis. Results Immunohistochemistry revealed MAO-B staining in neurons in the frontal cortex, hippocampus CA1 and entorhinal cortex in postmortem human brain. Interestingly, the neuronal staining intensity was higher in AD brain than in control brain in these regions. Mass spectrometric data from affinity purified γ-secretase suggested that MAO-B is a γ-secretase-associated protein, which was confirmed by immunoprecipitation and PLA, and a neuronal location of the interaction was shown. Strikingly, intraneuronal Aβ42 levels correlated with MAO-B levels, and siRNA silencing of MAO-B resulted in significantly reduced levels of intraneuronal Aβ42. Furthermore, overexpression of MAO-B enhanced Aβ production. Conclusions This study shows that MAO-B levels are increased not only in astrocytes but also in pyramidal neurons in AD brain. The study also suggests that MAO-B regulates Aβ production in neurons via γ-secretase and thereby provides a key to understanding the relationship between MAO-B and AD pathogenesis. Potentially, the γ-secretase/MAO-B association may be a target for reducing Aβ levels using protein–protein interaction breakers. Electronic supplementary material The online version of this article (doi:10.1186/s13195-017-0279-1) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sophia Schedin-Weiss
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden.
| | - Mitsuhiro Inoue
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden.,Present address: Dainippon Sumitomo Pharma Co., Ltd, Drug Development Research Laboratories, Osaka, Japan
| | - Lenka Hromadkova
- National Institute of Mental Health, Klecany, Czech Republic.,Faculty of Science, Charles University in Prague, Prague, Czech Republic
| | - Yasuhiro Teranishi
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden.,Present address: Dainippon Sumitomo Pharma Co., Ltd, Drug Development Research Laboratories, Osaka, Japan
| | - Natsuko Goto Yamamoto
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden.,Present address: Dainippon Sumitomo Pharma Co., Ltd, Drug Development Research Laboratories, Osaka, Japan
| | - Birgitta Wiehager
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Nenad Bogdanovic
- Department of Geriatric Medicine, University in Oslo, Memory Clinic, Oslo University Hospital, Oslo, Norway
| | - Bengt Winblad
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Anna Sandebring-Matton
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Susanne Frykman
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| | - Lars O Tjernberg
- Karolinska Institutet, Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Huddinge, Sweden
| |
Collapse
|
118
|
Arakawa R, Stenkrona P, Takano A, Nag S, Maior RS, Halldin C. Test-retest reproducibility of [ 11C]-L-deprenyl-D 2 binding to MAO-B in the human brain. EJNMMI Res 2017. [PMID: 28634836 PMCID: PMC5478550 DOI: 10.1186/s13550-017-0301-4] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Background [11C]-l-deprenyl-D2 is a positron emission tomography (PET) radioligand for measurement of the monoamine oxidase B (MAO-B) activity in vivo brain. The estimation of the test-retest reproducibility is important for accurate interpretation of PET studies. Results We performed two [11C]-l-deprenyl-D2 scans for six healthy subjects and evaluated the test-retest variability of this radioligand. MAO-B binding was quantified by two tissue compartment model (2TCM) with three rate constants (K1, k2, k3) using metabolite-corrected plasma radioactivity. The λk3 defined as (K1/k2) × k3 was also calculated. The correlation between MAO-B binding and age, and the effect of partial volume effect correction (PVEc) for the reproducibility were also estimated. %difference of k3 was 2.6% (medial frontal cortex) to 10.3% (hippocampus), and that of λk3 was 5.0% (thalamus) to 9.2% (cerebellum). Mean %difference of all regions were 5.3 and 7.0% in k3 and λk3, respectively. All regions showed below 10% variabilities except the hippocampus in k3 (10.3%). Intraclass correlation coefficient (ICC) of k3 was 0.78 (hippocampus) to 0.98 (medial frontal cortex), and that of λk3 was 0.78 (hippocampus) to 0.95 (thalamus). Mean ICC were 0.94 and 0.89 in k3 and λk3, respectively. The highest positive correlation with age was observed in the hippocampus, as r = 0.75 in k3 and 0.76 in λk3. After PVEc, mean %difference were 5.6 and 7.2% in k3 and λk3, respectively. Mean ICC were 0.92 and 0.90 for k3 and λk3, respectively. These values were almost the same as those before PVEc. Conclusions The present results indicate that k3 and λk3 of [11C]-l-deprenyl-D2 are reliable parameters for test-retest reproducibility with healthy subjects both before and after PVEc. The studies with patients of larger sample size are required for further clinical applications.
Collapse
Affiliation(s)
- Ryosuke Arakawa
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.
| | - Per Stenkrona
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Akihiro Takano
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Sangram Nag
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| | - Rafael S Maior
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden.,Primate Center and Laboratory of Neurosciences and Behavior, Department of Physiological Sciences, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Christer Halldin
- Department of Clinical Neuroscience, Center for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm, Sweden
| |
Collapse
|
119
|
Saadeh HA, Khasawneh MA, Samadi A, El-Haty IA, Satała G, Bojarski AJ, Ismaili L, Bautista-Aguilera ÓM, Yañez M, Mestres J, Marco-Contelles J. Design, Synthesis and Biological Evaluation of Potent Antioxidant 1-(2,5-Dimethoxybenzyl)-4-arylpiperazines and N
-Azolyl Substituted 2-(4-Arylpiperazin-1-yl). ChemistrySelect 2017. [DOI: 10.1002/slct.201700397] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Haythem A. Saadeh
- Department of Chemistry; College of Science; United Arab Emirates University; Al Ain 15551 UAE
- Department of Chemistry; Faculty of Science; The University of Jordan; Amman 11942 Jordan
| | - Mohammad A. Khasawneh
- Department of Chemistry; College of Science; United Arab Emirates University; Al Ain 15551 UAE
| | - Abdelouahid Samadi
- Department of Chemistry; College of Science; United Arab Emirates University; Al Ain 15551 UAE
| | - Ismail A. El-Haty
- Department of Chemistry; College of Science; United Arab Emirates University; Al Ain 15551 UAE
| | - Grzegorz Satała
- Institute of Pharmacology; Polish Academy of Sciences; 12 Smętna Street 31-343 Kraków Poland
| | - Andrzej J. Bojarski
- Institute of Pharmacology; Polish Academy of Sciences; 12 Smętna Street 31-343 Kraków Poland
| | - Lhassane Ismaili
- Neurosciences Intégratives et Cliniques, EA 481; Univ. Bourgogne Franche-Comté; Laboratoire de Chimie Organique et Thérapeutique, UFR SMP; 19, rue Ambroise Paré F-25000 Besançon France
| | - Óscar M. Bautista-Aguilera
- Neurosciences Intégratives et Cliniques, EA 481; Univ. Bourgogne Franche-Comté; Laboratoire de Chimie Organique et Thérapeutique, UFR SMP; 19, rue Ambroise Paré F-25000 Besançon France
| | - Matilde Yañez
- Facultad de Farmacia; Departamento de Farmacología; Universidad de Santiago de Compostela; Campus Vida, Santiago de Compostela La Coruña Spain
| | - Jordi Mestres
- Research Group on Systems Pharmacology; Research Program on Biomedical Informatics (GRIB); IMIM Hospital del Mar Institute of Medical Research; Universitat Pompeu Fabra; Doctor Aiguader 88 08003 Barcelona Spain
| | - José Marco-Contelles
- Laboratory of Medicinal Chemistry (IQOG, CSIC); C/ Juan de la Cierva 3 28006- Madrid Spain
| |
Collapse
|
120
|
Issaoui N, Ghalla H, Brandán SA, Bardak F, Flakus H, Atac A, Oujia B. Experimental FTIR and FT-Raman and theoretical studies on the molecular structures of monomer and dimer of 3-thiopheneacrylic acid. J Mol Struct 2017; 1135:209-221. [DOI: 10.1016/j.molstruc.2017.01.074] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
121
|
Akomolafe SF, Akinyemi AJ, Ogunsuyi OB, Oyeleye SI, Oboh G, Adeoyo OO, Allismith YR. Effect of caffeine, caffeic acid and their various combinations on enzymes of cholinergic, monoaminergic and purinergic systems critical to neurodegeneration in rat brain-In vitro. Neurotoxicology 2017; 62:6-13. [PMID: 28465162 DOI: 10.1016/j.neuro.2017.04.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 03/08/2017] [Accepted: 04/24/2017] [Indexed: 12/29/2022]
Abstract
Caffeine and caffeic acid are two bioactive compounds that are present in plant foods and are major constituent of coffee, cocoa, tea, cola drinks and chocolate. Although not structurally related, caffeine and caffeic acid has been reported to elicit neuroprotective properties. However, their different proportional distribution in food sources and possible effect of such interactions are not often taken into consideration. Therefore, in this study, we investigated the effect of caffeine, caffeic acid and their various combinations on activities of some enzymes [acetylcholinesterase (AChE), monoamine oxidase (MAO) ecto-nucleoside triphosphate diphosphohydrolase (E-NTPase), ecto-51-nucleotidase (E-NTDase) and Na+/K+ ATPase relevant to neurodegeneration in vitro in rat brain. The stock concentration of caffeine and caffiec acid and their various proportional combinations were prepared and their interactions with the activities of these enzymes were assessed (in vitro) in different brain structures. The Fe2+ and Cu2+ chelating abilities of the samples were also investigated. The results revealed that caffeine, caffeic acid and their various combinations exhibited inhibitory effect on activities of AChE, MAO, E-NTPase and E-NTDase, but stimulatory effect on Na+/K+ ATPase activity. The combinations also exhibited Fe2+ and Cu2+ chelating abilities. Considering the various combinations, a higher caffeine to caffeic acid ratio produced significantly highest enzyme modulatory effects; these were significantly lower to the effect of caffeine alone but significantly higher than the effect of caffeic acid alone. These findings may provide new insight into the effect of proportional combination of these bioactive compounds as obtained in many foods especially with respect to their neuroprotective effects.
Collapse
Affiliation(s)
- S F Akomolafe
- Department of Biochemistry, Ekiti State University, Ado-Ekiti, Private Mail Bag 5363, Nigeria.
| | - A J Akinyemi
- Department of Biochemistry, Afe Babalola University, Ado-Ekiti, Private Mail Bag 5454, Nigeria
| | - O B Ogunsuyi
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology Akure, Private Mail Bag 704, Akure 340001, Nigeria; Department of Biomedical Technology, School of Health and Health Technology, Federal University of Technology Akure, Private Mail Bag 704, Akure 340001, Nigeria
| | - S I Oyeleye
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology Akure, Private Mail Bag 704, Akure 340001, Nigeria; Department of Biomedical Technology, School of Health and Health Technology, Federal University of Technology Akure, Private Mail Bag 704, Akure 340001, Nigeria
| | - G Oboh
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology Akure, Private Mail Bag 704, Akure 340001, Nigeria
| | - O O Adeoyo
- Functional Foods and Nutraceuticals Unit, Department of Biochemistry, Federal University of Technology Akure, Private Mail Bag 704, Akure 340001, Nigeria
| | - Y R Allismith
- Department of Biochemistry, Ekiti State University, Ado-Ekiti, Private Mail Bag 5363, Nigeria
| |
Collapse
|
122
|
Schain M, Kreisl WC. Neuroinflammation in Neurodegenerative Disorders—a Review. Curr Neurol Neurosci Rep 2017; 17:25. [DOI: 10.1007/s11910-017-0733-2] [Citation(s) in RCA: 187] [Impact Index Per Article: 23.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
123
|
Sturm S, Forsberg A, Nave S, Stenkrona P, Seneca N, Varrone A, Comley RA, Fazio P, Jamois C, Nakao R, Ejduk Z, Al-Tawil N, Akenine U, Halldin C, Andreasen N, Ricci B. Positron emission tomography measurement of brain MAO-B inhibition in patients with Alzheimer's disease and elderly controls after oral administration of sembragiline. Eur J Nucl Med Mol Imaging 2017; 44:382-391. [PMID: 27633250 PMCID: PMC5281649 DOI: 10.1007/s00259-016-3510-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 08/29/2016] [Indexed: 11/24/2022]
Abstract
PURPOSE In Alzheimer's disease (AD), increased metabolism of monoamines by monoamine oxidase type B (MAO-B) leads to the production of toxic reactive oxygen species (ROS), which are thought to contribute to disease pathogenesis. Inhibition of the MAO-B enzyme may restore brain levels of monoaminergic neurotransmitters, reduce the formation of toxic ROS and reduce neuroinflammation (reactive astrocytosis), potentially leading to neuroprotection. Sembragiline (also referred as RO4602522, RG1577 and EVT 302 in previous communications) is a potent, selective and reversible inhibitor of MAO-B developed as a potential treatment for AD. METHODS This study assessed the relationship between plasma concentration of sembragiline and brain MAO-B inhibition in patients with AD and in healthy elderly control (EC) subjects. Positron emission tomography (PET) scans using [11C]-L-deprenyl-D2 radiotracer were performed in ten patients with AD and six EC subjects, who received sembragiline each day for 6-15 days. RESULTS At steady state, the relationship between sembragiline plasma concentration and MAO-B inhibition resulted in an Emax of ∼80-90 % across brain regions of interest and in an EC50 of 1-2 ng/mL. Data in patients with AD and EC subjects showed that near-maximal inhibition of brain MAO-B was achieved with 1 mg sembragiline daily, regardless of the population, whereas lower doses resulted in lower and variable brain MAO-B inhibition. CONCLUSIONS This PET study confirmed that daily treatment of at least 1 mg sembragiline resulted in near-maximal inhibition of brain MAO-B enzyme in patients with AD.
Collapse
Affiliation(s)
- Stefan Sturm
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Grenzacherstrasse 124, Basel, Switzerland.
- F. Hoffmann-La Roche Ltd, Grenzacherstrasse 124, 4070, Basel, Switzerland.
| | - Anton Forsberg
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Stephane Nave
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Grenzacherstrasse 124, Basel, Switzerland
| | - Per Stenkrona
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Andrea Varrone
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | | | - Patrik Fazio
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Candice Jamois
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Grenzacherstrasse 124, Basel, Switzerland
| | - Ryuji Nakao
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Zbigniew Ejduk
- Internal Disease and Gastroenterology, Miedzyleski Specialistic Hospital, Warsaw, Poland
| | - Nabil Al-Tawil
- Karolinska Trial Alliance Phase 1 Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrika Akenine
- Karolinska Institutet Alzheimer Disease Research Centre and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden
| | - Christer Halldin
- Department of Clinical Neuroscience, Centre for Psychiatric Research, Karolinska Institutet, Stockholm, Sweden
| | - Niels Andreasen
- Karolinska Institutet Alzheimer Disease Research Centre and Clinical Trial Unit, Geriatric Clinic, Karolinska University Hospital, Huddinge, Sweden
| | - Benedicte Ricci
- Roche Innovation Center Basel, Roche Pharmaceutical Research and Early Development, Grenzacherstrasse 124, Basel, Switzerland
| |
Collapse
|
124
|
Wu WY, Dai YC, Li NG, Dong ZX, Gu T, Shi ZH, Xue X, Tang YP, Duan JA. Novel multitarget-directed tacrine derivatives as potential candidates for the treatment of Alzheimer's disease. J Enzyme Inhib Med Chem 2017; 32:572-587. [PMID: 28133981 PMCID: PMC6009885 DOI: 10.1080/14756366.2016.1210139] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder, which is complex and progressive; it has not only threatened the health of elderly people, but also burdened the whole social medical and health system. The available therapy for AD is limited and the efficacy remains unsatisfactory. In view of the prevalence and expected increase in the incidence of AD, the design and development of efficacious and safe anti-AD agents has become a hotspot in the field of pharmaceutical research. Due to the multifactorial etiology of AD, the multitarget-directed ligands (MTDLs) approach is promising in search for new drugs for AD. Tacrine, which is the first acetylcholinesterase (AChE) inhibitor, has been selected as the ideal active fragment because of its simple structure, clear activity, and its superiority in the structural modification, thus it could be introduced into the overall molecular skeletons of the multi-target-directed anti-AD agents. In this review, we have summarized the recent advances (2012 to the present) in the chemical modification of tacrine, which could provide the reference for the further study of novel multi-target-directed tacrine derivatives to treat AD.
Collapse
Affiliation(s)
- Wen-Yu Wu
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Yu-Chen Dai
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Nian-Guang Li
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Ze-Xi Dong
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Ting Gu
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Zhi-Hao Shi
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,c Department of Organic Chemistry , China Pharmaceutical University , Nanjing , Jiangsu , China
| | - Xin Xue
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Yu-Ping Tang
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| | - Jin-Ao Duan
- a Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Key Laboratory for High Technology Research of TCM Formulae, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China.,b Department of Medicinal Chemistry , Nanjing University of Chinese Medicine , Nanjing , Jiangsu , China
| |
Collapse
|
125
|
Gu F, Chauhan V, Chauhan A. Monoamine oxidase-A and B activities in the cerebellum and frontal cortex of children and young adults with autism. J Neurosci Res 2017; 95:1965-1972. [DOI: 10.1002/jnr.24027] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Revised: 12/29/2016] [Accepted: 12/30/2016] [Indexed: 11/07/2022]
Affiliation(s)
- Feng Gu
- NYS Institute for Basic Research in Developmental Disabilities; Staten Island New York
| | - Ved Chauhan
- NYS Institute for Basic Research in Developmental Disabilities; Staten Island New York
| | - Abha Chauhan
- NYS Institute for Basic Research in Developmental Disabilities; Staten Island New York
| |
Collapse
|
126
|
Issaoui N, Ghalla H, Bardak F, Karabacak M, Aouled Dlala N, Flakus H, Oujia B. Combined experimental and theoretical studies on the molecular structures, spectroscopy, and inhibitor activity of 3-(2-thienyl)acrylic acid through AIM, NBO,FT-IR, FT-Raman, UV and HOMO-LUMO analyses, and molecular docking. J Mol Struct 2017; 1130:659-668. [DOI: 10.1016/j.molstruc.2016.11.019] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
127
|
Entzeroth M, Ratty AK. Monoamine Oxidase Inhibitors—Revisiting a Therapeutic Principle. ACTA ACUST UNITED AC 2017. [DOI: 10.4236/ojd.2017.62004] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
128
|
Kim D, Baik SH, Kang S, Cho SW, Bae J, Cha MY, Sailor MJ, Mook-Jung I, Ahn KH. Close Correlation of Monoamine Oxidase Activity with Progress of Alzheimer's Disease in Mice, Observed by in Vivo Two-Photon Imaging. ACS CENTRAL SCIENCE 2016; 2:967-975. [PMID: 28058286 PMCID: PMC5200925 DOI: 10.1021/acscentsci.6b00309] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Indexed: 05/26/2023]
Abstract
Monoamine oxidases (MAOs) play an important role in Alzheimer's disease (AD) pathology. We report in vivo comonitoring of MAO activity and amyloid-β (Aβ) plaques dependent on the aging of live mice with AD, using a two-photon fluorescence probe. The probe under the catalytic action of MAO produces a dipolar fluorophore that senses Aβ plaques, a general AD biomarker, enabling us to comonitor the enzyme activity and the progress of AD indicated by Aβ plaques. The results show that the progress of AD has a close correlation with MAO activity, which can be categorized into three stages: slow initiation stage up to three months, an aggressive stage, and a saturation stage from nine months. Histological analysis also reveals elevation of MAO activity around Aβ plaques in aged mice. The close correlation between the MAO activity and AD progress observed by in vivo monitoring for the first time prompts us to investigate the enzyme as a potential biomarker of AD.
Collapse
Affiliation(s)
- Dokyoung Kim
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Sung Hoon Baik
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Seokjo Kang
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Seo Won Cho
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
| | - Juryang Bae
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
| | - Moon-Yong Cha
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Michael J. Sailor
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| | - Inhee Mook-Jung
- Department
of Biochemistry and Biomedical Sciences, College of Medicine, Seoul National University, 103 Daehak-Ro, Jongro-Gu, Seoul 110-799, Republic
of Korea
| | - Kyo Han Ahn
- Department
of Chemistry, Pohang University of Science
and Technology (POSTECH), 77 Cheongam-Ro, Nam-Gu, Pohang, Gyungbuk 37673, Republic
of Korea
| |
Collapse
|
129
|
Tripathi RKP, Ayyannan SR. Design, Synthesis, and Evaluation of 2-Amino-6-nitrobenzothiazole-Derived Hydrazones as MAO Inhibitors: Role of the Methylene Spacer Group. ChemMedChem 2016; 11:1551-67. [DOI: 10.1002/cmdc.201600202] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Indexed: 11/05/2022]
Affiliation(s)
- Rati K. P. Tripathi
- Pharmaceutical Chemistry Research Laboratory; Department of Pharmaceutics; Indian Institute of Technology; Banaras Hindu University; Varanasi 221005 Uttar Pradesh India), Fax
| | - Senthil R. Ayyannan
- Pharmaceutical Chemistry Research Laboratory; Department of Pharmaceutics; Indian Institute of Technology; Banaras Hindu University; Varanasi 221005 Uttar Pradesh India), Fax
| |
Collapse
|
130
|
Zarmouh NO, Messeha SS, Elshami FM, Soliman KFA. Natural Products Screening for the Identification of Selective Monoamine Oxidase-B Inhibitors. ACTA ACUST UNITED AC 2016; 15. [PMID: 27341283 PMCID: PMC4898948 DOI: 10.9734/ejmp/2016/26453] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aims Monoamine oxidase-B inhibitors (MAO-BIs) are used for the initial therapy of Parkinson’s disease. Also, MAO-BIs have shown to be effective neuroprotective agents in several neurodegenerative diseases. However, some concerns exist regarding the long-term use of these compounds. Meanwhile, natural compounds showed potential MAO-B selective inhibitions. To date, few selective natural MAO-BIs have been identified. Therefore, the current study is designed to identify plants with potent and specific MAO-B inhibition. Study Design In this work, we utilized high throughput screening to evaluate the different plants ethanolic extract for their effectiveness to inhibit recombinant human (h)MAO-A and hMAO-B and to determine the relative selectivity of the top MAO-BI. Methodology Recombinant human isozymes were verified by Western blotting, and the 155 plants were screened. A continuous fluorometric screening assay was performed followed by two separate hMAO-A and hMAO-B microtiter screenings and IC50 determinations for the top extracts. Results In the screened plants, 9% of the extracts showed more than 1.5-fold relative inhibition of hMAO-B (RIB) and another 9% showed more than 1.5-fold relative inhibition of hMAO-A. The top extracts with the most potent RIBs were Psoralea corylifolia seeds, Phellodendron amurense bark, Glycyrrhiza uralensis roots, and Ferula assafoetida roots, with the highest RIB of 5.9-fold. Furthermore, extensive maceration of the promising extracts led to increase inhibitory effects with a preserved RIB as confirmed with luminescence assay. The top four extracts hMAO-BIs were equally potent (IC50= 1.3 to 3.8 μg/mL) with highly significant relative selectivities to inhibit hMAO-B (4.1- to 13.4-fold). Conclusion The obtained results indicate that Psoralea corylifolia seeds, Ferula assafoetida, Glycyrrhiza uralensis roots, and Phellodendron amurense ethanolic extracts have selective inhibitions for human MAO-B. Investigating these plant extracts as natural resources for novel selective MAO-BIs may lead to the development of molecules that can be used in the therapeutic management of neurodegenerative diseases including Parkinson’s disease.
Collapse
Affiliation(s)
- Najla O Zarmouh
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Samia S Messeha
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Faisel M Elshami
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| | - Karam F A Soliman
- College of Pharmacy and Pharmaceutical Sciences, Florida A&M University, Tallahassee, Florida 32307, USA
| |
Collapse
|
131
|
Declercq LD, Vandenberghe R, Van Laere K, Verbruggen A, Bormans G. Drug Development in Alzheimer's Disease: The Contribution of PET and SPECT. Front Pharmacol 2016; 7:88. [PMID: 27065872 PMCID: PMC4814730 DOI: 10.3389/fphar.2016.00088] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/16/2016] [Indexed: 12/13/2022] Open
Abstract
Clinical trials aiming to develop disease-altering drugs for Alzheimer’s disease (AD), a neurodegenerative disorder with devastating consequences, are failing at an alarming rate. Poorly defined inclusion-and outcome criteria, due to a limited amount of objective biomarkers, is one of the major concerns. Non-invasive molecular imaging techniques, positron emission tomography and single photon emission (computed) tomography (PET and SPE(C)T), allow visualization and quantification of a wide variety of (patho)physiological processes and allow early (differential) diagnosis in many disorders. PET and SPECT have the ability to provide biomarkers that permit spatial assessment of pathophysiological molecular changes and therefore objectively evaluate and follow up therapeutic response, especially in the brain. A number of specific PET/SPECT biomarkers used in support of emerging clinical therapies in AD are discussed in this review.
Collapse
Affiliation(s)
- Lieven D Declercq
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| | - Rik Vandenberghe
- Laboratory for Cognitive Neurology, Department of Neurosciences, KU Leuven Leuven, Belgium
| | - Koen Van Laere
- Nuclear Medicine and Molecular Imaging, Department of Imaging and Pathology, KU Leuven Leuven, Belgium
| | - Alfons Verbruggen
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmacy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven Leuven, Belgium
| |
Collapse
|
132
|
Influence of water-soluble derivatives of [60]fullerene on catalytic activity of monoaminе oxidase B and their membranotropic properties. Russ Chem Bull 2016. [DOI: 10.1007/s11172-016-1374-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
133
|
Rodriguez-Vieitez E, Saint-Aubert L, Carter SF, Almkvist O, Farid K, Schöll M, Chiotis K, Thordardottir S, Graff C, Wall A, Långström B, Nordberg A. Diverging longitudinal changes in astrocytosis and amyloid PET in autosomal dominant Alzheimer's disease. Brain 2016; 139:922-36. [PMID: 26813969 PMCID: PMC4766380 DOI: 10.1093/brain/awv404] [Citation(s) in RCA: 209] [Impact Index Per Article: 23.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/20/2015] [Indexed: 11/14/2022] Open
Abstract
See Schott and Fox (doi:
10.1093/brain/awv405
) for a scientific commentary on this article.
Alzheimer’s disease is a multifactorial dementia disorder characterized by early amyloid-β, tau deposition, glial activation and neurodegeneration, where the interrelationships between the different pathophysiological events are not yet well characterized. In this study, longitudinal multitracer positron emission tomography imaging of individuals with autosomal dominant or sporadic Alzheimer’s disease was used to quantify the changes in regional distribution of brain astrocytosis (tracer
11
C-deuterium-L-deprenyl), fibrillar amyloid-β plaque deposition (
11
C-Pittsburgh compound B), and glucose metabolism (
18
F-fluorodeoxyglucose) from early presymptomatic stages over an extended period to clinical symptoms. The 52 baseline participants comprised autosomal dominant Alzheimer’s disease mutation carriers (
n =
11; 49.6 ± 10.3 years old) and non-carriers (
n =
16; 51.1 ± 14.2 years old; 10 male), and patients with sporadic mild cognitive impairment (
n =
17; 61.9 ± 6.4 years old; nine male) and sporadic Alzheimer’s disease (
n =
8; 63.0 ± 6.5 years old; five male); for confidentiality reasons, the gender of mutation carriers is not revealed. The autosomal dominant Alzheimer’s disease participants belonged to families with known mutations in either presenilin 1 (
PSEN1
) or amyloid precursor protein (
APPswe
or
APParc
) genes. Sporadic mild cognitive impairment patients were further divided into
11
C-Pittsburgh compound B-positive (
n =
13; 62.0 ± 6.4; seven male) and
11
C-Pittsburgh compound B-negative (
n =
4; 61.8 ± 7.5 years old; two male) groups using a neocortical standardized uptake value ratio cut-off value of 1.41, which was calculated with respect to the cerebellar grey matter. All baseline participants underwent multitracer positron emission tomography scans, cerebrospinal fluid biomarker analysis and neuropsychological assessment. Twenty-six of the participants underwent clinical and imaging follow-up examinations after 2.8 ± 0.6 years. By using linear mixed-effects models, fibrillar amyloid-β plaque deposition was first observed in the striatum of presymptomatic autosomal dominant Alzheimer’s disease carriers from 17 years before expected symptom onset; at about the same time, astrocytosis was significantly elevated and then steadily declined. Diverging from the astrocytosis pattern, amyloid-β plaque deposition increased with disease progression. Glucose metabolism steadily declined from 10 years after initial amyloid-β plaque deposition. Patients with sporadic mild cognitive impairment who were
11
C-Pittsburgh compound B-positive at baseline showed increasing amyloid-β plaque deposition and decreasing glucose metabolism but, in contrast to autosomal dominant Alzheimer’s disease carriers, there was no significant longitudinal decline in astrocytosis over time. The prominent initially high and then declining astrocytosis in autosomal dominant Alzheimer’s disease carriers, contrasting with the increasing amyloid-β plaque load during disease progression, suggests astrocyte activation is implicated in the early stages of Alzheimer’s disease pathology.
Collapse
Affiliation(s)
- Elena Rodriguez-Vieitez
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Laure Saint-Aubert
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Stephen F Carter
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Ove Almkvist
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden 2 Department of Psychology, Stockholm University, 106 91 Stockholm, Sweden 3 Department of Geriatric Medicine, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden
| | - Karim Farid
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Michael Schöll
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Konstantinos Chiotis
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Steinunn Thordardottir
- 3 Department of Geriatric Medicine, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden 4 Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Caroline Graff
- 3 Department of Geriatric Medicine, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden 4 Department NVS, Center for Alzheimer Research, Division of Neurogeriatrics, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden
| | - Anders Wall
- 5 Department of Surgical Sciences, Section of Nuclear Medicine & PET, Uppsala University, 751 85 Uppsala, Sweden
| | - Bengt Långström
- 6 Department of Chemistry, Uppsala University, 701 05 Uppsala, Sweden
| | - Agneta Nordberg
- 1 Department NVS, Centre for Alzheimer Research, Division of Translational Alzheimer Neurobiology, Karolinska Institutet, 141 57 Huddinge, Stockholm, Sweden 3 Department of Geriatric Medicine, Karolinska University Hospital Huddinge, 141 86, Stockholm, Sweden
| |
Collapse
|
134
|
Distinto S, Meleddu R, Yanez M, Cirilli R, Bianco G, Sanna ML, Arridu A, Cossu P, Cottiglia F, Faggi C, Ortuso F, Alcaro S, Maccioni E. Drug design, synthesis, in vitro and in silico evaluation of selective monoaminoxidase B inhibitors based on 3-acetyl-2-dichlorophenyl-5-aryl-2,3-dihydro-1,3,4-oxadiazole chemical scaffold. Eur J Med Chem 2016; 108:542-552. [DOI: 10.1016/j.ejmech.2015.12.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2015] [Revised: 12/10/2015] [Accepted: 12/12/2015] [Indexed: 12/18/2022]
|
135
|
|
136
|
Fowler JS, Logan J, Shumay E, Alia-Klein N, Wang GJ, Volkow ND. Monoamine oxidase: radiotracer chemistry and human studies. J Labelled Comp Radiopharm 2015; 58:51-64. [PMID: 25678277 DOI: 10.1002/jlcr.3247] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2014] [Accepted: 10/31/2014] [Indexed: 11/09/2022]
Abstract
Monoamine oxidase (MAO) oxidizes amines from both endogenous and exogenous sources thereby regulating the concentration of neurotransmitter amines such as serotonin, norepinephrine, and dopamine as well as many xenobiotics. MAO inhibitor drugs are used in the treatment of Parkinson's disease and in depression stimulating the development of radiotracer tools to probe the role of MAO in normal human biology and in disease. Over the past 30 years since the first radiotracers were developed and the first positron emission tomography (PET) images of MAO in humans were carried out, PET studies of brain MAO in healthy volunteers and in patients have identified different variables that have contributed to different MAO levels in brain and in peripheral organs. MAO radiotracers and PET have also been used to study the current and developing MAO inhibitor drugs including the selection of doses for clinical trials. In this article, we describe the following: (1) the development of MAO radiotracers; (2) human studies including the relationship of brain MAO levels to genotype, personality, neurological, and psychiatric disorders; and (3) examples of the use of MAO radiotracers in drug research and development. We will conclude with outstanding needs to improve the radiotracers that are currently used and possible new applications.
Collapse
Affiliation(s)
- Joanna S Fowler
- Biological, Environmental and Climate Sciences Department, Brookhaven National Laboratory, Upton, NY, USA
| | | | | | | | | | | |
Collapse
|
137
|
Multifunctional coumarin derivatives: Monoamine oxidase B (MAO-B) inhibition, anti-β-amyloid (Aβ) aggregation and metal chelation properties against Alzheimer’s disease. Bioorg Med Chem Lett 2015; 25:508-13. [DOI: 10.1016/j.bmcl.2014.12.034] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 12/10/2014] [Accepted: 12/11/2014] [Indexed: 01/11/2023]
|
138
|
Hicks JW, Sadovski O, Parkes J, Houle S, Hay BA, Carter RL, Wilson AA, Vasdev N. Radiosynthesis and ex vivo evaluation of [18F]-(S)-3-(6-(3-fluoropropoxy)benzo[d]isoxazol-3-yl)-5-(methoxymethyl)oxazolidin-2-one for imaging MAO-B with PET. Bioorg Med Chem Lett 2015; 25:288-91. [DOI: 10.1016/j.bmcl.2014.11.048] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2014] [Revised: 11/14/2014] [Accepted: 11/17/2014] [Indexed: 11/17/2022]
|
139
|
Pisani L, Farina R, Nicolotti O, Gadaleta D, Soto-Otero R, Catto M, Di Braccio M, Mendez-Alvarez E, Carotti A. In silico design of novel 2H-chromen-2-one derivatives as potent and selective MAO-B inhibitors. Eur J Med Chem 2015; 89:98-105. [DOI: 10.1016/j.ejmech.2014.10.029] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Revised: 10/07/2014] [Accepted: 10/12/2014] [Indexed: 10/24/2022]
|
140
|
Lan JS, Pan LF, Xie SS, Wang XB, Kong LY. Synthesis and evaluation of 6-methylcoumarin derivatives as potent and selective monoamine oxidase B inhibitors. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00437j] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Compound 5n was a potent and selective inhibitor of hMAO-B.
Collapse
Affiliation(s)
- Jin-Shuai Lan
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- People's Republic of China
| | - Long-Fei Pan
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- People's Republic of China
| | - Sai-Sai Xie
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- People's Republic of China
| | - Xiao-Bing Wang
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- People's Republic of China
| | - Ling-Yi Kong
- State Key Laboratory of Natural Medicines
- Department of Natural Medicinal Chemistry
- China Pharmaceutical University
- Nanjing 210009
- People's Republic of China
| |
Collapse
|
141
|
Musunuri S, Kultima K, Richard BC, Ingelsson M, Lannfelt L, Bergquist J, Shevchenko G. Micellar extraction possesses a new advantage for the analysis of Alzheimer's disease brain proteome. Anal Bioanal Chem 2014; 407:1041-57. [PMID: 25416231 DOI: 10.1007/s00216-014-8320-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Revised: 10/30/2014] [Accepted: 11/03/2014] [Indexed: 12/11/2022]
Abstract
Integral membrane proteins (MPs), such as transporters, receptors, and ion channels, are of great interest because of their participation in various vital cellular functions including cell-cell interactions, ion transport, and signal transduction. However, studies of MPs are complicated because of their hydrophobic nature, heterogeneity, and low abundance. Cloud-point extraction (CPE) with the non-ionic surfactant Triton X-114 was performed to simultaneously extract and phase separate hydrophobic and hydrophilic proteins from Alzheimer's disease (AD) and unaffected control brain tissue. Quantitative proteomics analysis of temporal neocortex samples of AD patients and controls was performed using a shotgun approach based on stable isotope dimethyl labeling (DML) quantification technique followed by nanoLC-MS/MS analysis. A total of 1096 unique proteins were identified and quantified, with 40.3 % (211/524) predicted as integral MPs with at least one transmembrane domain (TMD) found in the detergent phase, and 10 % (80/798) in the detergent-depleted phase. Among these, 62 proteins were shown to be significantly altered (p-value <0.05), in AD versus control samples. In the detergent fraction, we found 10 hydrophobic transmembrane proteins containing up to 14 putative TMDs that were significantly up- or down-regulated in AD compared with control brains. Changes in four of these proteins, alpha-enolase (ENOA), lysosome-associated membrane glycoprotein 1 (LAMP1), 14-3-3 protein gamma (1433G), and sarcoplasmic/endoplasmic reticulum calcium ATPase2 (AT2A2) were validated by immunoblotting. Our results emphasize that separating hydrophobic MPs in CPE contributes to an increased understanding of the underlying molecular mechanisms in AD. Such knowledge can become useful for the development of novel disease biomarkers.
Collapse
Affiliation(s)
- Sravani Musunuri
- Analytical Chemistry, Department of Chemistry-BMC, Uppsala University, Uppsala, Sweden
| | | | | | | | | | | | | |
Collapse
|
142
|
Imaging neuroinflammation in Alzheimer's disease and other dementias: Recent advances and future directions. Alzheimers Dement 2014; 11:1110-20. [DOI: 10.1016/j.jalz.2014.08.105] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 04/21/2014] [Accepted: 08/12/2014] [Indexed: 12/13/2022]
|
143
|
Amhaoul H, Staelens S, Dedeurwaerdere S. Imaging brain inflammation in epilepsy. Neuroscience 2014; 279:238-52. [DOI: 10.1016/j.neuroscience.2014.08.044] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2014] [Revised: 08/27/2014] [Accepted: 08/27/2014] [Indexed: 01/15/2023]
|
144
|
Neudorfer C, Shanab K, Jurik A, Schreiber V, Neudorfer C, Vraka C, Schirmer E, Holzer W, Ecker G, Mitterhauser M, Wadsak W, Spreitzer H. Development of potential selective and reversible pyrazoline based MAO-B inhibitors as MAO-B PET tracer precursors and reference substances for the early detection of Alzheimer’s disease. Bioorg Med Chem Lett 2014; 24:4490-4495. [DOI: 10.1016/j.bmcl.2014.07.085] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2014] [Revised: 07/25/2014] [Accepted: 07/30/2014] [Indexed: 11/26/2022]
|
145
|
Choo ILH, Carter SF, Schöll ML, Nordberg A. Astrocytosis measured by ¹¹C-deprenyl PET correlates with decrease in gray matter density in the parahippocampus of prodromal Alzheimer's patients. Eur J Nucl Med Mol Imaging 2014; 41:2120-6. [PMID: 25077930 DOI: 10.1007/s00259-014-2859-7] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2014] [Accepted: 07/04/2014] [Indexed: 01/01/2023]
Abstract
PURPOSE The Alzheimer's disease (AD) pathology is characterized by fibrillar amyloid deposits and neurofibrillary tangles, as well as the activation of astrocytosis, microglia activation, atrophy, dysfunctional synapse, and cognitive impairments. The aim of this study was to test the hypothesis that astrocytosis is correlated with reduced gray matter density in prodromal AD. METHODS Twenty patients with AD or mild cognitive impairment (MCI) underwent multi-tracer positron emission tomography (PET) studies with (11)C-Pittsburgh compound B ((11)C-PIB), (18) F-Fluorodeoxyglucose ((18) F-FDG), and (11)C-deuterium-L-deprenyl ((11)C-DED) PET imaging, as well as magnetic resonance imaging (MRI) scanning, cerebrospinal fluid (CSF) biomarker analysis, and neuropsychological assessments. The parahippocampus was selected as a region of interest, and each value was calculated for four different imaging modalities. Correlation analysis was applied between DED slope values and gray matter (GM) densities by MRI. To further explore possible relationships, correlation analyses were performed between the different variables, including the CSF biomarker. RESULTS A significant negative correlation was obtained between DED slope values and GM density in the parahippocampus in PIB-positive (PIB + ve) MCI patients (p = 0.025) (prodromal AD). Furthermore, in exploratory analyses, a positive correlation was observed between PIB-PET retention and DED binding in AD patients (p = 0.014), and a negative correlation was observed between PIB retention and CSF Aβ42 levels in MCI patients (p = 0.021), while the GM density and CSF total tau levels were negatively correlated in both PIB + ve MCI (p = 0.002) and MCI patients (p = 0.001). No significant correlation was observed with FDG-PET and with any of the other PET, MRI, or CSF biomarkers. CONCLUSIONS High astrocytosis levels in the parahippocampus of PIB + ve MCI (prodromal AD) patients suggest an early preclinical influence on cellular tissue loss. The lack of correlation between astrocytosis and CSF tau levels, and a positive correlation between astrocytosis and fibrillar amyloid deposition in clinical demented AD together indicate that parahippocampal astrocytosis might have some causality within the amyloid pathology.
Collapse
Affiliation(s)
- I L Han Choo
- Department NVS, Center for Alzheimer Research, Translational Alzheimer Neurobiology, Karolinska Institutet, Stockholm, Sweden,
| | | | | | | |
Collapse
|
146
|
Kersemans K, Van Laeken N, De Vos F. Radiochemistry devoted to the production of monoamine oxidase (MAO-A and MAO-B) ligands for brain imaging with positron emission tomography. J Labelled Comp Radiopharm 2014; 56:78-88. [PMID: 24285313 DOI: 10.1002/jlcr.3007] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2012] [Revised: 11/14/2012] [Accepted: 11/15/2012] [Indexed: 11/11/2022]
Abstract
Monoamine oxidase (MAO) belongs to a family of flavin-containing integral enzymes that are present in the outer mitochondrial membrane in neurons and glial cells in the central nervous system. These enzymes catalyze the oxidative deamination of various neurotransmitters, biogenic amines, and xenobiotics, thereby influencing their availability and physiological activity in brain and body. Over the past decades, many potential positron emission tomography tracers have been put forward to visualize MAO in the brain with varying success, and recent publications on the topic illustrate the continuing interest in the field. The present review gives an overview of the compounds that have been put forward as possible MAO tracers in the brain and focuses on the radiochemical procedures that have been developed to produce them up till now. Relevant radioligands are grouped by the main radiochemical strategies that have been employed to synthesize them, and some interesting details and findings that are crucial to the radiosyntheses are provided.
Collapse
Affiliation(s)
- Ken Kersemans
- Laboratory for Radiopharmacy, Gent University, Gent, Belgium
| | | | | |
Collapse
|
147
|
Youdim MBH, Kupershmidt L, Amit T, Weinreb O. Promises of novel multi-target neuroprotective and neurorestorative drugs for Parkinson's disease. Parkinsonism Relat Disord 2014; 20 Suppl 1:S132-6. [PMID: 24262165 DOI: 10.1016/s1353-8020(13)70032-4] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The cascade of neurotoxic events involved in neuronal degeneration suggests that it is naive to think mono-target drugs can induce disease modification by slowing the process of neurodegeneration in Parkinson's disease (PD). Employing the pharmacophore of rasagiline (N-propargyl-1-R-aminoindan), we have developed a series of novel multi-target neuroprotective drugs, including: (A) drugs [ladostigil, TV-3326 (N-propargyl-3R-aminoindan-5yl)-ethyl methylcarbamate)] with both cholinesterase-butyrylesterase (Ch-BuE) and brain-selective monamine oxidase-AB (MAO-AB) inhibitory activities and (B) iron chelator-radical scavenging drugs (M30) possessing brain-selective MAO-AB inhibitor activity and the neuroprotective-neurorescue propargylamine moiety of rasagiline. This was considered to be valid since brain MAO and iron increase in PD and aging, which could lead to oxidative stress-dependent neurodegeneration. The multi-target iron chelator, M30, has all the properties of ladostigil, but is not an acetylcholinesterase (CHE) inhibitor. However, M30 has both neuroprotective and neurorestorative activities for nigrostriatal dopamine neurons in post-lesion MPTP, lactacystin and 6-hydroxydopamine animal models of PD. The neurorestorative activity has been identified as being related to the ability of the drug to activate hypoxia-inducible factor (HIF) by inhibiting prolyl-4-hydroxylase. M30 regulates cell cycle arrest and induces the neurotrophins brain-derived neurotrophic factor (BDNF), vascular endothelial growth factor (VEGF), erythropoietin (EPO), as well as glia-derived neurotrophic factor (GDNF). These unique multiple actions of M30 make it potentially useful as a disease modifying drug for the treatment of PD.
Collapse
Affiliation(s)
- Moussa B H Youdim
- Technion-Rappaport Faculty of Medicine, Eve Topf and NPF Centers of Excellence for Neurodegenerative Diseases, Haifa, Israel Abital Pharma Pipeline Ltd., Tel Aviv, Israel.
| | | | | | | |
Collapse
|
148
|
Tracking neuroinflammation in Alzheimer's disease: the role of positron emission tomography imaging. J Neuroinflammation 2014; 11:120. [PMID: 25005532 PMCID: PMC4099095 DOI: 10.1186/1742-2094-11-120] [Citation(s) in RCA: 84] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2014] [Accepted: 06/20/2014] [Indexed: 12/02/2022] Open
Abstract
Alzheimer’s disease (AD) has been reconceptualized as a dynamic pathophysiological process, where the accumulation of amyloid-beta (Aβ) is thought to trigger a cascade of neurodegenerative events resulting in cognitive impairment and, eventually, dementia. In addition to Aβ pathology, various lines of research have implicated neuroinflammation as an important participant in AD pathophysiology. Currently, neuroinflammation can be measured in vivo using positron emission tomography (PET) with ligands targeting diverse biological processes such as microglial activation, reactive astrocytes and phospholipase A2 activity. In terms of therapeutic strategies, despite a strong rationale and epidemiological studies suggesting that the use of non-steroidal anti-inflammatory drugs (NSAIDs) may reduce the prevalence of AD, clinical trials conducted to date have proven inconclusive. In this respect, it has been hypothesized that NSAIDs may only prove protective if administered early on in the disease course, prior to the accumulation of significant AD pathology. In order to test various hypotheses pertaining to the exact role of neuroinflammation in AD, studies in asymptomatic carriers of mutations deterministic for early-onset familial AD may prove of use. In this respect, PET ligands for neuroinflammation may act as surrogate markers of disease progression, allowing for the development of more integrative models of AD, as well as for the measuring of target engagement in the context of clinical trials using NSAIDs. In this review, we address the biological basis of neuroinflammatory changes in AD, underscore therapeutic strategies using anti-inflammatory compounds, and shed light on the possibility of tracking neuroinflammation in vivo using PET imaging ligands.
Collapse
|
149
|
Jo S, Yarishkin O, Hwang YJ, Chun YE, Park M, Woo DH, Bae JY, Kim T, Lee J, Chun H, Park HJ, Lee DY, Hong J, Kim HY, Oh SJ, Park SJ, Lee H, Yoon BE, Kim Y, Jeong Y, Shim I, Bae YC, Cho J, Kowall NW, Ryu H, Hwang E, Kim D, Lee CJ. GABA from reactive astrocytes impairs memory in mouse models of Alzheimer's disease. Nat Med 2014; 20:886-96. [PMID: 24973918 DOI: 10.1038/nm.3639] [Citation(s) in RCA: 581] [Impact Index Per Article: 52.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Accepted: 06/23/2014] [Indexed: 12/11/2022]
Abstract
In Alzheimer's disease (AD), memory impairment is the most prominent feature that afflicts patients and their families. Although reactive astrocytes have been observed around amyloid plaques since the disease was first described, their role in memory impairment has been poorly understood. Here, we show that reactive astrocytes aberrantly and abundantly produce the inhibitory gliotransmitter GABA by monoamine oxidase-B (Maob) and abnormally release GABA through the bestrophin 1 channel. In the dentate gyrus of mouse models of AD, the released GABA reduces spike probability of granule cells by acting on presynaptic GABA receptors. Suppressing GABA production or release from reactive astrocytes fully restores the impaired spike probability, synaptic plasticity, and learning and memory in the mice. In the postmortem brain of individuals with AD, astrocytic GABA and MAOB are significantly upregulated. We propose that selective inhibition of astrocytic GABA synthesis or release may serve as an effective therapeutic strategy for treating memory impairment in AD.
Collapse
Affiliation(s)
- Seonmi Jo
- 1] Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea. [2] WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea. [3]
| | - Oleg Yarishkin
- 1] WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea. [2]
| | - Yu Jin Hwang
- Center for Neuro-Medicine, Brain Science Institute, KIST, Seoul, Republic of Korea
| | - Ye Eun Chun
- 1] WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea. [2] Neuroscience Program, Korea University of Science and Technology, Daejeon, Republic of Korea
| | - Mijeong Park
- 1] Neuroscience Program, Korea University of Science and Technology, Daejeon, Republic of Korea. [2] Center for Neuroscience, Brain Science Institute, KIST, Seoul, Republic of Korea
| | - Dong Ho Woo
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jin Young Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Taekeun Kim
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jaekwang Lee
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Heejung Chun
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyun Jung Park
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Da Yong Lee
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jinpyo Hong
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hye Yun Kim
- Center for Neuro-Medicine, Brain Science Institute, KIST, Seoul, Republic of Korea
| | - Soo-Jin Oh
- Center for Neuroscience, Brain Science Institute, KIST, Seoul, Republic of Korea
| | - Seung Ju Park
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Hyo Lee
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Bo-Eun Yoon
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - YoungSoo Kim
- Center for Neuro-Medicine, Brain Science Institute, KIST, Seoul, Republic of Korea
| | - Yong Jeong
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Insop Shim
- Department of Science in Korean Medicine, Graduate School, College of Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Yong Chul Bae
- Department of Anatomy and Neurobiology, School of Dentistry, Kyungpook National University, Daegu, Republic of Korea
| | - Jeiwon Cho
- 1] Neuroscience Program, Korea University of Science and Technology, Daejeon, Republic of Korea. [2] Center for Neuroscience, Brain Science Institute, KIST, Seoul, Republic of Korea
| | - Neil W Kowall
- 1] Boston University Alzheimer's Disease Center, Boston University School of Medicine, Boston, Massachusetts, USA. [2] Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA. [3] VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Hoon Ryu
- 1] Center for Neuro-Medicine, Brain Science Institute, KIST, Seoul, Republic of Korea. [2] Boston University Alzheimer's Disease Center, Boston University School of Medicine, Boston, Massachusetts, USA. [3] Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA. [4] VA Boston Healthcare System, Boston, Massachusetts, USA
| | - Eunmi Hwang
- WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Daesoo Kim
- Department of Biological Sciences, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - C Justin Lee
- 1] WCI Center for Functional Connectomics, Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea. [2] Neuroscience Program, Korea University of Science and Technology, Daejeon, Republic of Korea. [3] Center for Neuroscience, Brain Science Institute, KIST, Seoul, Republic of Korea. [4] KU-KIST Graduate School of Converging Science of Technology, Korea University, Seoul, Republic of Korea
| |
Collapse
|
150
|
Andrade JM, Passos CDS, Dresch RR, Kieling-Rubio MA, Moreno PRH, Henriques AT. Chemical analysis, antioxidant, antichemotactic and monoamine oxidase inhibition effects of some pteridophytes from Brazil. Pharmacogn Mag 2014; 10:S100-9. [PMID: 24914288 PMCID: PMC4047592 DOI: 10.4103/0973-1296.127354] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2013] [Revised: 07/07/2013] [Accepted: 02/21/2014] [Indexed: 01/09/2023] Open
Abstract
Background: Ferns are a group of plants that have been little explored from a chemical and biological perspective but that have interesting potential, occurring in various parts of the world. Objective: This work investigates the chemical profile and the biological effects of ferns from Brazil. Materials and Methods: Analyses were performed using rapid performance liquid chromatography (RP-LC) with a diode array detector (DAD). Extracts were tested for their in vitro antioxidant activity, by the total reactive antioxidant potential method and for their antichemotactic potential, by the Boyden chamber method. Cytotoxic effects were assessed by lactate dehydrogenase levels, while the monoamine oxidase (MAO) assay was carried out using a fluorescence-based method. Results: Different chemical compositions were found for the studied ferns, such as Asplenium gastonis, in which hesperidin was identified in its extract, while A. serra showed the presence of xanthone mangiferin. The most samples with highest antioxidant activity were the Asplenium serra, Lastreopsis amplissima and Cyathea dichromatolepis extracts, at 10 μg/mL. High antichemotactic activity was found for A. serra (94.06%) and Didymochlaena truncatula (93.41%), at 10 μg/mL. The extracts showed no cytotoxicity at the highest concentration. Against MAO-A, D. truncatula (82.61%), Alsophila setosa (82.21%), Cyathea phalerata (74.07%) and C. delgadii (70.32%) were the most active extracts (100 μg/mL). Conclusion: The hypothesis was considered that phenolics and triterpenes are responsible for these pronounced activities.
Collapse
Affiliation(s)
- Juliana Mm Andrade
- Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Av. Ipiranga 2752, 90.610-000, Porto Alegre, RS, Brazil
| | - Carolina Dos S Passos
- Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Av. Ipiranga 2752, 90.610-000, Porto Alegre, RS, Brazil
| | - Roger R Dresch
- Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Av. Ipiranga 2752, 90.610-000, Porto Alegre, RS, Brazil
| | | | - Paulo Roberto H Moreno
- Department of Fundamental Chemistry, University of São Paulo (USP), Av. Prof. Lineu Prestes 748 B11, 05.508-000, São Paulo, SP, Brazil
| | - Amélia T Henriques
- Faculty of Pharmacy, Federal University of Rio Grande do Sul (UFRGS), Av. Ipiranga 2752, 90.610-000, Porto Alegre, RS, Brazil
| |
Collapse
|