251
|
Hampel H, Schneider LS, Giacobini E, Kivipelto M, Sindi S, Dubois B, Broich K, Nisticò R, Aisen PS, Lista S. Advances in the therapy of Alzheimer's disease: targeting amyloid beta and tau and perspectives for the future. Expert Rev Neurother 2014; 15:83-105. [PMID: 25537424 DOI: 10.1586/14737175.2015.995637] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Worldwide multidisciplinary translational research has led to a growing knowledge of the genetics and molecular pathogenesis of Alzheimer's disease (AD) indicating that pathophysiological brain alterations occur decades before clinical signs and symptoms of cognitive decline can be diagnosed. Consequently, therapeutic concepts and targets have been increasingly focused on early-stage illness before the onset of dementia; and distinct classes of compounds are now being tested in clinical trials. At present, there is a growing consensus that therapeutic progress in AD delaying disease progression would significantly decrease the expanding global burden. The evolving hypothesis- and evidence-based generation of new diagnostic research criteria for early-stage AD has positively impacted the development of clinical trial designs and the characterization of earlier and more specific target populations for trials in prodromal as well as in pre- and asymptomatic at-risk stages of AD.
Collapse
|
252
|
Cárdenas-Aguayo MDC, Gómez-Virgilio L, DeRosa S, Meraz-Ríos MA. The role of tau oligomers in the onset of Alzheimer's disease neuropathology. ACS Chem Neurosci 2014; 5:1178-91. [PMID: 25268947 DOI: 10.1021/cn500148z] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Most neurodegenerative diseases are characterized by the presence of protein aggregates. Alzheimer's disease (AD) is the most common cause of dementia in people over age 60. One of the histopathological hallmarks of AD is the presence of tau protein aggregates. Historically, it has been thought that paired helical filaments (PHFs) were the toxic form of tau that assembled to form neurofibrillary tangles (NFTs), but recently there has been evidence that tau oligomers, which form before PHFs and NFTs, could be the structures mediating neurodegeneration even before the fibrillary tau is deposited. Here, we discuss the recent advances in tau oligomer research, their implications on AD and other tauopathies, the mechanisms of tau turnover by the principal protein clearance systems (the proteasome and autophagy), and the potential use of tau oligomers as drug targets for the development of new therapeutic approaches.
Collapse
Affiliation(s)
- María del Carmen Cárdenas-Aguayo
- Molecular
Biomedicine Department, CINVESTAV-IPN, Ave. Politécnico 2508, Colonia
San Pedro Zacatenco, México City, D.F. 07360, México
| | - Laura Gómez-Virgilio
- Molecular
Biomedicine Department, CINVESTAV-IPN, Ave. Politécnico 2508, Colonia
San Pedro Zacatenco, México City, D.F. 07360, México
| | - Steven DeRosa
- Center
for Dementia Research, Nathan S. Kline Institute for Psychiatric Research, 140 Old Orangeburg Road, Orangeburg, New York 10962, United States
| | - Marco Antonio Meraz-Ríos
- Molecular
Biomedicine Department, CINVESTAV-IPN, Ave. Politécnico 2508, Colonia
San Pedro Zacatenco, México City, D.F. 07360, México
| |
Collapse
|
253
|
Brinet D, Kaffy J, Oukacine F, Glumm S, Ongeri S, Taverna M. An improved capillary electrophoresis method for in vitro monitoring of the challenging early steps of Aβ1-42 peptide oligomerization: application to anti-Alzheimer's drug discovery. Electrophoresis 2014; 35:3302-9. [PMID: 25219962 DOI: 10.1002/elps.201400271] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2014] [Revised: 08/22/2014] [Accepted: 08/22/2014] [Indexed: 11/06/2022]
Abstract
We report an improved CE method to monitor in vitro the self-assembly of monomeric amyloid β-peptide (42 amino acids amyloid β-peptide, Aβ1-42 ) and in particular the crucial early steps involved in the formation of the neurotoxic oligomers. In order to start the kinetics from the beginning, sample preparation was optimized to provide samples containing exclusively the monomeric form. The CE method was also improved using a dynamic coating and by reducing the separation distance. Using this method, the disappearance of the monomer as well as the progressive formation of four species during the self-assembly process can now be monitored and quantified over time. The hydrodynamic radius of the species present at the initial kinetics step was estimated around 1.8 nm by Taylor dispersion analysis while SDS-PAGE analyses showed the predominance of the monomer. These results confirmed that the Aβ1-42 species present at this initial time was the monomer. Methylene blue, an anti-Alzheimer disease candidate, was then evaluated. In spite of an oligomerization inhibition, the enhanced disappearance of the Aβ1-42 monomer provoked by methylene blue was demonstrated for the first time. This method, allowing the monomeric and smallest oligomeric species to be monitored, represents a new accurate and precise way to evaluate compounds for drug discovery.
Collapse
Affiliation(s)
- Dimitri Brinet
- Protéines et Nanotechnologies en Sciences Séparatives, Institut Galien de Paris Sud, Faculté de Pharmacie, Université Paris-Sud, Châtenay-Malabry, France; Molécules Fluorées et Chimie Médicinale, Faculté de Pharmacie, Université Paris-Sud, Châtenay-Malabry, France
| | | | | | | | | | | |
Collapse
|
254
|
Schilling LP, Leuzy A, Zimmer ER, Gauthier S, Rosa-Neto P. Nonamyloid PET biomarkers and Alzheimer's disease: current and future perspectives. FUTURE NEUROLOGY 2014. [DOI: 10.2217/fnl.14.40] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
ABSTRACT Recent advances in neurobiology and PET have helped redefine Alzheimer's disease (AD) as a dynamic pathophysiological process, clinically characterized by preclinical, mild cognitive impairment due to AD and dementia stages. Though a majority of PET studies conducted within these populations have to date focused on β-amyloid, various ‘nonamyloid’ radiopharmaceuticals exist for evaluating neurodegeneration, neuroinflammation and perturbations in neurotransmission across the spectrum of AD. Importantly, findings using such tracers have been shown to correlate with various clinical, cognitive and behavioral measures. In the context of a growing shift toward early diagnosis and symptomatic and disease-modifying clinical trials, nonamyloid PET radiotracers will prove of use, and, potentially, contribute to improved therapeutic prospects for AD.
Collapse
Affiliation(s)
- Lucas Porcello Schilling
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
- Alzheimer's Disease Research Unit, McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
- Brain Institute of Rio Grande do Sul, Pontifical Catholic University of Rio Grande do Sul (PUCRS), Porto Alegre, Brazil
| | - Antoine Leuzy
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
- Alzheimer's Disease Research Unit, McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
| | - Eduardo Rigon Zimmer
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
- Alzheimer's Disease Research Unit, McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
- Department of Biochemistry, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Serge Gauthier
- Alzheimer's Disease Research Unit, McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
| | - Pedro Rosa-Neto
- Translational Neuroimaging Laboratory (TNL), McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
- Alzheimer's Disease Research Unit, McGill Center for Studies in Aging (MCSA), Douglas Mental Health Research Institute, Montreal, Canada
| |
Collapse
|
255
|
Fuse S, Matsumura K, Fujita Y, Sugimoto H, Takahashi T. Development of dual targeting inhibitors against aggregations of amyloid-β and tau protein. Eur J Med Chem 2014; 85:228-34. [DOI: 10.1016/j.ejmech.2014.07.095] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2014] [Revised: 07/25/2014] [Accepted: 07/25/2014] [Indexed: 11/28/2022]
|
256
|
Mori T, Koyama N, Segawa T, Maeda M, Maruyama N, Kinoshita N, Hou H, Tan J, Town T. Methylene blue modulates β-secretase, reverses cerebral amyloidosis, and improves cognition in transgenic mice. J Biol Chem 2014; 289:30303-30317. [PMID: 25157105 DOI: 10.1074/jbc.m114.568212] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
Amyloid precursor protein (APP) proteolysis is required for production of amyloid-β (Aβ) peptides that comprise β-amyloid plaques in the brains of patients with Alzheimer disease (AD). Here, we tested whether the experimental agent methylene blue (MB), used for treatment of methemoglobinemia, might improve AD-like pathology and behavioral deficits. We orally administered MB to the aged transgenic PSAPP mouse model of cerebral amyloidosis and evaluated cognitive function and cerebral amyloid pathology. Beginning at 15 months of age, animals were gavaged with MB (3 mg/kg) or vehicle once daily for 3 months. MB treatment significantly prevented transgene-associated behavioral impairment, including hyperactivity, decreased object recognition, and defective spatial working and reference memory, but it did not alter nontransgenic mouse behavior. Moreover, brain parenchymal and cerebral vascular β-amyloid deposits as well as levels of various Aβ species, including oligomers, were mitigated in MB-treated PSAPP mice. These effects occurred with inhibition of amyloidogenic APP proteolysis. Specifically, β-carboxyl-terminal APP fragment and β-site APP cleaving enzyme 1 protein expression and activity were attenuated. Additionally, treatment of Chinese hamster ovary cells overexpressing human wild-type APP with MB significantly decreased Aβ production and amyloidogenic APP proteolysis. These results underscore the potential for oral MB treatment against AD-related cerebral amyloidosis by modulating the amyloidogenic pathway.
Collapse
Affiliation(s)
- Takashi Mori
- Departments of Biomedical Sciences and Saitama Medical Center and University, Kawagoe, Saitama 350-8550, Japan; Departments of Pathology, Saitama Medical Center and University, Kawagoe, Saitama 350-8550, Japan,.
| | - Naoki Koyama
- Departments of Biomedical Sciences and Saitama Medical Center and University, Kawagoe, Saitama 350-8550, Japan
| | - Tatsuya Segawa
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Masahiro Maeda
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Nobuhiro Maruyama
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Noriaki Kinoshita
- Immuno-Biological Laboratories Co., Ltd., Fujioka, Gunma 375-0005, Japan
| | - Huayan Hou
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center and University of South Florida, Tampa, Florida 33613
| | - Jun Tan
- Rashid Laboratory for Developmental Neurobiology, Silver Child Development Center and University of South Florida, Tampa, Florida 33613; Neuroimmunology Laboratory, Department of Psychiatry and Behavioral Neurosciences, Morsoni College of Medicine, University of South Florida, Tampa, Florida 33613, and
| | - Terrence Town
- Zilkha Neurogenetic Institute, Department of Physiology and Biophysics, Keck School of Medicine, University of Southern California, Los Angeles, California 90089-2821.
| |
Collapse
|
257
|
Gheyara AL, Ponnusamy R, Djukic B, Craft RJ, Ho K, Guo W, Finucane MM, Sanchez PE, Mucke L. Tau reduction prevents disease in a mouse model of Dravet syndrome. Ann Neurol 2014; 76:443-56. [PMID: 25042160 PMCID: PMC4338764 DOI: 10.1002/ana.24230] [Citation(s) in RCA: 109] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2014] [Revised: 07/08/2014] [Accepted: 07/09/2014] [Indexed: 12/12/2022]
Abstract
Objective Reducing levels of the microtubule-associated protein tau has shown promise as a potential treatment strategy for diseases with secondary epileptic features such as Alzheimer disease. We wanted to determine whether tau reduction may also be of benefit in intractable genetic epilepsies. Methods We studied a mouse model of Dravet syndrome, a severe childhood epilepsy caused by mutations in the human SCN1A gene encoding the voltage-gated sodium channel subunit Nav1.1. We genetically deleted 1 or 2 Tau alleles in mice carrying an Nav1.1 truncation mutation (R1407X) that causes Dravet syndrome in humans, and examined their survival, epileptic activity, related hippocampal alterations, and behavioral abnormalities using observation, electroencephalographic recordings, acute slice electrophysiology, immunohistochemistry, and behavioral assays. Results Tau ablation prevented the high mortality of Dravet mice and reduced the frequency of spontaneous and febrile seizures. It reduced interictal epileptic spikes in vivo and drug-induced epileptic activity in brain slices ex vivo. Tau ablation also prevented biochemical changes in the hippocampus indicative of epileptic activity and ameliorated abnormalities in learning and memory, nest building, and open field behaviors in Dravet mice. Deletion of only 1 Tau allele was sufficient to suppress epileptic activity and improve survival and nesting performance. Interpretation Tau reduction may be of therapeutic benefit in Dravet syndrome and other intractable genetic epilepsies. Ann Neurol 2014;76:443–456
Collapse
Affiliation(s)
- Ania L Gheyara
- Gladstone Institute of Neurological Disease, University of California, San Francisco, San Francisco, CA; Departments of Pathology, University of California, San Francisco, San Francisco, CA
| | | | | | | | | | | | | | | | | |
Collapse
|
258
|
Kovacic P, Somanathan R. Toxicity of imine-iminium dyes and pigments: electron transfer, radicals, oxidative stress and other physiological effects. J Appl Toxicol 2014; 34:825-34. [PMID: 24852913 DOI: 10.1002/jat.3005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 02/10/2014] [Indexed: 12/12/2022]
Abstract
Although conjugation is well known as an important contributor to color, there is scant recognition concerning involvement of imine and iminium functions in the physiological effects of this class of dyes and pigments. The group includes the dyes methylene blue, rhodamine, malachite green, fuchsin, crystal violet, auramine and cyanins, in addition to the pigments consisting of pyocyanine, phthalocyanine and pheophytin. The physiological effects consist of both toxicity and beneficial aspects. The unifying theme of electron transfer-reactive oxygen species-oxidative stress is used as the rationale in both cases. Toxicity is frequently prevented or alleviated by antioxidants. The apparent dichotomy of methylene blue action as both oxidant and antioxidant is rationalized based on similar previous cases. This mechanistic approach may have practical benefit. This review is important in conveying, for the first time, a unifying mechanism for toxicity based on electron transfer-reactive oxygen species-oxidative stress arising from imine-iminium.
Collapse
Affiliation(s)
- Peter Kovacic
- Department of Chemistry and Biochemistry, San Diego State University, San Diego, CA, USA
| | | |
Collapse
|
259
|
Potential therapeutic strategies for Alzheimer's disease targeting or beyond β-amyloid: insights from clinical trials. BIOMED RESEARCH INTERNATIONAL 2014; 2014:837157. [PMID: 25136630 PMCID: PMC4124758 DOI: 10.1155/2014/837157] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2014] [Revised: 06/23/2014] [Accepted: 06/25/2014] [Indexed: 01/25/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder with two hallmarks: β-amyloid plagues and neurofibrillary tangles. It is one of the most alarming illnesses to elderly people. No effective drugs and therapies have been developed, while mechanism-based explorations of therapeutic approaches have been intensively investigated. Outcomes of clinical trials suggested several pitfalls in the choice of biomarkers, development of drug candidates, and interaction of drug-targeted molecules; however, they also aroused concerns on the potential deficiency in our understanding of pathogenesis of AD, and ultimately stimulated the advent of novel drug targets tests. The anticipated increase of AD patients in next few decades makes development of better therapy an urgent issue. Here we attempt to summarize and compare putative therapeutic strategies that have completed clinical trials or are currently being tested from various perspectives to provide insights for treatments of Alzheimer's disease.
Collapse
|
260
|
Boutajangout A, Wisniewski T. Tau-based therapeutic approaches for Alzheimer's disease - a mini-review. Gerontology 2014; 60:381-5. [PMID: 24732638 PMCID: PMC4149810 DOI: 10.1159/000358875] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2013] [Accepted: 01/21/2014] [Indexed: 11/19/2022] Open
Abstract
The accumulation of aggregated, hyperphosphorylated tau as neurofibrillary tangles and neuropil threads are cardinal features of Alzheimer's disease (AD). The other lesions found in AD include amyloid plaques and congophilic amyloid angiopathy, both associated with the extracellular accumulation of the amyloid-beta (Aβ) peptide. AD is the most common cause of dementia globally. Currently, there are no effective means to treat AD or even to slow it down. The dominant theory for the causation of AD is the amyloid cascade hypothesis, which suggests that the aggregation of Aβ as oligomers and amyloid plaques is central to the pathogenesis of AD. Numerous therapies have been developed directed to Aβ-related pathology, in particular various immunotherapeutic approaches. So far all of these have failed in clinical trials. Recently, there has been more focus on therapy directed to tau-related pathology, which correlates better with the cognitive status of patients, compared to the amyloid burden. Immunotherapeutic targeting of tau pathology has shown great potential in treating tau pathologies in mouse models of AD. A number of studies have shown the efficacy of both passive and active immunization. This review summarizes recent advances in therapy targeting pathological tau protein, in particular focusing on immunotherapeutic approaches which are showing great promise.
Collapse
Affiliation(s)
- Allal Boutajangout
- Department of Neurology, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Psychiatry, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Physiology & Neuroscience, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
| | - Thomas Wisniewski
- Department of Neurology, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Pathology, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
- Department of Psychiatry, New York University School of Medicine, 450 East 29 Street, New York. NY. 10016
| |
Collapse
|
261
|
Gonzalez-Lima F, Barksdale BR, Rojas JC. Mitochondrial respiration as a target for neuroprotection and cognitive enhancement. Biochem Pharmacol 2014; 88:584-93. [DOI: 10.1016/j.bcp.2013.11.010] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2013] [Revised: 11/16/2013] [Accepted: 11/18/2013] [Indexed: 10/25/2022]
|
262
|
Fan LY, Chiu MJ. Combotherapy and current concepts as well as future strategies for the treatment of Alzheimer's disease. Neuropsychiatr Dis Treat 2014; 10:439-51. [PMID: 24648738 PMCID: PMC3956689 DOI: 10.2147/ndt.s45143] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
It has been estimated that 35.6 million people globally had dementia in 2010 and the prevalence of dementia has been predicted to double every 20 years. Thus, 115.4 million people may be living with dementia in 2050. Alzheimer's disease (AD) is the leading cause of dementia and is present in 60%-70% of people with dementia. Unfortunately, there are few approved drugs that can alleviate the cognitive or behavioral symptoms of AD dementia. Recent studies have revealed that pathophysiological changes related to AD occur decades before the appearance of clinical symptoms of dementia. This extended preclinical phase of AD provides a critical chance for disease-modifying agents to halt or delay the relentless process of AD. Although several trials targeting various pathological processes are ongoing, the examination of the combined use of different approaches to combat AD seems warranted. In this article, we will review current therapies, future strategies, and ongoing clinical trials for the treatment of AD with a special focus on combination therapies. Furthermore, preventive strategies for cognitively normal subjects in the presymptomatic stages of AD will also be addressed. In this review, we discuss current hypotheses of the disease process. In the decades since the approval of cholinesterase inhibitors, no new drug has ultimately demonstrated clear success in clinical trials. Given the difficulties that have been encountered in attempts to identify a single drug that can treat AD, we must pursue effective multi-target strategies, ie, combination therapies. The combination of cholinesterase inhibitors and memantine is considered well tolerated and safe, and this combination benefits patients with moderate-to-severe AD. In contrast, with the exception of adjuvant therapies of conventional drugs, combinations of different disease-modifying agents with different mechanisms may have promising synergic effects and benefit cognition, behavior, and daily living function.
Collapse
Affiliation(s)
- Ling-Yun Fan
- Department of Neurology, En Chu Kong Hospital, Taipei, Taiwan
- Department of Neurology, College of Medicine, Graduate Institute of Brain and Mind Sciences, Taipei, Taiwan
| | - Ming-Jang Chiu
- Department of Neurology, College of Medicine, Graduate Institute of Brain and Mind Sciences, Taipei, Taiwan
- Graduate Institute of Psychology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Biomedical Engineering and Bioinformatics, National Taiwan University Hospital, Taipei, Taiwan
| |
Collapse
|
263
|
Stack C, Jainuddin S, Elipenahli C, Gerges M, Starkova N, Starkov AA, Jové M, Portero-Otin M, Launay N, Pujol A, Kaidery NA, Thomas B, Tampellini D, Beal MF, Dumont M. Methylene blue upregulates Nrf2/ARE genes and prevents tau-related neurotoxicity. Hum Mol Genet 2014; 23:3716-32. [PMID: 24556215 DOI: 10.1093/hmg/ddu080] [Citation(s) in RCA: 113] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Methylene blue (MB, methylthioninium chloride) is a phenothiazine that crosses the blood brain barrier and acts as a redox cycler. Among its beneficial properties are its abilities to act as an antioxidant, to reduce tau protein aggregation and to improve energy metabolism. These actions are of particular interest for the treatment of neurodegenerative diseases with tau protein aggregates known as tauopathies. The present study examined the effects of MB in the P301S mouse model of tauopathy. Both 4 mg/kg MB (low dose) and 40 mg/kg MB (high dose) were administered in the diet ad libitum from 1 to 10 months of age. We assessed behavior, tau pathology, oxidative damage, inflammation and numbers of mitochondria. MB improved the behavioral abnormalities and reduced tau pathology, inflammation and oxidative damage in the P301S mice. These beneficial effects were associated with increased expression of genes regulated by NF-E2-related factor 2 (Nrf2)/antioxidant response element (ARE), which play an important role in antioxidant defenses, preventing protein aggregation, and reducing inflammation. The activation of Nrf2/ARE genes is neuroprotective in other transgenic mouse models of neurodegenerative diseases and it appears to be an important mediator of the neuroprotective effects of MB in P301S mice. Moreover, we used Nrf2 knock out fibroblasts to show that the upregulation of Nrf2/ARE genes by MB is Nrf2 dependent and not due to secondary effects of the compound. These findings provide further evidence that MB has important neuroprotective effects that may be beneficial in the treatment of human neurodegenerative diseases with tau pathology.
Collapse
Affiliation(s)
- Cliona Stack
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Shari Jainuddin
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Ceyhan Elipenahli
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Meri Gerges
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Natalia Starkova
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Anatoly A Starkov
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Mariona Jové
- Department de Medicina Experimental, Universitat de Lleida-IRBLLEIDA, Spain
| | | | - Nathalie Launay
- Neurometabolic Diseases Laboratory-IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Spain, CIBERER, Spanish Network for Rare Diseases, ISCIII, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory-IDIBELL, Hospital Duran i Reynals, 08908 L'Hospitalet de Llobregat, Barcelona, Spain, CIBERER, Spanish Network for Rare Diseases, ISCIII, Spain, ICREA, Catalan Institution for Research and Advanced Studies, Spain
| | - Navneet Ammal Kaidery
- Department of Pharmacology and Toxicology and Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Bobby Thomas
- Department of Pharmacology and Toxicology and Department of Neurology, Medical College of Georgia, Georgia Regents University, Augusta, GA 30912, USA
| | - Davide Tampellini
- Hospital Kremlin Bicêtre, UMR 788, Institut National de la Santé et de la Recherche Médicale (INSERM), Université Paris Sud, Le Kremlin Bicêtre, France and
| | - M Flint Beal
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA
| | - Magali Dumont
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA, IHU-A-ICM, Hospital Pitié-Salpêtrière, 75013 Paris, France
| |
Collapse
|
264
|
In vivo evaluation of a novel tau imaging tracer for Alzheimer's disease. Eur J Nucl Med Mol Imaging 2014; 41:816-26. [PMID: 24514874 DOI: 10.1007/s00259-013-2681-7] [Citation(s) in RCA: 131] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2013] [Accepted: 12/20/2013] [Indexed: 10/25/2022]
Abstract
PURPOSE Diagnosis of tauopathies such as Alzheimer's disease (AD) still relies on post-mortem examination of the human brain. A non-invasive method of determining brain tau burden in vivo would allow a better understanding of the pathophysiology of tauopathies. The purpose of the study was to evaluate (18)F-THK523 as a potential tau imaging tracer. METHODS Ten healthy elderly controls, three semantic dementia (SD) and ten AD patients underwent neuropsychological examination, MRI as well as (18)F-THK523 and (11)C-Pittsburgh compound B (PIB) positron emission tomography (PET) scans. Composite memory and non-memory scores, global and hippocampal brain volume, and partial volume-corrected tissue ratios for (18)F-THK523 and (11)C-PIB were estimated for all participants. Correlational analyses were performed between global and regional (18)F-THK523, (11)C-PIB, cognition and brain volumetrics. RESULTS (18)F-THK523 presented with fast reversible kinetics. Significantly higher (18)F-THK523 retention was observed in the temporal, parietal, orbitofrontal and hippocampi of AD patients when compared to healthy controls and SD patients. White matter retention was significantly higher than grey matter retention in all participants. The pattern of cortical (18)F-THK523 retention did not correlate with Aβ distribution as assessed by (11)C-PIB and followed the known distribution of tau in the AD brain, being higher in temporal and parietal areas than in the frontal region. Unlike (11)C-PIB, hippocampal (18)F-THK523 retention was correlated with several cognitive parameters and with hippocampal atrophy. CONCLUSION (18)F-THK523 does not bind to Aβ in vivo, while following the known distribution of paired helical filaments (PHF)-tau in the brain. Significantly higher cortical (18)F-THK523 retention in AD patients as well as the association of hippocampal (18)F-THK523 retention with cognitive parameters and hippocampal volume suggests (18)F-THK523 selectively binds to tau in AD patients. Unfortunately, the very high (18)F-THK523 retention in white matter precludes simple visual inspection of the images, preventing its use in research or clinical settings.
Collapse
|
265
|
Petzer A, Harvey BH, Petzer JP. The interactions of azure B, a metabolite of methylene blue, with acetylcholinesterase and butyrylcholinesterase. Toxicol Appl Pharmacol 2014; 274:488-93. [DOI: 10.1016/j.taap.2013.10.014] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 10/04/2013] [Accepted: 10/14/2013] [Indexed: 02/06/2023]
|
266
|
Spires-Jones TL, Friedman T, Pitstick R, Polydoro M, Roe A, Carlson GA, Hyman BT. Methylene blue does not reverse existing neurofibrillary tangle pathology in the rTg4510 mouse model of tauopathy. Neurosci Lett 2014; 562:63-8. [PMID: 24462887 DOI: 10.1016/j.neulet.2014.01.013] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2013] [Revised: 12/18/2013] [Accepted: 01/10/2014] [Indexed: 10/25/2022]
Abstract
Alzheimer's disease is characterized pathologically by aggregation of amyloid beta into senile plaques and aggregation of pathologically modified tau into neurofibrillary tangles. While changes in amyloid processing are strongly implicated in disease initiation, the recent failure of amyloid-based therapies has highlighted the importance of tau as a therapeutic target. "Tangle busting" compounds including methylene blue and analogous molecules are currently being evaluated as therapeutics in Alzheimer's disease. Previous studies indicated that methylene blue can reverse tau aggregation in vitro after 10 min, and subsequent studies suggested that high levels of drug reduce tau protein levels (assessed biochemically) in vivo. Here, we tested whether methylene blue could remove established neurofibrillary tangles in the rTg4510 model of tauopathy, which develops robust tangle pathology. We find that 6 weeks of methylene blue dosing in the water from 16 months to 17.5 months of age decreases soluble tau but does not remove sarkosyl insoluble tau, or histologically defined PHF1 or Gallyas positive tangle pathology. These data indicate that methylene blue treatment will likely not rapidly reverse existing tangle pathology.
Collapse
Affiliation(s)
- Tara L Spires-Jones
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA.
| | - Taylor Friedman
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | | - Manuela Polydoro
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | - Allyson Roe
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| | | | - Bradley T Hyman
- Massachusetts General Hospital, 114 16th Street, Charlestown, MA 02129, USA
| |
Collapse
|
267
|
Léger GC, Massoud F. Novel disease-modifying therapeutics for the treatment of Alzheimer’s disease. Expert Rev Clin Pharmacol 2014; 6:423-42. [DOI: 10.1586/17512433.2013.811237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
268
|
Guerrero-Muñoz MJ, Castillo-Carranza DL, Kayed R. Therapeutic approaches against common structural features of toxic oligomers shared by multiple amyloidogenic proteins. Biochem Pharmacol 2014; 88:468-78. [PMID: 24406245 DOI: 10.1016/j.bcp.2013.12.023] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Revised: 12/18/2013] [Accepted: 12/19/2013] [Indexed: 02/03/2023]
Abstract
Impaired proteostasis is one of the main features of all amyloid diseases, which are associated with the formation of insoluble aggregates from amyloidogenic proteins. The aggregation process can be caused by overproduction or poor clearance of these proteins. However, numerous reports suggest that amyloid oligomers are the most toxic species, rather than insoluble fibrillar material, in Alzheimer's, Parkinson's, and Prion diseases, among others. Although the exact protein that aggregates varies between amyloid disorders, they all share common structural features that can be used as therapeutic targets. In this review, we focus on therapeutic approaches against shared features of toxic oligomeric structures and future directions.
Collapse
Affiliation(s)
- Marcos J Guerrero-Muñoz
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Diana L Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|
269
|
Affiliation(s)
- Karen Chiang
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093; ,
| | - Edward H. Koo
- Department of Neurosciences, University of California, San Diego, La Jolla, California 92093; ,
| |
Collapse
|
270
|
Paranjape SR, Chiang YM, Sanchez JF, Entwistle R, Wang CCC, Oakley BR, Gamblin TC. Inhibition of Tau aggregation by three Aspergillus nidulans secondary metabolites: 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde. PLANTA MEDICA 2014; 80:77-85. [PMID: 24414310 PMCID: PMC6442474 DOI: 10.1055/s-0033-1360180] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
The aggregation of the microtubule-associated protein tau is a significant event in many neurodegenerative diseases including Alzheimer's disease. The inhibition or reversal of tau aggregation is therefore a potential therapeutic strategy for these diseases. Fungal natural products have proven to be a rich source of useful compounds having wide varieties of biological activity. We have screened Aspergillus nidulans secondary metabolites containing aromatic ring structures for their ability to inhibit tau aggregation in vitro using an arachidonic acid polymerization protocol and the previously identified aggregation inhibitor emodin as a positive control. While several compounds showed some activity, 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde were potent aggregation inhibitors as determined by both a filter trap assay and electron microscopy. In this study, these three compounds were stronger inhibitors than emodin, which has been shown in a prior study to inhibit the heparin induction of tau aggregation with an IC50 of 1-5 µM. Additionally, 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde reduced, but did not block, tau stabilization of microtubules. 2,ω-Dihydroxyemodin and asperthecin have similar structures to previously identified tau aggregation inhibitors, while asperbenzaldehyde represents a new class of compounds with tau aggregation inhibitor activity. Asperbenzaldehyde can be readily modified into compounds with strong lipoxygenase inhibitor activity, suggesting that compounds derived from asperbenzaldehyde could have dual activity. Together, our data demonstrates the potential of 2,ω-dihydroxyemodin, asperthecin, and asperbenzaldehyde as lead compounds for further development as therapeutics to inhibit tau aggregation in Alzheimer's disease and neurodegenerative tauopathies.
Collapse
Affiliation(s)
- Smita R. Paranjape
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave., Lawrence, KS 66045, USA
| | - Yi-Ming Chiang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
- Graduate Institute of Pharmaceutical Science, Chia Nan University School of Pharmacy and Science, Tainan 71710, Taiwan
| | - James F. Sanchez
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
| | - Ruth Entwistle
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave., Lawrence, KS 66045, USA
| | - Clay C. C. Wang
- Department of Pharmacology and Pharmaceutical Sciences, School of Pharmacy, University of Southern California, Los Angeles, CA 90089, USA
- Department of Chemistry, Dornsife College of Letters, Arts, and Sciences, University of Southern California, Los Angeles, CA 90089, USA
| | - Berl R. Oakley
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave., Lawrence, KS 66045, USA
| | - T. Chris Gamblin
- Department of Molecular Biosciences, University of Kansas, 1200 Sunnyside Ave., Lawrence, KS 66045, USA
| |
Collapse
|
271
|
Wischik CM, Harrington CR, Storey JMD. Tau-aggregation inhibitor therapy for Alzheimer's disease. Biochem Pharmacol 2013; 88:529-39. [PMID: 24361915 DOI: 10.1016/j.bcp.2013.12.008] [Citation(s) in RCA: 190] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2013] [Revised: 12/08/2013] [Accepted: 12/09/2013] [Indexed: 12/22/2022]
Abstract
Many trials of drugs aimed at preventing or clearing β-amyloid pathology have failed to demonstrate efficacy in recent years and further trials continue with drugs aimed at the same targets and mechanisms. The Alzheimer neurofibrillary tangle is composed of tau and the core of its constituent filaments are made of a truncated fragment from the repeat domain of tau. This truncated tau can catalyse the conversion of normal soluble tau into aggregated oligomeric and fibrillar tau which, in turn, can spread to neighbouring neurons. Tau aggregation is not a late-life process and onset of Braak stage 1 peaks in people in their late 40s or early 50s. Tau aggregation pathology at Braak stage 1 or beyond affects 50% of the population over the age of 45. The initiation of tau aggregation requires its binding to a non-specific substrate to expose a high affinity tau-tau binding domain and it is self-propagating thereafter. The initiating substrate complex is most likely formed as a consequence of a progressive loss of endosomal-lysosomal processing of neuronal proteins, particularly of membrane proteins from mitochondria. Mutations in the APP/presenilin membrane complex may simply add to the age-related endosomal-lysosomal processing failure, bringing forward, but not directly causing, the tau aggregation cascade in carriers. Methylthioninium chloride (MTC), the first identified tau aggregation inhibitor (TAI), offers an alternative to the amyloid approach. Phase 3 trials are underway with a novel stabilized reduced form of methylthioninium (LMTX) that has improved tolerability and absorption.
Collapse
Affiliation(s)
- Claude M Wischik
- TauRx Therapeutics Ltd., Singapore; School of Medicine and Dentistry, University of Aberdeen, Scotland, United Kingdom.
| | - Charles R Harrington
- TauRx Therapeutics Ltd., Singapore; School of Medicine and Dentistry, University of Aberdeen, Scotland, United Kingdom
| | - John M D Storey
- TauRx Therapeutics Ltd., Singapore; Department of Chemistry, University of Aberdeen, Scotland, United Kingdom
| |
Collapse
|
272
|
Investigation on the aggregation behaviors and filament morphology of tau protein by a simple 90° angle light-scattering assay. ScientificWorldJournal 2013; 2013:354730. [PMID: 24163620 PMCID: PMC3791826 DOI: 10.1155/2013/354730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 08/06/2013] [Indexed: 11/24/2022] Open
Abstract
The in vitro aggregation of tau constructs was monitored by a simple 90° angle light-scattering (LS) approach which was conducted directly on fluorescence instrument. At the optimum incident wavelength (550 nm, unpolarized), the sensitivity of LS was high enough to detect tau aggregation at micromolar range. The nucleation and elongation, different events in the aggregation process of 4RMBD construct (corresponding with the four repeated units of tau Microtubule Binding Domain) could be observed by this approach, as compared with ThS fluorescence assay. The validity of this technique was demonstrated over a range of tau concentrations with different tau filaments. Linear regression of scattering light against concentration yielded the x-intercept, the critical concentrations of tau constructs. The critical concentrations of 4RMBD and its S305N mutant are 5.26 μM and 4.04 μM respectively, indicating point mutation S305N, which is associated with FTDP-17, appear to enhance the heparin-induced tau aggregation in vitro. Furthermore, the slopes of concentration dependence curves, as well as the angle dependence, were discussed based on the filaments morphology examined by electron microscopy and ultrasonication experiment.
Collapse
|
273
|
Kim SJ, Ha DJ, Koo TS. Simultaneous quantification of methylene blue and its major metabolite, azure B, in plasma by LC-MS/MS and its application for a pharmacokinetic study. Biomed Chromatogr 2013; 28:518-24. [PMID: 24122875 DOI: 10.1002/bmc.3063] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2013] [Revised: 08/06/2013] [Accepted: 09/09/2013] [Indexed: 11/06/2022]
Abstract
A simple and sensitive liquid chromatography-tandem mass spectrometry (LC-MS/MS) method was developed for the quantification of methylene blue (MB) and its major metabolite, azure B (AZB), in rat plasma. A simple protein precipitation using acetonitrile was followed by injection of the supernatant on to a Zorbax HILIC Plus column (3.5 µm, 2.1 × 100 mm) with isocratic mobile phase consisting of 5 mM ammonium acetate in 10:90 (v/v) water:methanol at a flow rate of 0.3 mL/min and detection in positive ionization mode. The standard curve was linear over the concentration range from 1 to 1000 ng/mL for MB and AZB with coefficient of determination above 0.9930. The lower limit of quantification was 1 ng/mL using 20 μL of rat plasma sample. The intra- and inter-assay precision and accuracy were <12%. The developed analytical method was successfully applied to the pharmacokinetic study of MB and AZB in rats.
Collapse
Affiliation(s)
- Soo-Jin Kim
- Translational Medicine, Life Science R&D Park, SK Biopharmaceuticals, Daejeon, 305-712, Korea
| | | | | |
Collapse
|
274
|
Aprahamian I, Stella F, Forlenza OV. New treatment strategies for Alzheimer's disease: is there a hope? Indian J Med Res 2013; 138:449-60. [PMID: 24434253 PMCID: PMC3868059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2013] [Indexed: 10/29/2022] Open
Abstract
Alzheimer's disease (AD) is a progressive and irreversible neurodegenerative disease, and corresponds to the most common cause of dementia worldwide. Although not fully understood, the pathophysiology of AD is largely represented by the neurotoxic events triggered by the beta-amyloid cascade and by cytoskeletal abnormalities subsequent to the hyperphosphorylation of microtubule-associated Tau protein in neurons. These processes lead respectively to the formation of neuritic plaques and neurofibrillary tangles, which are the pathological hallmarks of the disease. Clinical benefits of the available pharmacological treatment for AD with antidementia drugs (namely cholinesterase inhibitors and memantine) are unquestionable, although limited to a temporary, symptomatic support to cognitive and related functions. Over the past decade, substantial funding and research have been dedicated to the search and development of new pharmaceutical compounds with disease-modifying properties. The rationale of such approach is that by tackling key pathological processes in AD it may be possible to attenuate or even change its natural history. In the present review, we summarize the available evidence on the new therapeutic approaches that target amyloid and Tau pathology in AD, focusing on pharmaceutical compounds undergoing phase 2 and phase 3 randomized controlled trials.
Collapse
Affiliation(s)
- Ivan Aprahamian
- Laboratory of Neuroscience (LIM 27) Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Brazil
| | - Florindo Stella
- Laboratory of Neuroscience (LIM 27) Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Brazil
- UNESP, Biosciences Institute, Campus of Rio Claro-SP, Brazil
| | - Orestes V. Forlenza
- Laboratory of Neuroscience (LIM 27) Department & Institute of Psychiatry, Faculty of Medicine, University of São Paulo, Brazil
| |
Collapse
|
275
|
Schafer KN, Cisek K, Huseby CJ, Chang E, Kuret J. Structural determinants of Tau aggregation inhibitor potency. J Biol Chem 2013; 288:32599-32611. [PMID: 24072703 DOI: 10.1074/jbc.m113.503474] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Small-molecule Tau aggregation inhibitors are under investigation as potential therapeutic agents against Alzheimer disease. Many such inhibitors have been identified in vitro, but their potency-driving features, and their molecular targets in the Tau aggregation pathway, have resisted identification. Previously we proposed ligand polarizability, a measure of electron delocalization, as a candidate descriptor of inhibitor potency. Here we tested this hypothesis by correlating the ground state polarizabilities of cyanine, phenothiazine, and arylmethine derivatives calculated using ab initio quantum methods with inhibitory potency values determined in the presence of octadecyl sulfate inducer under reducing conditions. A series of rhodanine analogs was analyzed as well using potency values disclosed in the literature. Results showed that polarizability and inhibitory potency directly correlated within all four series. To identify putative binding targets, representative members of the four chemotypes were added to aggregation reactions, where they were found to stabilize soluble, but SDS-resistant Tau species at the expense of filamentous aggregates. Using SDS resistance as a secondary assay, and a library of Tau deletion and missense mutants as targets, interaction with cyanine was localized to the microtubule binding repeat region. Moreover, the SDS-resistant phenotype was completely dependent on the presence of octadecyl sulfate inducer, but not intact PHF6/PH6* hexapeptide motifs, indicating that cyanine interacted with a species in the aggregation pathway prior to nucleus formation. Together the data suggest that flat, highly polarizable ligands inhibit Tau aggregation by interacting with folded species in the aggregation pathway and driving their assembly into soluble but highly stable Tau oligomers.
Collapse
Affiliation(s)
- Kelsey N Schafer
- From the Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Katryna Cisek
- From the Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Carol J Huseby
- From the Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Edward Chang
- From the Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio 43210
| | - Jeff Kuret
- From the Department of Molecular and Cellular Biochemistry, College of Medicine, The Ohio State University, Columbus, Ohio 43210.
| |
Collapse
|
276
|
Potential synergy between tau aggregation inhibitors and tau chaperone modulators. ALZHEIMERS RESEARCH & THERAPY 2013; 5:41. [PMID: 24041111 PMCID: PMC3979086 DOI: 10.1186/alzrt207] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Tau is a soluble, microtubule-associated protein known to aberrantly form
amyloid-positive aggregates. This pathology is characteristic for more than 15
neuropathies, the most common of which is Alzheimer’s disease. Finding
therapeutics to reverse or remove this non-native tau state is of great
interest; however, at this time only one drug is entering phase III clinical
trials for treating tauopathies. Generally, tau manipulation by therapeutics can
either directly or indirectly alter tau aggregation and stability. Drugs that
bind and change the conformation of tau itself are largely classified as
aggregation inhibitors, while drugs that alter the activity of a tau-effector
protein fall into several categories, such as kinase inhibitors, microtubule
stabilizers, or chaperone modulators. Chaperone inhibitors that have proven
effective in tau models include heat shock protein 90 inhibitors, heat shock
protein 70 inhibitors and activators, as well as inducers of heat shock
proteins. While many of these compounds can alter tau levels and/or aggregation
states, it is possible that combining these approaches may produce the most
optimal outcome. However, because many of these compounds have multiple
off-target effects or poor blood–brain barrier permeability, the
development of this synergistic therapeutic strategy presents significant
challenges. This review will summarize many of the drugs that have been
identified to alter tau biology, with special focus on therapeutics that prevent
tau aggregation and regulate chaperone-mediated clearance of tau.
Collapse
|
277
|
Appleby BS, Nacopoulos D, Milano N, Zhong K, Cummings JL. A review: treatment of Alzheimer's disease discovered in repurposed agents. Dement Geriatr Cogn Disord 2013; 35:1-22. [PMID: 23307039 DOI: 10.1159/000345791] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/13/2012] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND/AIMS Many compounds that have already been approved for alternate diagnoses have been studied in relation to Alzheimer's disease (AD). The purpose of this review is to summarize these studies and discuss the rationale and benefits of repurposing drugs for AD treatment. METHODS Studies of drugs related to AD treatment that were relevant to a disease-modifying mechanism of action (MOA) and are already approved by the Food and Drug Administration for non-AD diagnoses were collected from PubMed. RESULTS Many drugs already approved for the treatment of other diseases have been studied in relation to AD treatment. Numerous drugs with known toxicity profiles have the potential to be repurposed as a treatment for AD. CONCLUSION Known MOA, toxicology, and pharmacodynamic profiles would accelerate the process and increase the odds of finding a more timely disease-modifying treatment for AD.
Collapse
Affiliation(s)
- Brian S Appleby
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland, OH, USA
| | | | | | | | | |
Collapse
|
278
|
Abstract
The pathway leading from soluble and monomeric to hyperphosphorylated, insoluble and filamentous tau protein is at the centre of many human neurodegenerative diseases, collectively referred to as tauopathies. Dominantly inherited mutations in MAPT, the gene that encodes tau, cause forms of frontotemporal dementia and parkinsonism, proving that dysfunction of tau is sufficient to cause neurodegeneration and dementia. However, most cases of tauopathy are not inherited in a dominant manner. The first tau aggregates form in a few nerve cells in discrete brain areas. These become self propagating and spread to distant brain regions in a prion-like manner. The prevention of tau aggregation and propagation is the focus of attempts to develop mechanism-based treatments for tauopathies.
Collapse
Affiliation(s)
- Maria Grazia Spillantini
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
279
|
Weinstein JD, Gonzalez ER, Egleton RD, Hunt DA. A Paradigm Shift for Evaluating Pharmacotherapy for Alzheimer's Disease: The 10-Patient Screening Protocol. ACTA ACUST UNITED AC 2013; 28:443-54. [DOI: 10.4140/tcp.n.2013.443] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
280
|
Selectivity of phenothiazine cholinesterase inhibitors for neurotransmitter systems. Bioorg Med Chem Lett 2013; 23:3822-5. [DOI: 10.1016/j.bmcl.2013.04.082] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2013] [Revised: 04/23/2013] [Accepted: 04/29/2013] [Indexed: 11/22/2022]
|
281
|
Abstract
Protein misfolding disorders, such as Alzheimer's disease and Parkinson's disease, have in common that a protein accumulates in an insoluble form in the affected tissue. The process of aggregation follows a mechanism of seeded polymerization. Although the toxic species is still not well defined, the process, rather than the end product, of fibril formation is likely the main culprit in amyloid toxicity. These findings suggest that therapeutic strategies directed against the protein misfolding cascade should focus on depleting aggregation intermediates rather than on large fibrillar aggregates. Recent studies involving natural compounds have suggested new intervention strategies. The polyphenol epi-gallocatechine-3-gallate (EGCG), the main polyphenol in Camilla sinensis, binds directly to a large number of proteins that are involved in protein misfolding diseases and inhibits their fibrillization. Instead, it promotes the formation of stable, spherical aggregates. These spherical aggregates are not cytotoxic, have a lower β-sheet content than fibrils, and do not catalyze fibril formation. Correspondingly, epi-gallocatechine-3-gallate remodels amyloid fibrils into aggregates with the same properties. Derivatives of Orcein, which is a phenoxazine dye that can be isolated from the lichen Roccella tinctoria, form a second promising class of natural compounds. They accelerate fibril formation of the Alzheimer's disease-related amyloid-beta peptide. At the same time these compounds deplete oligomeric and protofibrillar forms of the peptide. These compounds may serve as proof-of-principle for the strategies of promoting and redirecting fibril formation. Both may emerge as two promising new therapeutic approaches to intervening into protein misfolding processes.
Collapse
Affiliation(s)
- Jan Bieschke
- Department of Biomedical Engineering, Washington University in St Louis, One Brookings Drive, St Louis, MO 63130, USA.
| |
Collapse
|
282
|
Yoshiyama Y, Lee VMY, Trojanowski JQ. Therapeutic strategies for tau mediated neurodegeneration. J Neurol Neurosurg Psychiatry 2013; 84:784-95. [PMID: 23085937 PMCID: PMC3912572 DOI: 10.1136/jnnp-2012-303144] [Citation(s) in RCA: 95] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Based on the amyloid hypothesis, controlling β-amyloid protein (Aβ) accumulation is supposed to suppress downstream pathological events, tau accumulation, neurodegeneration and cognitive decline. However, in recent clinical trials, Aβ removal or reducing Aβ production has shown limited efficacy. Moreover, while active immunisation with Aβ resulted in the clearance of Aβ, it did not prevent tau pathology or neurodegeneration. This prompts the concern that it might be too late to employ Aβ targeting therapies once tau mediated neurodegeneration has occurred. Therefore, it is timely and very important to develop tau directed therapies. The pathomechanisms of tau mediated neurodegeneration are unclear but hyperphosphorylation, oligomerisation, fibrillisation and propagation of tau pathology have been proposed as the likely pathological processes that induce loss of function or gain of toxic function of tau, causing neurodegeneration. Here we review the strategies for tau directed treatments based on recent progress in research on tau and our understanding of the pathomechanisms of tau mediated neurodegeneration.
Collapse
Affiliation(s)
- Yasumasa Yoshiyama
- Department of Neurology, Chiba East National Hospital, 673 Nitona, Chuo Ward, Chiba, Chiba 260-8712, Japan.
| | | | | |
Collapse
|
283
|
Bulic B, Pickhardt M, Mandelkow E. Progress and developments in tau aggregation inhibitors for Alzheimer disease. J Med Chem 2013; 56:4135-55. [PMID: 23484434 DOI: 10.1021/jm3017317] [Citation(s) in RCA: 87] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Pharmacological approaches directed toward Alzheimer disease are diversifying in parallel with a growing number of promising targets. Investigations on the microtubule-associated protein tau yielded innovative targets backed by recent findings about the central role of tau in numerous neurodegenerative diseases. In this review, we summarize the recent evolution in the development of nonpeptidic small molecules tau aggregation inhibitors (TAGIs) and their advancement toward clinical trials. The compounds are classified according to their chemical structures, providing correlative insights into their pharmacology. Overall, shared structure-activity traits are emerging, as well as specific binding modes related to their ability to engage in hydrogen bonding. Medicinal chemistry efforts on TAGIs together with encouraging in vivo data argue for successful translation to the clinic.
Collapse
Affiliation(s)
- Bruno Bulic
- Laboratory of Organic Synthesis of Functional Systems, Humboldt-Universität zu Berlin, Brook-Taylor-Strasse 2, 12489 Berlin, Germany.
| | | | | |
Collapse
|
284
|
Ramachandran G, Udgaonkar JB. Mechanistic studies unravel the complexity inherent in tau aggregation leading to Alzheimer's disease and the tauopathies. Biochemistry 2013; 52:4107-26. [PMID: 23721410 DOI: 10.1021/bi400209z] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The aggregation of the protein tau into amyloid fibrils is known to be involved in the causation of the neurodegenerative tauopathies and the progression of cognitive decline in Alzheimer's disease. This review surveys the mechanism of tau aggregation with special emphasis on the information obtained from biochemical and biophysical studies. First, tau is described from a structure-function perspective. Subsequently, the connection of tau to neurodegeneration is explained, and a description of the tau amyloid fibril is provided. Lastly, studies of the mechanism of tau fibril formation are reviewed, and the physiological significance of these studies with reference to how they can clarify many aspects of disease progression is described. The aim of this review is to underscore how mechanistic studies reveal the complexity of the tau fibril formation pathway and the plethora of species populated on or off the pathway of aggregation, and how this information can be beneficial in the design of inhibitors or drugs that ameliorate neurodegeneration.
Collapse
Affiliation(s)
- Gayathri Ramachandran
- National Centre for Biological Sciences, Tata Institute of Fundamental Research , Bangalore 560065, India
| | | |
Collapse
|
285
|
Clavaguera F, Lavenir I, Falcon B, Frank S, Goedert M, Tolnay M. "Prion-like" templated misfolding in tauopathies. Brain Pathol 2013; 23:342-9. [PMID: 23587140 PMCID: PMC8028860 DOI: 10.1111/bpa.12044] [Citation(s) in RCA: 100] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2013] [Accepted: 02/11/2013] [Indexed: 12/20/2022] Open
Abstract
The soluble microtubule-associated protein tau forms hyperphosphorylated, insoluble and filamentous inclusions in a number of neurodegenerative diseases referred to as "tauopathies." In Alzheimer's disease, tau pathology develops in a stereotypical manner, with the first lesions appearing in the locus coeruleus and entorhinal cortex, from where they appear to spread to the hippocampus and neocortex. Propagation of tau pathology is also a characteristic of argyrophilic grain disease, where the tau lesions spread throughout the limbic system. Significantly, isoform composition and morphology of tau filaments can differ between tauopathies, suggesting the existence of distinct tau strains. Extensive experimental findings indicate that prion-like mechanisms underly the pathogenesis of tauopathies.
Collapse
Affiliation(s)
- Florence Clavaguera
- Department of NeuropathologyInstitute of PathologyUniversity Hospital BaselBaselSwitzerland
| | | | - Ben Falcon
- MRC Laboratory of Molecular BiologyCambridgeUK
| | - Stephan Frank
- Department of NeuropathologyInstitute of PathologyUniversity Hospital BaselBaselSwitzerland
| | | | - Markus Tolnay
- Department of NeuropathologyInstitute of PathologyUniversity Hospital BaselBaselSwitzerland
| |
Collapse
|
286
|
Trippier PC, Labby KJ, Hawker DD, Mataka JJ, Silverman RB. Target- and mechanism-based therapeutics for neurodegenerative diseases: strength in numbers. J Med Chem 2013; 56:3121-47. [PMID: 23458846 PMCID: PMC3637880 DOI: 10.1021/jm3015926] [Citation(s) in RCA: 113] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The development of new therapeutics for the treatment of neurodegenerative pathophysiologies currently stands at a crossroads. This presents an opportunity to transition future drug discovery efforts to target disease modification, an area in which much still remains unknown. In this Perspective we examine recent progress in the areas of neurodegenerative drug discovery, focusing on some of the most common targets and mechanisms: N-methyl-d-aspartic acid (NMDA) receptors, voltage gated calcium channels (VGCCs), neuronal nitric oxide synthase (nNOS), oxidative stress from reactive oxygen species, and protein aggregation. These represent the key players identified in neurodegeneration and are part of a complex, intertwined signaling cascade. The synergistic delivery of two or more compounds directed against these targets, along with the design of small molecules with multiple modes of action, should be explored in pursuit of more effective clinical treatments for neurodegenerative diseases.
Collapse
Affiliation(s)
- Paul C. Trippier
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Kristin Jansen Labby
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Dustin D. Hawker
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Jan J. Mataka
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
| | - Richard B. Silverman
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208-3113, USA
- Department of Molecular Biosciences, Chemistry of Life Processes Institute, Center for Molecular Innovation and Drug Discovery, Northwestern University, Evanston, IL, USA
| |
Collapse
|
287
|
Crowe A, James MJ, Lee VMY, Smith AB, Trojanowski JQ, Ballatore C, Brunden KR. Aminothienopyridazines and methylene blue affect Tau fibrillization via cysteine oxidation. J Biol Chem 2013; 288:11024-37. [PMID: 23443659 DOI: 10.1074/jbc.m112.436006] [Citation(s) in RCA: 111] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Alzheimer disease and several other neurodegenerative disorders are characterized by the accumulation of intraneuronal fibrils comprised of the protein Tau. Tau is normally a soluble protein that stabilizes microtubules, with splice isoforms that contain either three (3-R) or four (4-R) microtubule binding repeats. The formation of Tau fibrils is thought to result in neuronal damage, and inhibitors of Tau fibrillization may hold promise as therapeutic agents. The process of Tau fibrillization can be replicated in vitro, and a number of small molecules have been identified that inhibit Tau fibril formation. However, little is known about how these molecules affect Tau fibrillization. Here, we examined the mechanism by which the previously described aminothieno pyridazine (ATPZ) series of compounds inhibit Tau fibrillization. Active ATPZs were found to promote the oxidation of the two cysteine residues within 4-R Tau by a redox cycling mechanism, resulting in the formation of a disulfide-containing compact monomer that was refractory to fibrillization. Moreover, the ATPZs facilitated intermolecular disulfide formation between 3-R Tau monomers, leading to dimers that were capable of fibrillization. The ATPZs also caused cysteine oxidation in molecules unrelated to Tau. Interestingly, methylene blue, an inhibitor of Tau fibrillization under evaluation in Alzheimer disease clinical trials, caused a similar oxidation of cysteines in Tau and other molecules. These findings reveal that the ATPZs and methylene blue act by a mechanism that may affect their viability as potential therapeutic agents.
Collapse
Affiliation(s)
- Alex Crowe
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
288
|
Phenotypic identification of the redox dye methylene blue as an antagonist of heat shock response gene expression in metastatic melanoma cells. Int J Mol Sci 2013; 14:4185-202. [PMID: 23429201 PMCID: PMC3588094 DOI: 10.3390/ijms14024185] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2013] [Revised: 01/24/2013] [Accepted: 01/29/2013] [Indexed: 11/17/2022] Open
Abstract
Repurposing approved and abandoned non-oncological drugs is an alternative developmental strategy for the identification of anticancer therapeutics that has recently attracted considerable attention. Due to the essential role of the cellular heat shock response in cytoprotection through the maintenance of proteostasis and suppression of apoptosis, small molecule heat shock response antagonists can be harnessed for targeted induction of cytotoxic effects in cancer cells. Guided by gene expression array analysis and a phenotypic screen interrogating a collection of 3,7-diamino-phenothiazinium derivatives, we have identified the redox-drug methylene blue (MB), used clinically for the infusional treatment of methemoglobinemia, as a negative modulator of heat shock response gene expression in human metastatic melanoma cells. MB-treatment blocked thermal (43 °C) and pharmacological (celastrol, geldanamycin) induction of heat shock response gene expression, suppressing Hsp70 (HSPA1A) and Hsp27 (HSPB1) upregulation at the mRNA and protein level. MB sensitized melanoma cells to the apoptogenic activity of geldanamycin, an Hsp90 antagonist known to induce the counter-regulatory upregulation of Hsp70 expression underlying cancer cell resistance to geldanamycin chemotherapy. Similarly, MB-cotreatment sensitized melanoma cells to other chemotherapeutics (etoposide, doxorubicin). Taken together, these data suggest feasibility of repurposing the non-oncological redox drug MB as a therapeutic heat shock response antagonist for cancer cell chemosensitization.
Collapse
|
289
|
Akoury E, Pickhardt M, Gajda M, Biernat J, Mandelkow E, Zweckstetter M. Mechanistic basis of phenothiazine-driven inhibition of Tau aggregation. Angew Chem Int Ed Engl 2013; 52:3511-5. [PMID: 23401175 DOI: 10.1002/anie.201208290] [Citation(s) in RCA: 115] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2012] [Revised: 12/14/2012] [Indexed: 01/05/2023]
Affiliation(s)
- Elias Akoury
- Max-Planck-Institut für Biophysikalische Chemie, Am Fassberg 11, 37077 Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
290
|
Akoury E, Pickhardt M, Gajda M, Biernat J, Mandelkow E, Zweckstetter M. Mechanismus der Phenothiazin-induzierten Hemmung der Tau-Aggregation. Angew Chem Int Ed Engl 2013. [DOI: 10.1002/ange.201208290] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
|
291
|
Akoury E, Gajda M, Pickhardt M, Biernat J, Soraya P, Griesinger C, Mandelkow E, Zweckstetter M. Inhibition of Tau Filament Formation by Conformational Modulation. J Am Chem Soc 2013; 135:2853-62. [DOI: 10.1021/ja312471h] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Elias Akoury
- Department for NMR-based Structural
Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen,
Germany
| | - Michal Gajda
- Department for NMR-based Structural
Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen,
Germany
| | - Marcus Pickhardt
- German Center for
Neurodegenerative
Diseases (DZNE), Ludwig−Erhard−Allee 2, 53175 Bonn,
Germany
| | - Jacek Biernat
- German Center for
Neurodegenerative
Diseases (DZNE), Ludwig−Erhard−Allee 2, 53175 Bonn,
Germany
| | - Pornsuwan Soraya
- RG Electron Spin Resonance Spectroscopy,
Max Planck Institute for Biophysical Chemistry, Am Fassberg 11, 37077
Göttingen, Germany
| | - Christian Griesinger
- Department for NMR-based Structural
Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen,
Germany
| | - Eckhard Mandelkow
- German Center for
Neurodegenerative
Diseases (DZNE), Ludwig−Erhard−Allee 2, 53175 Bonn,
Germany
- CAESAR Research Center,
Ludwig−Erhard−Allee
2, 53175 Bonn, Germany
| | - Markus Zweckstetter
- Department for NMR-based Structural
Biology, Max Planck Institute for Biophysical Chemistry, 37077 Göttingen,
Germany
- German Center for Neurodegenerative
Diseases (DZNE), Göttingen, Germany
| |
Collapse
|
292
|
IRITANI SHUJI. What happens in the brain of schizophrenia patients?: an investigation from the viewpoint of neuropathology. NAGOYA JOURNAL OF MEDICAL SCIENCE 2013; 75:11-28. [PMID: 23544264 PMCID: PMC4345712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/05/2022]
Abstract
Studies that seek to determine the etiology of schizophrenia through pathological images and morphological abnormalities of the brain have been conducted since the era of E. Kraepelin, and pioneers in neuropathology such as A. Alzheimer have also eagerly pursued such studies. However, there have been no disease-specific findings, and there was a brief era in which it was said that "schizophrenia is the graveyard of neuropathologists." However, since the 1980s, neuroimaging studies with CT and MRI etc., have been used in many reports of cases of schizophrenia with abnormal brain morphology, thus generating renewed interest in developments within brain tissue and leading to new neuropathological studies. There are now many reports in which, in addition to morphological observations, cell distribution and the like are image-processed and statistically processed through computers. Due to methodological problems in making progress in the field of cerebral pathology, we have not yet been able to observe disease-specific findings, although there are several findings with high certainty. However, the neurodevelopmental hypothesis has been supported as being able to reasonably explain the accumulated findings of previous studies. At the same time, results of recent molecular-biological studies have revealed the risk genes for this disease, and because many of those genes are associated with functions related to nerve differentiation, development, and plasticity, there is growing interest in their correlations with cerebral pathology. We are now on the verge of uncovering the etiology of this disease by integrating cerebral neuroimaging, molecular genetics, and cerebral neuropathology. In that sense, neuropathological studies of this disease from new viewpoints have become essential.
Collapse
Affiliation(s)
- SHUJI IRITANI
- Department of Psychiatry, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
293
|
Natural products as a rich source of tau-targeting drugs for Alzheimer's disease. Future Med Chem 2013; 4:1751-61. [PMID: 22924511 DOI: 10.4155/fmc.12.124] [Citation(s) in RCA: 73] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder and the most common form of dementia, affecting more than 5.4 million people in the USA. Although the cause of AD is not well understood, the cholinergic, amyloid and tau hypotheses were proposed to explain its development. Drug discovery for AD based on the cholinergic and amyloid theories have not been effective. In this article we summarize tau-based natural products as AD therapeutics from a variety of biological sources, including the anti-amyloid agent curcumin, isolated from turmeric, the microtubule stabilizer paclitaxel, from the Pacific Yew Taxus brevifolia, and the Streptomyces-derived Hsp90 inhibitor, geldanamycin. The overlooked approach of clearing tau aggregation will most likely be the next objective for AD drug discovery.
Collapse
|
294
|
Jung M, Metzger D. Methylene blue protects mitochondrial respiration from ethanol withdrawal stress. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/abb.2013.47a2004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
295
|
Maity S, Kumar R, Maity SK, Jana P, Bera S, Haldar D. Synthesis and study of 2-acetyl amino-3-[4-(2-amino-5-sulfo-phenylazo)-phenyl]-propionic acid: a new class of inhibitor for hen egg white lysozyme amyloidogenesis. MEDCHEMCOMM 2013. [DOI: 10.1039/c2md20236k] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
296
|
Binding of methylene blue to a surface cleft inhibits the oligomerization and fibrillization of prion protein. Biochim Biophys Acta Mol Basis Dis 2013; 1832:20-8. [DOI: 10.1016/j.bbadis.2012.09.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2012] [Revised: 09/14/2012] [Accepted: 09/17/2012] [Indexed: 01/08/2023]
|
297
|
Hosokawa M, Arai T, Masuda-Suzukake M, Nonaka T, Yamashita M, Akiyama H, Hasegawa M. Methylene blue reduced abnormal tau accumulation in P301L tau transgenic mice. PLoS One 2012; 7:e52389. [PMID: 23285020 PMCID: PMC3527507 DOI: 10.1371/journal.pone.0052389] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 11/14/2012] [Indexed: 11/24/2022] Open
Abstract
In neurodegenerative disorders, abnormally hyperphosphorylated and aggregated tau accumulates intracellularly, a mechanism which is thought to induce neuronal cell death. Methylene blue, a type of phenothiazine, has been reported to inhibit tau aggregation in vitro. However, the effect of methylene blue in vivo has remained unknown. Therefore, we examined whether methylene blue suppresses abnormal tau accumulation using P301L tau transgenic mice. At 8 to 11 months of age, these mice were orally administered methylene blue for 5 months. Subsequent results of Western blotting analysis revealed that this agent reduced detergent-insoluble phospho-tau. Methylene blue may have potential as a drug candidate for the treatment of tauopathy.
Collapse
Affiliation(s)
- Masato Hosokawa
- Department of Dementia and Higher Brain Function, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
298
|
Wasik U, Schneider G, Mietelska-Porowska A, Mazurkiewicz M, Fabczak H, Weis S, Zabke C, Harrington CR, Filipek A, Niewiadomska G. Calcyclin binding protein and Siah-1 interacting protein in Alzheimer's disease pathology: neuronal localization and possible function. Neurobiol Aging 2012; 34:1380-8. [PMID: 23260124 DOI: 10.1016/j.neurobiolaging.2012.11.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2012] [Revised: 10/18/2012] [Accepted: 11/16/2012] [Indexed: 01/13/2023]
Abstract
The calcyclin binding protein and Siah-1 interacting protein (CacyBP/SIP) protein was shown to play a role in the organization of microtubules. In this work we have examined the neuronal distribution and possible function of CacyBP/SIP in cytoskeletal pathophysiology. We have used brain tissue from Alzheimer's disease (AD) patients and from transgenic mice modeling 2 different pathologies characteristic for AD: amyloid and tau. In the brain from AD patients, CacyBP/SIP was found to be almost exclusively present in neuronal somata, and in control patients it was seen in the somata and neuronal processes. In mice doubly transgenic for amyloid precursor protein and presenilin 1 there was no difference in CacyBP/SIP neuronal localization in comparison with the nontransgenic animals. By contrast in tau transgenic mice, localization of CacyBP/SIP was similar to that observed for AD patients. To find the relation between CacyBP/SIP and tau we examined dephosphorylation of tau by CacyBP/SIP. We found that indeed it exhibited phosphatase activity toward tau. Altogether, our results suggest that CacyBP/SIP might play a role in AD pathology.
Collapse
Affiliation(s)
- Urszula Wasik
- Nencki Institute of Experimental Biology, Warsaw, Poland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
299
|
Abstract
Tau aggregates are present in several neurodegenerative diseases and correlate with the severity of memory deficit in AD (Alzheimer's disease). However, the triggers of tau aggregation and tau-induced neurodegeneration are still elusive. The impairment of protein-degradation systems might play a role in such processes, as these pathways normally keep tau levels at a low level which may prevent aggregation. Some proteases can process tau and thus contribute to tau aggregation by generating amyloidogenic fragments, but the complete clearance of tau mainly relies on the UPS (ubiquitin-proteasome system) and the ALS (autophagy-lysosome system). In the present paper, we focus on the regulation of the degradation of tau by the UPS and ALS and its relation to tau aggregation. We anticipate that stimulation of these two protein-degradation systems might be a potential therapeutic strategy for AD and other tauopathies.
Collapse
|
300
|
Monti MC, Margarucci L, Riccio R, Casapullo A. Modulation of tau protein fibrillization by oleocanthal. JOURNAL OF NATURAL PRODUCTS 2012; 75:1584-1588. [PMID: 22988908 DOI: 10.1021/np300384h] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Among the phenolic compounds extracted from extra virgin olive oil, oleocanthal (1) has attracted considerable attention in the modulation of many human diseases, such as inflammation and Alzheimer's disease (AD). Indeed, 1 is capable of altering the fibrillization of tau protein, which is one of the key factors at the basis of neurodegenerative diseases, and of covalently reacting with lysine ε-amino groups of the tau fragment K18 in an unspecific fashion. In the present study, an investigation of the recognition process and the reaction profile between 1 and the wild-type tau protein has been conducted by a circular dichroism, surface plasmon resonance, fluorescence, and mass spectrometry combined approach. As a result, 1 has been found to interact with tau-441, inducing stable conformational modifications of the protein secondary structure and also interfering with tau aggregation. These findings provide experimental support for the potential reduced risk of AD and related neurodegenerative diseases associated with olive oil consumption and may offer a new chemical scaffold for the development of AD-modulating agents.
Collapse
Affiliation(s)
- Maria Chiara Monti
- Dipartimento di Scienze Farmaceutiche e Biomediche, Università degli Studi di Salerno, Via Ponte don Melillo, 84084 Fisciano, Italy
| | | | | | | |
Collapse
|