101
|
Selkoe DJ. The therapeutics of Alzheimer's disease: where we stand and where we are heading. Ann Neurol 2015; 74:328-36. [PMID: 25813842 DOI: 10.1002/ana.24001] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2013] [Revised: 08/06/2013] [Accepted: 08/06/2013] [Indexed: 12/11/2022]
Abstract
Few diagnoses in modern medicine evoke more apprehension in patients and their families than Alzheimer disease (AD). Defined as a clinical and pathological entity a century ago, the disorder only came under intense molecular scrutiny in the mid-1980s. Genetic, histopathological, biochemical, and animal modeling studies have combined to provide evidence that the disease may begin with an imbalance between the production and clearance of the self-aggregating amyloid β protein (Aβ) in brain regions serving memory and cognition. This concept has been furthered by recent analyses in humans of cerebrospinal fluid and neuroimaging biomarkers that suggest an approximate sequence of AD-type brain alterations beginning >2 decades before the onset of dementia. Although the Aβ hypothesis of Alzheimer causation does not explain all features of this multifactorial syndrome, experimental agents that lower or neutralize Aβ have become the major focus of therapeutic research. Several clinical trials in mild-to-moderate AD have not met standard cognitive and functional endpoints, but there were important shortcomings in the agent and/or the trial design in each case. Based on the lessons learned, the field has moved on to test potentially disease-modifying agents in mild AD patients or via secondary prevention in presymptomatic subjects bearing amyloid plaques. Immunotherapeutic agents are receiving the most study, but other antiamyloid strategies and, importantly, nonamyloid targets such as tau and neuroinflammation are of great interest. The pace of recent developments augurs well for 1 or more experimental agents being shown to slow cognitive decline without major side effects. However, research funding from all sources will need to increase dramatically and soon to stave off the approaching tsunami of AD.
Collapse
Affiliation(s)
- Dennis J Selkoe
- Center for Neurologic Diseases, Department of Neurology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
102
|
The role of dietary coconut for the prevention and treatment of Alzheimer's disease: potential mechanisms of action. Br J Nutr 2015; 114:1-14. [PMID: 25997382 DOI: 10.1017/s0007114515001452] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Coconut, Cocos nucifera L., is a tree that is cultivated to provide a large number of products, although it is mainly grown for its nutritional and medicinal values. Coconut oil, derived from the coconut fruit, has been recognised historically as containing high levels of saturated fat; however, closer scrutiny suggests that coconut should be regarded more favourably. Unlike most other dietary fats that are high in long-chain fatty acids, coconut oil comprises medium-chain fatty acids (MCFA). MCFA are unique in that they are easily absorbed and metabolised by the liver, and can be converted to ketones. Ketone bodies are an important alternative energy source in the brain, and may be beneficial to people developing or already with memory impairment, as in Alzheimer's disease (AD). Coconut is classified as a highly nutritious 'functional food'. It is rich in dietary fibre, vitamins and minerals; however, notably, evidence is mounting to support the concept that coconut may be beneficial in the treatment of obesity, dyslipidaemia, elevated LDL, insulin resistance and hypertension - these are the risk factors for CVD and type 2 diabetes, and also for AD. In addition, phenolic compounds and hormones (cytokinins) found in coconut may assist in preventing the aggregation of amyloid-β peptide, potentially inhibiting a key step in the pathogenesis of AD. The purpose of the present review was to explore the literature related to coconut, outlining the known mechanistic physiology, and to discuss the potential role of coconut supplementation as a therapeutic option in the prevention and management of AD.
Collapse
|
103
|
Tanaka K, Takanaka S, Yoshida KI. A second-generation Bacillus cell factory for rare inositol production. Bioengineered 2015; 5:331-4. [PMID: 25482235 DOI: 10.4161/bioe.29897] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Some rare inositol stereoisomers are known to exert specific health-promoting effects, including scyllo-inositol (SI), which is a promising therapeutic agent for Alzheimer disease. We recently reported a Bacillus subtilis cell factory that performed the efficient production of SI from the cheapest and most abundant isomer myo-inositol (MI). In the cell factory all "useless" genes involved in MI and SI metabolism were deleted and overexpression of the key enzymes, IolG and IolW, was appended. It converted 10 g/L MI into the same amount of SI in 48 h of cultivation. In this addendum, we discuss further improvement in the cell factory and its possible applications.
Collapse
Affiliation(s)
- Kosei Tanaka
- a Organization of Advanced Science and Technology; Kobe University; Kobe, Japan
| | | | | |
Collapse
|
104
|
Comparation of activity against Aβ aggregation between RR and LPFFD. CHINESE JOURNAL OF POLYMER SCIENCE 2015. [DOI: 10.1007/s10118-015-1646-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
105
|
Morrone CD, Liu M, Black SE, McLaurin J. Interaction between therapeutic interventions for Alzheimer's disease and physiological Aβ clearance mechanisms. Front Aging Neurosci 2015; 7:64. [PMID: 25999850 PMCID: PMC4419721 DOI: 10.3389/fnagi.2015.00064] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2015] [Accepted: 04/13/2015] [Indexed: 01/05/2023] Open
Abstract
Most therapeutic agents are designed to target a molecule or pathway without consideration of the mechanisms involved in the physiological turnover or removal of that target. In light of this and in particular for Alzheimer’s disease, a number of therapeutic interventions are presently being developed/investigated which target the amyloid-β peptide (Aβ). However, the literature has not adequately considered which Aβ physiological clearance pathways are necessary and sufficient for the effective action of these therapeutics. In this review, we evaluate the therapeutic strategies targeting Aβ presently in clinical development, discuss the possible interaction of these treatments with pathways that under normal physiological conditions are responsible for the turnover of Aβ and highlight possible caveats. We consider immunization strategies primarily reliant on a peripheral sink mechanism of action, small molecules that are reliant on entry into the CNS and thus degradation pathways within the brain, as well as lifestyle interventions that affect vascular, parenchymal and peripheral degradation pathways. We propose that effective development of Alzheimer’s disease therapeutic strategies targeting Aβ peptide will require consideration of the age- and disease-specific changes to endogenous Aβ clearance mechanisms in order to elicit maximal efficacy.
Collapse
Affiliation(s)
- Christopher D Morrone
- Biological Sciences, Sunnybrook Research Institute Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada
| | - Mingzhe Liu
- Biological Sciences, Sunnybrook Research Institute Toronto, ON, Canada
| | - Sandra E Black
- Canadian Partnership for Stroke Recovery, Sunnybrook Research Institute Toronto, ON, Canada ; Department of Medicine (Neurology), University of Toronto Toronto, ON, Canada ; Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre and University of Toronto Toronto, ON, Canada
| | - JoAnne McLaurin
- Biological Sciences, Sunnybrook Research Institute Toronto, ON, Canada ; Department of Laboratory Medicine and Pathobiology, University of Toronto Toronto, ON, Canada
| |
Collapse
|
106
|
MRZ-99030 – A novel modulator of Aβ aggregation: II – Reversal of Aβ oligomer-induced deficits in long-term potentiation (LTP) and cognitive performance in rats and mice. Neuropharmacology 2015; 92:170-82. [DOI: 10.1016/j.neuropharm.2014.12.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Revised: 11/28/2014] [Accepted: 12/02/2014] [Indexed: 11/21/2022]
|
107
|
Dias C, Rauter AP. Carbohydrates and Glycomimetics in Alzheimer's Disease Therapeutics and Diagnosis. CARBOHYDRATES IN DRUG DESIGN AND DISCOVERY 2015. [DOI: 10.1039/9781849739993-00180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Alzheimer's disease is the most prevalent form of late-life dementia, affecting millions worldwide. The devastating nature of the disease, unsuccessful treatment options and high socio-economic impact has inspired scientists to develop new structures with neuroprotective properties. Although currently available drugs target cholinergic neurotransmission, investigation towards disease-modifying therapies has been growing and carbohydrates have been playing an active role in the latest discoveries. Sugars, as polyfunctional compounds particularly important in biology and widely involved in human health and disease, have great potential to generate bioactive and bioavailable interesting molecules. Herein we discuss the importance of carbohydrates and glycomimetic structures, addressing different aspects of neuroprotection under investigation, targeting amyloid, tau and cholinergic hypotheses. The potential of carbohydrates in diagnosis is also discussed.
Collapse
Affiliation(s)
- Catarina Dias
- Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016 Lisbon Portugal
| | - Amélia P. Rauter
- Centro de Química e Bioquímica, Faculdade de Ciências, Universidade de Lisboa, Campo Grande 1749-016 Lisbon Portugal
| |
Collapse
|
108
|
Schneider S. Inositol transport proteins. FEBS Lett 2015; 589:1049-58. [DOI: 10.1016/j.febslet.2015.03.012] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/05/2015] [Accepted: 03/18/2015] [Indexed: 12/27/2022]
|
109
|
Zheng X, Liu D, Klärner FG, Schrader T, Bitan G, Bowers MT. Amyloid β-protein assembly: The effect of molecular tweezers CLR01 and CLR03. J Phys Chem B 2015; 119:4831-41. [PMID: 25751170 PMCID: PMC4415044 DOI: 10.1021/acs.jpcb.5b00692] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
![]()
The early oligomerization of amyloid
β-protein (Aβ)
has been shown to be an important event in the pathology of Alzheimer’s
disease (AD). Designing small molecule inhibitors targeting Aβ
oligomerization is one attractive and promising strategy for AD treatment.
Here we used ion mobility spectrometry coupled to mass spectrometry
(IMS-MS) to study the different effects of the molecular tweezers
CLR01 and CLR03 on Aβ self-assembly. CLR01 was found to bind
to Aβ directly and disrupt its early oligomerization. Moreover,
CLR01 remodeled the early oligomerization of Aβ42 by compacting
the structures of dimers and tetramers and as a consequence eliminated
higher-order oligomers. Unexpectedly, the negative-control derivative,
CLR03, which lacks the hydrophobic arms of the tweezer structure,
was found to facilitate early Aβ oligomerization. Our study
provides an example of IMS as a powerful tool to study and better
understand the interaction between small molecule modulators and Aβ
oligomerization, which is not attainable by other methods, and provides
important insights into therapeutic development of molecular tweezers
for AD treatment.
Collapse
Affiliation(s)
- Xueyun Zheng
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Deyu Liu
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| | - Frank-Gerrit Klärner
- ‡Institute of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Thomas Schrader
- ‡Institute of Organic Chemistry, University of Duisburg-Essen, Essen 45117, Germany
| | - Gal Bitan
- §Department of Neurology, David Geffen School of Medicine, Brain Research Institute, and Molecular Biology Institute, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Michael T Bowers
- †Department of Chemistry and Biochemistry, University of California, Santa Barbara, California 93106, United States
| |
Collapse
|
110
|
Bohm C, Chen F, Sevalle J, Qamar S, Dodd R, Li Y, Schmitt-Ulms G, Fraser PE, St George-Hyslop PH. Current and future implications of basic and translational research on amyloid-β peptide production and removal pathways. Mol Cell Neurosci 2015; 66:3-11. [PMID: 25748120 PMCID: PMC4503820 DOI: 10.1016/j.mcn.2015.02.016] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2015] [Revised: 02/26/2015] [Accepted: 02/27/2015] [Indexed: 01/12/2023] Open
Abstract
Inherited variants in multiple different genes are associated with increased risk for Alzheimer's disease (AD). In many of these genes, the inherited variants alter some aspect of the production or clearance of the neurotoxic amyloid β-peptide (Aβ). Thus missense, splice site or duplication mutants in the presenilin 1 (PS1), presenilin 2 (PS2) or the amyloid precursor protein (APP) genes, which alter the levels or shift the balance of Aβ produced, are associated with rare, highly penetrant autosomal dominant forms of Familial Alzheimer's Disease (FAD). Similarly, the more prevalent late-onset forms of AD are associated with both coding and non-coding variants in genes such as SORL1, PICALM and ABCA7 that affect the production and clearance of Aβ. This review summarises some of the recent molecular and structural work on the role of these genes and the proteins coded by them in the biology of Aβ. We also briefly outline how the emerging knowledge about the pathways involved in Aβ generation and clearance can be potentially targeted therapeutically. This article is part of Special Issue entitled "Neuronal Protein".
Collapse
Affiliation(s)
- C Bohm
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - F Chen
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - J Sevalle
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - S Qamar
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - R Dodd
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - Y Li
- Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK
| | - G Schmitt-Ulms
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada
| | - P H St George-Hyslop
- Tanz Centre for Research in Neurodegenerative Diseases, Departments of Medicine, Laboratory Medicine and Pathobiology and Medical Biophysics, University of Toronto, Krembil Discovery Tower, 6th Floor-6KD417, 60 Leonard Avenue, Toronto, Ontario M5T 2S8, Canada; Cambridge Institute for Medical Research, Wellcome Trust MRC Building, Addenbrookes Hospital, Hills Road, Cambridge CB2 0XY, UK.
| |
Collapse
|
111
|
Determining binding sites of polycyclic aromatic small molecule-based amyloid-beta peptide aggregation modulators using sequence-specific antibodies. Anal Biochem 2015; 470:61-70. [DOI: 10.1016/j.ab.2014.10.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Revised: 10/21/2014] [Accepted: 10/27/2014] [Indexed: 12/22/2022]
|
112
|
Parrott MD, Winocur G, Bazinet RP, Ma DW, Greenwood CE. Whole-food diet worsened cognitive dysfunction in an Alzheimer's disease mouse model. Neurobiol Aging 2015; 36:90-9. [DOI: 10.1016/j.neurobiolaging.2014.08.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 08/07/2014] [Accepted: 08/12/2014] [Indexed: 12/13/2022]
|
113
|
Kang JH, Ryoo NY, Shin DW, Trojanowski JQ, Shaw LM. Role of cerebrospinal fluid biomarkers in clinical trials for Alzheimer's disease modifying therapies. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2014; 18:447-56. [PMID: 25598657 PMCID: PMC4296032 DOI: 10.4196/kjpp.2014.18.6.447] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/19/2014] [Revised: 09/02/2014] [Accepted: 10/07/2014] [Indexed: 12/27/2022]
Abstract
Until now, a disease-modifying therapy (DMT) that has an ability to slow or arrest Alzheimer's disease (AD) progression has not been developed, and all clinical trials involving AD patients enrolled by clinical assessment alone also have not been successful. Given the growing consensus that the DMT is likely to require treatment initiation well before full-blown dementia emerges, the early detection of AD will provide opportunities to successfully identify new drugs that slow the course of AD pathology. Recent advances in early detection of AD and prediction of progression of the disease using various biomarkers, including cerebrospinal fluid (CSF) Aβ1-42, total tau and p-tau181 levels, and imagining biomarkers, are now being actively integrated into the designs of AD clinical trials. In terms of therapeutic mechanisms, monitoring these markers may be helpful for go/no-go decision making as well as surrogate markers for disease severity or progression. Furthermore, CSF biomarkers can be used as a tool to enrich patients for clinical trials with prospect of increasing statistical power and reducing costs in drug development. However, the standardization of technical aspects of analysis of these biomarkers is an essential prerequisite to the clinical uses. To accomplish this, global efforts are underway to standardize CSF biomarker measurements and a quality control program supported by the Alzheimer's Association. The current review summarizes therapeutic targets of developing drugs in AD pathophysiology, and provides the most recent advances in the
Collapse
Affiliation(s)
- Ju-Hee Kang
- Department of Pharmacology and Clinical Pharmacology, Inha University School of Medicine, Incheon 400-712, Korea. ; Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon 400-712, Korea. ; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Na-Young Ryoo
- Hypoxia-related Disease Research Center, Inha University School of Medicine, Incheon 400-712, Korea. ; Department of Anatomy, Inha University School of Medicine, Incheon 400-712, Korea
| | - Dong Wun Shin
- Department of Emergency Medicine, Inje University Ilsan Paik Hospital, Ilsan 411-706, Korea
| | - John Q Trojanowski
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. ; Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Leslie M Shaw
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA. ; Institute on Aging and Center for Neurodegenerative Disease Research, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, 19104, USA
| |
Collapse
|
114
|
Kang B, Sutou T, Wang Y, Kuwano S, Yamaoka Y, Takasu K, Yamada KI. N-Heterocyclic Carbene-Catalyzed Benzoin Strategy for Divergent Synthesis of Cyclitol Derivatives from Alditols. Adv Synth Catal 2014. [DOI: 10.1002/adsc.201400712] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
115
|
Wang H, Raleigh DP. General amyloid inhibitors? A critical examination of the inhibition of IAPP amyloid formation by inositol stereoisomers. PLoS One 2014; 9:e104023. [PMID: 25260075 PMCID: PMC4178012 DOI: 10.1371/journal.pone.0104023] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Accepted: 07/09/2014] [Indexed: 01/01/2023] Open
Abstract
Islet amyloid polypeptide (IAPP or amylin) forms amyloid deposits in the islets of Langerhans; a process that is believed to contribute to the progression of type 2 diabetes and to the failure of islet transplants. An emerging theme in amyloid research is the hypothesis that the toxic species produced during amyloid formation by different polypeptides share common features and exert their effects by common mechanisms. If correct, this suggests that inhibitors of amyloid formation by one polypeptide might be effective against other amyloidogenic sequences. IAPP and Aβ, the peptide responsible for amyloid formation in Alzheimer's disease, are particularly interesting in this regard as they are both natively unfolded in their monomeric states and share some common characteristics. Comparatively little effort has been expended on the design of IAPP amyloid inhibitors, thus it is natural to inquire if Aβ inhibitors are effective against IAPP, especially since no IAPP inhibitors have been clinically approved. A range of compounds inhibit Aβ amyloid formation, including various stereoisomers of inositol. Myo-, scyllo-, and epi-inositol have been shown to induce conformational changes in Aβ and prevent Aβ amyloid fibril formation by stabilizing non-fibrillar β-sheet structures. We investigate the ability of inositol stereoisomers to inhibit amyloid formation by IAPP. The compounds do not induce a conformational change in IAPP and are ineffective inhibitors of IAPP amyloid formation, although some do lead to modest apparent changes in IAPP amyloid fibril morphology. Thus not all classes of Aβ inhibitors are effective against IAPP. This work provides a basis of comparison to work on polyphenol based inhibitors of IAPP amyloid formation and helps provide clues as to the features which render them effective. The study also helps provide information for further efforts in rational inhibitor design.
Collapse
Affiliation(s)
- Hui Wang
- Department of Chemistry, Stony Brook University, Stony Brook, New York, United States of America
| | - Daniel P. Raleigh
- Department of Chemistry, Stony Brook University, Stony Brook, New York, United States of America
- Graduate Program in Biochemistry and Structural Biology, Graduate Program in Biophysics, Stony Brook University, Stony Brook, New York, United States of America
- * E-mail:
| |
Collapse
|
116
|
Lu Z, Wang J, Li M, Liu Q, Wei D, Yang M, Kong L. (1)H NMR-based metabolomics study on a goldfish model of Parkinson's disease induced by 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). Chem Biol Interact 2014; 223:18-26. [PMID: 25242684 DOI: 10.1016/j.cbi.2014.09.006] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 08/22/2014] [Accepted: 09/09/2014] [Indexed: 11/19/2022]
Abstract
A goldfish (Carassius auratus) model of Parkinson's disease (PD) was constructed by a single dose of 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) according to previously reported methods. Global metabolite changes in brain of the MPTP induced goldfish model of PD were investigated. (1)H NMR-based metabolomics combined with various statistical methods such as orthogonal partial least squares discriminant analysis (OPLS-DA) and two-dimensional statistical total correlation spectroscopy (2D-STOCSY) found significant increase of leucine, isoleucine, valine, alanine, alanylalanine, creatinine, myo-inositol, 18:2 fatty acid, total fatty acids, arachic alcohol, taurine and significant decrease of N-acetylaspartate, (phospho)creatine, (phospho)choline, betaine, glutamine, 3-hexenedioate, acetamide, malonate, isocitrate, scyllo-inositol, phosphatidylcholines, cholesterols, n-3 fatty acids, polyunsaturated fatty acids (PUFAs) in brain of MPTP induced PD goldfish. These disturbed metabolite levels were involved in oxidative stress, energy failure, neuronal cell injury and death, consistent with those observed in clinical PD patients, and rodents and primates model of PD, indicating that the acute MPTP model of goldfish was an ideal and valuable model for PD research. In addition, several unusual metabolites in brain were significantly changed between MPTP induced PD and control goldfish, which might also play an important role in the pathogenesis of PD. This study also demonstrated the applicability and potential of (1)H NMR-based metabolomics approach for evaluation of animal models of disease induced by chemicals, such as MPTP-induced PD goldfish.
Collapse
Affiliation(s)
- Zhaoguang Lu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Junsong Wang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, 200 Xiao Ling Wei Street, Nanjing 210094, PR China.
| | - Minghui Li
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Qingwang Liu
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Dandan Wei
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Minghua Yang
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China
| | - Lingyi Kong
- State Key Laboratory of Natural Medicines, Department of Natural Medicinal Chemistry, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, PR China.
| |
Collapse
|
117
|
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: 42] [Impact Index Per Article: 3.8] [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
|
118
|
α-Melanocyte stimulating hormone prevents GABAergic neuronal loss and improves cognitive function in Alzheimer's disease. J Neurosci 2014; 34:6736-45. [PMID: 24828629 DOI: 10.1523/jneurosci.5075-13.2014] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
In Alzheimer's disease (AD), appropriate excitatory-inhibitory balance required for memory formation is impaired. Our objective was to elucidate deficits in the inhibitory GABAergic system in the TgCRND8 mouse model of AD to establish a link between GABAergic dysfunction and cognitive function. We sought to determine whether the neuroprotective peptide α-melanocyte stimulating hormone (α-MSH) attenuates GABAergic loss and thus improves cognition. TgCRND8 mice with established β-amyloid peptide pathology and nontransgenic littermates were treated with either α-MSH or vehicle via daily intraperitoneal injections for 28 d. TgCRND8 mice exhibited spatial memory deficits and altered anxiety that were rescued after α-MSH treatment. The expression of GABAergic marker glutamic acid decarboxylase 67 (GAD67) and the number of GABAergic GAD67+ interneurons expressing neuropeptide Y and somatostatin are reduced in the hippocampus in vehicle-treated TgCRND8 mice. In the septohippocampal pathway, GABAergic deficits are observed before cholinergic deficits, suggesting that GABAergic loss may underlie behavior deficits in vehicle-treated TgCRND8 mice. α-MSH preserves GAD67 expression and prevents loss of the somatostatin-expressing subtype of GABAergic GAD67+ inhibitory interneurons. Without decreasing β-amyloid peptide load in the brain, α-MSH improves spatial memory in TgCRND8 mice and prevents alterations in anxiety. α-MSH modulated the excitatory-inhibitory balance in the brain by restoring GABAergic inhibition and, as a result, improved cognition in TgCRND8 mice.
Collapse
|
119
|
Nair S, Traini M, Dawes IW, Perrone GG. Genome-wide analysis of Saccharomyces cerevisiae identifies cellular processes affecting intracellular aggregation of Alzheimer's amyloid-β42: importance of lipid homeostasis. Mol Biol Cell 2014; 25:2235-49. [PMID: 24870034 PMCID: PMC4116298 DOI: 10.1091/mbc.e13-04-0216] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Amyloid-β (Aβ)-containing plaques are a major neuropathological feature of Alzheimer's disease (AD). The two major isoforms of Aβ peptide associated with AD are Aβ40 and Aβ42, of which the latter is highly prone to aggregation. Increased presence and aggregation of intracellular Aβ42 peptides is an early event in AD progression. Improved understanding of cellular processes affecting Aβ42 aggregation may have implications for development of therapeutic strategies. Aβ42 fused to green fluorescent protein (Aβ42-GFP) was expressed in ∼4600 mutants of a Saccharomyces cerevisiae genome-wide deletion library to identify proteins and cellular processes affecting intracellular Aβ42 aggregation by assessing the fluorescence of Aβ42-GFP. This screening identified 110 mutants exhibiting intense Aβ42-GFP-associated fluorescence. Four major cellular processes were overrepresented in the data set, including phospholipid homeostasis. Disruption of phosphatidylcholine, phosphatidylserine, and/or phosphatidylethanolamine metabolism had a major effect on intracellular Aβ42 aggregation and localization. Confocal microscopy indicated that Aβ42-GFP localization in the phospholipid mutants was juxtaposed to the nucleus, most likely associated with the endoplasmic reticulum (ER)/ER membrane. These data provide a genome-wide indication of cellular processes that affect intracellular Aβ42-GFP aggregation and may have important implications for understanding cellular mechanisms affecting intracellular Aβ42 aggregation and AD disease progression.
Collapse
Affiliation(s)
- S Nair
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, Australia
| | - M Traini
- Atherosclerosis Laboratory, ANZAC Research Institute, Concord Hospital, Concord, NSW 2139, Australia
| | - I W Dawes
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW 2052, AustraliaRamaciotti Centre for Gene Function Analysis, University of New South Wales, Sydney, NSW 2052, Australia
| | - G G Perrone
- School of Science and Health, University of Western Sydney, Penrith, NSW 1797, Australia
| |
Collapse
|
120
|
Rana P, Gupta M, Khan AR, Hemanth Kumar BS, Roy R, Khushu S. NMR based metabolomics reveals acute hippocampal metabolic fluctuations during cranial irradiation in murine model. Neurochem Int 2014; 74:1-7. [PMID: 24787771 DOI: 10.1016/j.neuint.2014.04.013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 04/15/2014] [Accepted: 04/21/2014] [Indexed: 01/28/2023]
Abstract
Cranial irradiation is widely used as a treatment modality or prophylactic treatment in cancer patients, but it is frequently related to neurocognitive impairment in cancer survivors. Though most of radiation-induced changes occur during early and late delayed phase of radiation sickness, recent reports have supported the evidence of impaired neurogenesis within 24-48 h of radiation exposure that may implicate changes in acute phase as well. Inspection of these acute changes could be considered important as they may have long lasting effect on cognitive development and functions. In the present study, (1)H NMR spectroscopy based metabolomic approach was used to obtain comprehensive information of hippocampus metabolic physiology during acute phase of radiation sickness in a mouse model for single dose 8 Gy cranial irradiation. The analysis demonstrated reduced metabolic activity in irradiated animals compared to controls, typically evident in citric acid cycle intermediates, glutamine/glutamate and ketone bodies metabolism thus providing strong indication that the hippocampus is metabolically responsive to radiation exposure. The data suggested reduced glucose utilization, altered intermediary and neurotransmitter metabolism in hippocampus tissue extract. To the best of our knowledge this is the first metabolomic study to document cranial irradiation induced acute metabolic changes using in vitro(1)H NMR spectroscopy.
Collapse
Affiliation(s)
- Poonam Rana
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Mamta Gupta
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Ahmad Raza Khan
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - B S Hemanth Kumar
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India
| | - Raja Roy
- Centre for Biomedical Magnetic Resonance (CBMR), SGPGIMS Campus, Lucknow, Uttar Pradesh, India
| | - Subash Khushu
- NMR Research Centre, Institute of Nuclear Medicine and Allied Sciences, Delhi, India.
| |
Collapse
|
121
|
Lee SH, Kim Y, Kim HY, Kim YH, Kim MS, Kong JY, Lee MH, Kim DJ, Ahn YG. Aminostyrylbenzofuran directly reduces oligomeric amyloid-β and reverses cognitive deficits in Alzheimer transgenic mice. PLoS One 2014; 9:e95733. [PMID: 24760018 PMCID: PMC3997483 DOI: 10.1371/journal.pone.0095733] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2013] [Accepted: 03/29/2014] [Indexed: 11/18/2022] Open
Abstract
Alzheimer's disease is an irreversible neurodegenerative disorder that is characterized by the abnormal aggregation of amyloid-β into neurotoxic oligomers and plaques. Although many disease-modifying molecules are currently in Alzheimer clinical trials, a small molecule that inhibits amyloid-β aggregation and ameliorates the disorder has not been approved to date. Herein, we report the effects of a potent small molecule, 6-methoxy-2-(4-dimethylaminostyryl) benzofuran (KMS88009), that directly disrupts amyloid-β oligomerization, preserving cognitive behavior when used prophylactically and reversing declines in cognitive behavior when used therapeutically. KMS88009 exhibited excellent pharmacokinetic profiles with extensive brain uptake and a high level of safety. When orally administered before and after the onset of Alzheimer's disease symptoms, KMS88009 significantly reduced assembly of amyloid-β oligomers and improved cognitive behaviors in the APP/PS1 double transgenic mouse model. The unique dual mode of action indicates that KMS88009 may be a powerful therapeutic candidate for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Sang-Hyun Lee
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong-si, Gyonggi-do, Republic of Korea
- Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - YoungSoo Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Biological Chemistry Program, University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hye Yun Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- Biological Chemistry Program, University of Science and Technology (UST), Daejeon, Republic of Korea
- Department of Biochemistry and Biomedical Sciences, Seoul National University, College of Medicine, Seoul, Republic of Korea
| | - Young Hoon Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong-si, Gyonggi-do, Republic of Korea
| | - Maeng Sup Kim
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong-si, Gyonggi-do, Republic of Korea
| | - Jae Yang Kong
- College of Pharmacy, Keimyung University, Daegu, Republic of Korea
| | - Mun-Han Lee
- Department of Biochemistry, College of Veterinary Medicine, Seoul National University, Seoul, Republic of Korea
| | - Dong Jin Kim
- Center for Neuro-Medicine, Brain Science Institute, Korea Institute of Science and Technology, Seoul, Republic of Korea
- * E-mail: (DJK); (YGA)
| | - Young Gil Ahn
- Hanmi Research Center, Hanmi Pharmaceutical Co., Ltd., Hwaseong-si, Gyonggi-do, Republic of Korea
- * E-mail: (DJK); (YGA)
| |
Collapse
|
122
|
Abstract
A hallmark of Alzheimer's disease (AD) brain is the amyloid β (Aβ) plaque, which is comprised of Aβ peptides. Multiple lines of evidence suggest that Aβ oligomers are more toxic than other peptide forms. We sought to develop a robust assay to quantify oligomers from CSF. Antibody 19.3 was compared in one-site and competitive ELISAs for oligomer binding specificity. A two-site ELISA for oligomers was developed using 19.3 coupled to a sensitive, bead-based fluorescent platform able to detect single photons of emitted light. The two-site ELISA was >2500× selective for Aβ oligomers over Aβ monomers with a limit of detection ∼ 0.09 pg/ml in human CSF. The lower limit of reliable quantification of the assay was 0.18 pg/ml and the antibody pairs recognized Aβ multimers comprised of either synthetic standards, or endogenous oligomers isolated from confirmed human AD and healthy control brain. Using the assay, a significant 3- to 5-fold increase in Aβ oligomers in human AD CSF compared with comparably aged controls was demonstrated. The increase was seen in three separate human cohorts, totaling 63 AD and 54 controls. CSF oligomers ranged between 0.1 and 10 pg/ml. Aβ oligomer levels did not strongly associate with age or gender, but had an inverse correlation with MMSE score. The C statistic for the Aβ oligomer ROC curve was 0.86, with 80% sensitivity and 88% specificity to detect AD, suggesting reasonable discriminatory power for the AD state and the potential for utility as a diagnostic marker.
Collapse
|
123
|
Klohs J, Rudin M, Shimshek DR, Beckmann N. Imaging of cerebrovascular pathology in animal models of Alzheimer's disease. Front Aging Neurosci 2014; 6:32. [PMID: 24659966 PMCID: PMC3952109 DOI: 10.3389/fnagi.2014.00032] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Accepted: 02/19/2014] [Indexed: 01/04/2023] Open
Abstract
In Alzheimer's disease (AD), vascular pathology may interact with neurodegeneration and thus aggravate cognitive decline. As the relationship between these two processes is poorly understood, research has been increasingly focused on understanding the link between cerebrovascular alterations and AD. This has at last been spurred by the engineering of transgenic animals, which display pathological features of AD and develop cerebral amyloid angiopathy to various degrees. Transgenic models are versatile for investigating the role of amyloid deposition and vascular dysfunction, and for evaluating novel therapeutic concepts. In addition, research has benefited from the development of novel imaging techniques, which are capable of characterizing vascular pathology in vivo. They provide vascular structural read-outs and have the ability to assess the functional consequences of vascular dysfunction as well as to visualize and monitor the molecular processes underlying these pathological alterations. This article focusses on recent in vivo small animal imaging studies addressing vascular aspects related to AD. With the technical advances of imaging modalities such as magnetic resonance, nuclear and microscopic imaging, molecular, functional and structural information related to vascular pathology can now be visualized in vivo in small rodents. Imaging vascular and parenchymal amyloid-β (Aβ) deposition as well as Aβ transport pathways have been shown to be useful to characterize their dynamics and to elucidate their role in the development of cerebral amyloid angiopathy and AD. Structural and functional imaging read-outs have been employed to describe the deleterious affects of Aβ on vessel morphology, hemodynamics and vascular integrity. More recent imaging studies have also addressed how inflammatory processes partake in the pathogenesis of the disease. Moreover, imaging can be pivotal in the search for novel therapies targeting the vasculature.
Collapse
Affiliation(s)
- Jan Klohs
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland
| | - Markus Rudin
- Institute for Biomedical Engineering, University of Zurich and ETH Zurich Zurich, Switzerland ; Neuroscience Center Zurich, University of Zurich and ETH Zurich Zurich, Switzerland ; Institute of Pharmacology and Toxicology, University of Zurich Zurich, Switzerland
| | - Derya R Shimshek
- Autoimmunity, Transplantation and Inflammation/Neuroinflammation Department, Novartis Institutes for BioMedical Research Basel, Switzerland
| | - Nicolau Beckmann
- Analytical Sciences and Imaging, Novartis Institutes for BioMedical Research Basel, Switzerland
| |
Collapse
|
124
|
Modulating nitric oxide signaling in the CNS for Alzheimer's disease therapy. Future Med Chem 2014; 5:1451-68. [PMID: 23919554 DOI: 10.4155/fmc.13.111] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Nitric oxide (NO)/solube GC (sGC)/cGMP signaling is important for modulating synaptic transmission and plasticity in the hippocampus and cerebral cortex, which are critical for learning and memory. Physiological concentrations of NO also elicit anti-apoptotic/prosurvival effects against various neurotoxic challenges and brain insults through multiple mechanisms. Depression of the NO/sGC pathway is a feature of Alzheimer's disease (AD), attributed to amyloid-β neuropathology, and altered expression and activity of NOS, sGC and PDE enzymes. Different classes of NO-releasing hybrid drugs, including nomethiazoles, NO-NSAIDs and NO-acetylcholinesterase inhibitors were designed to deliver low concentrations of exogenous NO to the CNS while targeting other underlying disease mechanisms, such as excitotoxicity, neuro-inflammation and acetylcholine deficiency, respectively. Incorporating a NO-donating moiety may also reduce gastrointestinal and liver toxicity of the parent drugs. Progress has also been made in targeting downstream sGC and PDE enzymes. The PDE9 inhibitor PF-04447943 has completed Phase II clinical trials for AD. The search for effective NO-donating hybrid drugs, CNS-targeting sGC stimulators/activators and selective PDE inhibitors is an important goal for pharmacotherapy that manipulates NO biochemical pathways involved in cognitive function and neuroprotection. Rigorous preclinical validation of target engagement, and optimization of pharmacokinetic and toxicity profiles are likely to advance more drug candidates into clinical trials for mild cognitive impairment and early stage AD.
Collapse
|
125
|
Liu J, Wang W, Zhang Q, Zhang S, Yuan Z. Study on the Efficiency and Interaction Mechanism of a Decapeptide Inhibitor of β-Amyloid Aggregation. Biomacromolecules 2014; 15:931-9. [DOI: 10.1021/bm401795e] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- Jing Liu
- Key Laboratory
of Functional
Polymer Materials of Ministry of Education, and Institute of Polymer
Chemistry, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Wei Wang
- Key Laboratory
of Functional
Polymer Materials of Ministry of Education, and Institute of Polymer
Chemistry, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Qian Zhang
- Key Laboratory
of Functional
Polymer Materials of Ministry of Education, and Institute of Polymer
Chemistry, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Saihui Zhang
- Key Laboratory
of Functional
Polymer Materials of Ministry of Education, and Institute of Polymer
Chemistry, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| | - Zhi Yuan
- Key Laboratory
of Functional
Polymer Materials of Ministry of Education, and Institute of Polymer
Chemistry, Nankai University, and Collaborative Innovation Center of Chemical Science and Engineering, Tianjin 300071, China
| |
Collapse
|
126
|
Kamp JA, Moursel LG, Haan J, Terwindt GM, Lesnik Oberstein SA, van Duinen SG, van Roon-Mom WM. Amyloid β in hereditary cerebral hemorrhage with amyloidosis-Dutch type. Rev Neurosci 2014; 25:641-51. [DOI: 10.1515/revneuro-2014-0008] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Accepted: 04/17/2014] [Indexed: 12/23/2022]
|
127
|
Thomason LAM, Stefanovic B, McLaurin J. Cerebrovascular contributions to Alzheimer's disease pathophysiology and potential therapeutic interventions in mouse models. Eur J Neurosci 2013; 37:1994-2004. [PMID: 23773069 DOI: 10.1111/ejn.12181] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 02/08/2013] [Accepted: 02/11/2013] [Indexed: 10/26/2022]
Abstract
The inter-relationship between vascular dysfunction and Alzheimer's disease pathology is not clearly understood; however, it is clear that the accumulation of amyloid-beta peptide and loss of vascular function contribute to the cognitive decline detected in patients. At present, imaging modalities can monitor the downstream effects of vascular dysfunction such as cerebral blood flow alterations, white and gray matter lacunes, and ischemic lesions; however, they cannot distinguish parenchymal plaques from cerebrovascular amyloid. Much of our understanding regarding the relationship between amyloid and vascular dysfunction has come from longitudinal population studies and mouse models. In this review, we will discuss the breadth of data generated on vascular function in mouse models of Alzheimer's disease and cerebrovascular amyloid angiopathy. We will also discuss therapeutic strategies targeting the reduction of cerebrovascular amyloid angiopathy and improvement of vascular function.
Collapse
Affiliation(s)
- Lynsie A M Thomason
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Circle, Toronto, ON M5S 1A8, Canada
| | | | | |
Collapse
|
128
|
Lai AY, Lan CP, Hasan S, Brown ME, McLaurin J. scyllo-Inositol promotes robust mutant Huntingtin protein degradation. J Biol Chem 2013; 289:3666-76. [PMID: 24352657 DOI: 10.1074/jbc.m113.501635] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Huntington disease is characterized by neuronal aggregates and inclusions containing polyglutamine-expanded huntingtin protein and peptide fragments (polyQ-Htt). We have used an established cell-based assay employing a PC12 cell line overexpressing truncated exon 1 of Htt with a 103-residue polyQ expansion that yields polyQ-Htt aggregates to investigate the fate of polyQ-Htt-drug complexes. scyllo-Inositol is an endogenous inositol stereoisomer known to inhibit accumulation and toxicity of the amyloid-β peptide and α-synuclein. In light of these properties, we investigated the effect of scyllo-inositol on polyQ-Htt accumulation. We show that scyllo-inositol lowered the number of visible polyQ-Htt aggregates and robustly decreased polyQ-Htt protein abundance without concomitant cellular toxicity. We found that scyllo-inositol-induced polyQ-Htt reduction was by rescue of degradation pathways mediated by the lysosome and by the proteasome but not autophagosomes. The rescue of degradation pathways was not a direct result of scyllo-inositol on the lysosome or proteasome but due to scyllo-inositol-induced reduction in mutant polyQ-Htt protein levels.
Collapse
Affiliation(s)
- Aaron Y Lai
- From the Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario M5S 1A8, Canada
| | | | | | | | | |
Collapse
|
129
|
Tanaka K, Tajima S, Takenaka S, Yoshida KI. An improved Bacillus subtilis cell factory for producing scyllo-inositol, a promising therapeutic agent for Alzheimer's disease. Microb Cell Fact 2013; 12:124. [PMID: 24325193 PMCID: PMC3878828 DOI: 10.1186/1475-2859-12-124] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2013] [Accepted: 12/03/2013] [Indexed: 02/03/2023] Open
Abstract
Background Bacillus subtilis 168 possesses an efficient pathway to metabolize some of the stereoisomers of inositol, including myo-inositol (MI) and scyllo-inositol (SI). Previously we reported a prototype of a B. subtilis cell factory with modified inositol metabolism that converts MI into SI in the culture medium. However, it wasted half of initial 1.0% (w/v) MI, and the conversion was limited to produce only 0.4% (w/v) SI. To achieve a more efficient SI production, we attempted additional modifications. Results All “useless” genes involved in MI and SI metabolism were deleted. Although no elevation in SI production was observed in the deletion strain, it did result in no wastage of MI anymore. Thus additionally, overexpression of the key enzymes, IolG and IolW, was appended to demonstrate that simultaneous overexpression of them enabled complete conversion of all MI into SI. Conclusions The B. subtilis cell factory was improved to yield an SI production rate of 10 g/L/48 h at least. The improved conversion was achieved only in the presence of enriched nutrition in the form of 2% (w/v) Bacto soytone in the medium, which may be due to the increasing demand for regeneration of cofactors.
Collapse
Affiliation(s)
| | | | | | - Ken-ichi Yoshida
- Organization of Advanced Science and Technology, Kobe University, 1-1 Rokkodai, Nada, Kobe 657 8501, Japan.
| |
Collapse
|
130
|
Holland JP, Liang SH, Rotstein BH, Collier TL, Stephenson NA, Greguric I, Vasdev N. Alternative approaches for PET radiotracer development in Alzheimer's disease: imaging beyond plaque. J Labelled Comp Radiopharm 2013; 57:323-31. [PMID: 24327420 DOI: 10.1002/jlcr.3158] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/29/2013] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) and related dementias show increasing clinical prevalence, yet our understanding of the etiology and pathobiology of disease-related neurodegeneration remains limited. In this regard, noninvasive imaging with radiotracers for positron emission tomography (PET) presents a unique tool for quantifying spatial and temporal changes in characteristic biological markers of brain disease and for assessing potential drug efficacy. PET radiotracers targeting different protein markers are being developed to address questions pertaining to the molecular and/or genetic heterogeneity of AD and related dementias. For example, radiotracers including [(11) C]-PiB and [(18) F]-AV-45 (Florbetapir) are being used to measure the density of Aβ-plaques in AD patients and to interrogate the biological mechanisms of disease initiation and progression. Our focus is on the development of novel PET imaging agents, targeting proteins beyond Aβ-plaques, which can be used to investigate the broader mechanism of AD pathogenesis. Here, we present the chemical basis of various radiotracers which show promise in preclinical or clinical studies for use in evaluating the phenotypic or biochemical characteristics of AD. Radiotracers for PET imaging neuroinflammation, metal ion association with Aβ-plaques, tau protein, cholinergic and cannabinoid receptors, and enzymes including glycogen-synthase kinase-3β and monoamine oxidase B amongst others, and their connection to AD are highlighted.
Collapse
Affiliation(s)
- Jason P Holland
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Department of Radiology, Harvard Medical School, 55 Fruit St., White 427, Boston, Massachusetts, 02114, USA; Life Sciences, Australian Nuclear Science and Technology Organisation, Kirrawee, New South Wales, 2232, Australia
| | | | | | | | | | | | | |
Collapse
|
131
|
Latrepirdine: molecular mechanisms underlying potential therapeutic roles in Alzheimer's and other neurodegenerative diseases. Transl Psychiatry 2013; 3:e332. [PMID: 24301650 PMCID: PMC4030329 DOI: 10.1038/tp.2013.97] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Revised: 09/19/2013] [Accepted: 09/26/2013] [Indexed: 02/06/2023] Open
Abstract
Latrepirdine (Dimebon(TM)) was originally marketed as a non-selective antihistamine in Russia. It was repurposed as an effective treatment for patients suffering from Alzheimer's disease (AD) and Huntington's disease (HD) following preliminary reports showing its neuroprotective functions and ability to enhance cognition in AD and HD models. However, latrepirdine failed to show efficacy in phase III trials in AD and HD patients following encouraging phase II trials. The failure of latrepirdine in the clinical trials has highlighted the importance of understanding the precise mechanism underlying its cognitive benefits in neurodegenerative diseases before clinical evaluation. Latrepirdine has shown to affect a number of cellular functions including multireceptor activity, mitochondrial function, calcium influx and intracellular catabolic pathways; however, it is unclear how these properties contribute to its clinical benefits. Here, we review the studies investigating latrepirdine in cellular and animal models to provide a complete evaluation of its mechanisms of action in the central nervous system. In addition, we review recent studies that demonstrate neuroprotective functions for latrepirdine-related class of molecules including the β-carbolines and aminopropyl carbazoles in AD, Parkinson's disease and amyotrophic lateral sclerosis models. Assessment of their neuroprotective effects and underlying biological functions presents obvious value for developing structural analogues of latrepirdine for dementia treatment.
Collapse
|
132
|
Zhang X, Tian Y, Li Z, Tian X, Sun H, Liu H, Moore A, Ran C. Design and synthesis of curcumin analogues for in vivo fluorescence imaging and inhibiting copper-induced cross-linking of amyloid beta species in Alzheimer's disease. J Am Chem Soc 2013; 135:16397-409. [PMID: 24116384 DOI: 10.1021/ja405239v] [Citation(s) in RCA: 222] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In this article, we first designed and synthesized curcumin-based near-infrared (NIR) fluorescence imaging probes for detecting both soluble and insoluble amyloid beta (Aβ) species and then an inhibitor that could attenuate cross-linking of Aβ induced by copper. According to our previous results and the possible structural stereohindrance compatibility of the Aβ peptide and the hydrophobic/hydrophilic property of the Aβ13-20 (HHQKLVFF) fragment, NIR imaging probe CRANAD-58 was designed and synthesized. As expected CRANAD-58 showed significant fluorescence property changes upon mixing with both soluble and insoluble Aβ species in vitro. In vivo NIR imaging revealed that CRANAD-58 was capable of differentiating transgenic and wild-type mice as young as 4 months old, the age that lacks apparently visible Aβ plaques and Aβ is likely in its soluble forms. According to our limited studies on the interaction mechanism between CRANAD-58 and Aβ, we also designed CRANAD-17 to attenuate the cross-linking of Aβ42 induced by copper. It is well-known that the coordination of copper with imidazoles on Histidine-13 and 14 (H13, H14) of Aβ peptides could initialize covalent cross-linking of Aβ. In CRANAD-17, a curcumin scaffold was used as an anchoring moiety to usher the designed compound to the vicinity of H13 and H14 of Aβ, and imidazole rings were incorporated to compete with H13/H14 for copper binding. The results of SDS-PAGE gel and Western blot indicated that CRANAD-17 was capable of inhibiting Aβ42 cross-linking induced by copper. This raises a potential for CRANAD-17 to be considered for AD therapy.
Collapse
Affiliation(s)
- Xueli Zhang
- Molecular Imaging Laboratory, MGH/MIT/HMS Athinoula A. Martinos Center for Biomedical Imaging, Department of Radiology, Massachusetts General Hospital/Harvard Medical School , Building 75, Charlestown, Massachusetts 02129, United States
| | | | | | | | | | | | | | | |
Collapse
|
133
|
Combination therapy in a transgenic model of Alzheimer's disease. Exp Neurol 2013; 250:228-238. [PMID: 24120437 DOI: 10.1016/j.expneurol.2013.10.001] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2013] [Revised: 09/16/2013] [Accepted: 10/02/2013] [Indexed: 12/31/2022]
Abstract
The pathological accumulation of the β-amyloid protein (Aβ) has been closely associated with synaptic loss and neurotoxicity contributing to cognitive dysfunction in Alzheimer's disease (AD). Oligomers of Aβ42 appear to be the most neurotoxic form. Two of the most promising attempts to reduce Aβ accumulation have been with scyllo-inositol, an inositol steroisomer, that stabilizes Aβ42 peptide and prevents it from progressing to oligomers and fibrils and R-flurbiprofen, a purified enantiomer of the classical racemic non-steroidal anti-inflammatory drugs (NSAID), flurbiprofen, that retains the ability to specifically lower Aβ42. In the present study we evaluated the effects of scyllo-inositol and the combination treatment of scyllo-inositol+R-flurbiprofen on amyloid pathology and hippocampal-dependent memory function in 5XFAD mice, a model of Aβ pathology characterized by an enormous production of Aβ42. Our expectations were that the combination treatment of scyllo-inositol+R-flurbiprofen would have an additive effect in preventing Aβ accumulation and that cognition would be improved. Mice treated with scyllo-inositol exhibit 41 and 35% reduction in the deposition of the amyloid plaques stained by antibody against Aβ42 and Aβ40 respectively. Scyllo-inositol was not more effective when combined with R-flurbiprofen for the measures tested. Scyllo-inositol treated mice performed significantly better at the radial arm water maze (RAWM) task than untreated and scyllo-inositol+R-flurbiprofen treated mice.
Collapse
|
134
|
Mucke L, Selkoe DJ. Neurotoxicity of amyloid β-protein: synaptic and network dysfunction. Cold Spring Harb Perspect Med 2013; 2:a006338. [PMID: 22762015 DOI: 10.1101/cshperspect.a006338] [Citation(s) in RCA: 747] [Impact Index Per Article: 62.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Evidence for an ever-expanding variety of molecular mediators of amyloid β-protein neurotoxicity (membrane lipids, receptor proteins, channel proteins, second messengers and related signaling cascades, cytoskeletal proteins, inflammatory mediators, etc.) has led to the notion that the binding of hydrophobic Aβ assemblies to cellular membranes triggers multiple effects affecting diverse pathways. It appears unlikely that there are only one or two cognate receptors for neurotoxic forms of Aβ and also that there are just one or two assembly forms of the peptide that induce neuronal dysfunction. Rather, various soluble (diffusible) oligomers of Aβ that may be in dynamic equilibrium with insoluble, fibrillar deposits (amyloid plaques) and that can bind to different components of neuronal and non-neuronal plasma membranes appear to induce complex patterns of synaptic dysfunction and network disorganization that underlie the intermittent but gradually progressive cognitive manifestations of the clinical disorder. Modern analyses of this problem utilize electrophysiology coupled with synaptic biochemistry and behavioral phenotyping of animal models to elucidate the affected circuits and assess the effects of potential therapeutic interventions.
Collapse
Affiliation(s)
- Lennart Mucke
- Gladstone Institute of Neurological Disease and University of California, San Francisco, San Francisco, California, USA.
| | | |
Collapse
|
135
|
Jiang L, Liu C, Leibly D, Landau M, Zhao M, Hughes MP, Eisenberg DS. Structure-based discovery of fiber-binding compounds that reduce the cytotoxicity of amyloid beta. eLife 2013. [PMID: 23878726 DOI: 10.7554/elife.00857.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Amyloid protein aggregates are associated with dozens of devastating diseases including Alzheimer's, Parkinson's, ALS, and diabetes type 2. While structure-based discovery of compounds has been effective in combating numerous infectious and metabolic diseases, ignorance of amyloid structure has hindered similar approaches to amyloid disease. Here we show that knowledge of the atomic structure of one of the adhesive, steric-zipper segments of the amyloid-beta (Aβ) protein of Alzheimer's disease, when coupled with computational methods, identifies eight diverse but mainly flat compounds and three compound derivatives that reduce Aβ cytotoxicity against mammalian cells by up to 90%. Although these compounds bind to Aβ fibers, they do not reduce fiber formation of Aβ. Structure-activity relationship studies of the fiber-binding compounds and their derivatives suggest that compound binding increases fiber stability and decreases fiber toxicity, perhaps by shifting the equilibrium of Aβ from oligomers to fibers. DOI:http://dx.doi.org/10.7554/eLife.00857.001.
Collapse
Affiliation(s)
- Lin Jiang
- Departments of Chemistry and Biochemistry and Biological Chemistry , Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles , Los Angeles , United States
| | | | | | | | | | | | | |
Collapse
|
136
|
Jiang L, Liu C, Leibly D, Landau M, Zhao M, Hughes MP, Eisenberg DS. Structure-based discovery of fiber-binding compounds that reduce the cytotoxicity of amyloid beta. eLife 2013; 2:e00857. [PMID: 23878726 PMCID: PMC3713518 DOI: 10.7554/elife.00857] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Accepted: 06/10/2013] [Indexed: 12/15/2022] Open
Abstract
Amyloid protein aggregates are associated with dozens of devastating diseases including Alzheimer’s, Parkinson’s, ALS, and diabetes type 2. While structure-based discovery of compounds has been effective in combating numerous infectious and metabolic diseases, ignorance of amyloid structure has hindered similar approaches to amyloid disease. Here we show that knowledge of the atomic structure of one of the adhesive, steric-zipper segments of the amyloid-beta (Aβ) protein of Alzheimer’s disease, when coupled with computational methods, identifies eight diverse but mainly flat compounds and three compound derivatives that reduce Aβ cytotoxicity against mammalian cells by up to 90%. Although these compounds bind to Aβ fibers, they do not reduce fiber formation of Aβ. Structure-activity relationship studies of the fiber-binding compounds and their derivatives suggest that compound binding increases fiber stability and decreases fiber toxicity, perhaps by shifting the equilibrium of Aβ from oligomers to fibers. DOI:http://dx.doi.org/10.7554/eLife.00857.001 Alzheimer’s disease is the most common form of dementia, estimated to affect roughly five million people in the United States, and its incidence is steadily increasing as the population ages. A pathological hallmark of Alzheimer’s disease is the presence in the brain of aggregates of two proteins: tangles of a protein called tau; and fibers and smaller units (oligomers) of a peptide called amyloid beta. Many attempts have been made to screen libraries of natural and synthetic compounds to identify substances that might prevent the aggregation and toxicity of amyloid. Such studies revealed that polyphenols found in green tea and in the spice turmeric can inhibit the formation of amyloid fibrils. Moreover, a number of dyes reduce the toxic effects of amyloid on cells, although significant side effects prevent these from being used as drugs. Structure-based drug design, in which the structure of a target protein is used to help identify compounds that will interact with it, has been used to generate therapeutic agents for a number of diseases. Here, Jiang et al. report the first application of this technique in the hunt for compounds that inhibit the cytotoxicity of amyloid beta. Using the known atomic structure of the protein in complex with a dye, Jiang et al. performed a computational screen of 18,000 compounds in search of those that are likely to bind effectively. The compounds that showed the strongest predicted binding were then tested for their ability to interfere with the aggregation of amyloid beta and to protect cells grown in culture from its toxic effects. Compounds that reduced toxicity did not reduce the abundance of protein aggregates, but they appear to increase the stability of fibrils. This is consistent with other evidence suggesting that small, soluble forms (oligomers) of amyloid beta that break free from the fibrils may be the toxic agent in Alzheimer’s disease, rather than the fibrils themselves. In addition to uncovering compounds with therapeutic potential in Alzheimer’s disease, this work presents a new approach for identifying proteins that bind to amyloid fibrils. Given that amyloid accumulation is a feature of many other diseases, including Parkinson’s disease, Huntington’s disease and type 2 diabetes, the approach could have broad therapeutic applications. DOI:http://dx.doi.org/10.7554/eLife.00857.002
Collapse
Affiliation(s)
- Lin Jiang
- Departments of Chemistry and Biochemistry and Biological Chemistry , Howard Hughes Medical Institute, UCLA-DOE Institute for Genomics and Proteomics, University of California, Los Angeles , Los Angeles , United States
| | | | | | | | | | | | | |
Collapse
|
137
|
Luo Y, Vali S, Sun S, Chen X, Liang X, Drozhzhina T, Popugaeva E, Bezprozvanny I. Aβ42-binding peptoids as amyloid aggregation inhibitors and detection ligands. ACS Chem Neurosci 2013; 4:952-62. [PMID: 23427915 DOI: 10.1021/cn400011f] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is the most common form of dementia and currently affects 5.4 million Americans. A number of anti-Aβ (beta amyloid) therapeutic agents have been developed for AD, but so far all of them failed in clinic. Here we used peptoid chemistry to develop ligands selective for Aβ42. Peptoids are N-substituted glycine oligomers, a class of peptidomimics. We synthesized an on-bead peptoid library consisting of 38,416 unique peptoids. The generated peptoid library was screened and arrays of Aβ42-selective peptoid ligands were identified. One of those peptoid ligands, IAM1 (inhibitor of amyloid), and the dimeric form (IAM1)2 were synthesized and evaluated in a variety of biochemical assays. We discovered that IAM1 selectively binds to Aβ42, while the dimeric derivative (IAM1)2 has a higher affinity for Aβ42. Furthermore, we demonstrated that IAM1 and (IAM1)2 were able to inhibit the aggregation of Aβ42 in a concentration-dependent manner, and that (IAM1)2 protected primary hippocampal neurons from the Aβ-induced toxicity in vitro. These results suggest that IAM1 and (IAM1)2 are specific Aβ42 ligands with antiaggregation and neuroprotective properties. IAM1, (IAM1)2, and their derivatives hold promise as Aβ42 detection agents and as lead compounds for the development of AD therapeutic agents.
Collapse
Affiliation(s)
- Yuan Luo
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Sheetal Vali
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Suya Sun
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Xuesong Chen
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Xia Liang
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
| | - Tatiana Drozhzhina
- Laboratory of Molecular Neurodegeneration, State Technical University, St. Petersburg 195251,
Russia
| | - Elena Popugaeva
- Laboratory of Molecular Neurodegeneration, State Technical University, St. Petersburg 195251,
Russia
| | - Ilya Bezprozvanny
- Department of Physiology, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390, United States
- Laboratory of Molecular Neurodegeneration, State Technical University, St. Petersburg 195251,
Russia
| |
Collapse
|
138
|
Reactive astrocytes associated with plaques in TgCRND8 mouse brain and in human Alzheimer brain express phosphoprotein enriched in astrocytes (PEA-15). FEBS Lett 2013; 587:2448-54. [DOI: 10.1016/j.febslet.2013.06.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Revised: 04/30/2013] [Accepted: 06/06/2013] [Indexed: 01/02/2023]
|
139
|
Goutagny R, Gu N, Cavanagh C, Jackson J, Chabot JG, Quirion R, Krantic S, Williams S. Alterations in hippocampal network oscillations and theta-gamma coupling arise before Aβ overproduction in a mouse model of Alzheimer's disease. Eur J Neurosci 2013; 37:1896-902. [DOI: 10.1111/ejn.12233] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 03/26/2013] [Accepted: 03/28/2013] [Indexed: 12/31/2022]
Affiliation(s)
| | - Ning Gu
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| | - Chelsea Cavanagh
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| | - Jesse Jackson
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| | - Jean-Guy Chabot
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| | - Rémi Quirion
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| | - Slavica Krantic
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| | - Sylvain Williams
- Douglas Mental Health University Institute; McGill University; 6875 Lasalle blvd; Verdun; Montreal; H4H1R3; QC; Canada
| |
Collapse
|
140
|
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: 86] [Impact Index Per Article: 7.2] [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
|
141
|
Li G, Pomès R. Binding mechanism of inositol stereoisomers to monomers and aggregates of Aβ(16-22). J Phys Chem B 2013; 117:6603-13. [PMID: 23627280 DOI: 10.1021/jp311350r] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alzheimer's disease (AD) is a severe neurodegenerative disease with no cure. A potential therapeutic approach is to prevent or reverse the amyloid formation of Aβ42, a key pathological hallmark of AD. We examine the molecular basis for stereochemistry-dependent inhibition of the formation of Aβ fibrils in vitro by a polyol, scyllo-inositol. We present molecular dynamics simulations of the monomeric, disordered aggregate, and protofibrillar states of Aβ(16-22), an amyloid-forming peptide fragment of full-length Aβ, successively with and without scyllo-inositol and its inactive stereoisomer chiro-inositol. Both stereoisomers bind monomers and disordered aggregates with similar affinities of 10-120 mM, whereas binding to β-sheet-containing protofibrils yields affinities of 0.2-0.5 mM commensurate with in vitro inhibitory concentrations of scyllo-inositol. Moreover, scyllo-inositol displays a higher binding specificity for phenylalanine-lined grooves on the protofibril surface, suggesting that scyllo-inositol coats the surface of Aβ protofibrils and disrupts their lateral stacking into amyloid fibrils.
Collapse
Affiliation(s)
- Grace Li
- Department of Biochemistry, University of Toronto, 27 King's College Circle, Toronto, Ontario, Canada M5S 1A1
| | | |
Collapse
|
142
|
Yamashita Y, Yamaoka M, Hasunuma T, Ashida H, Yoshida KI. Detection of orally administered inositol stereoisomers in mouse blood plasma and their effects on translocation of glucose transporter 4 in skeletal muscle cells. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2013; 61:4850-4854. [PMID: 23641877 DOI: 10.1021/jf305322t] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Simple pharmacological studies on inositol stereoisomers are presented in this study. Male ICR mice were orally administered 1 g/kg BW of three inositol stereoisomers, myo-inositol (MI), d-chiro-inositol (DCI), and scyllo-inositol (SI), and blood plasma samples and skeletal muscle fractions were prepared after an hour. The plasma samples were subjected to gas chromatography-coupled time-of-flight mass spectrometry (GC-TOF-MS) analysis. None of the three stereoisomers was seen in untreated samples, but substantial amounts ranging from 2.5 to 6.5 mM were detected only after administration, indicating that orally administered inositol stereoisomers were readily absorbed and their levels elevated in the bloodstream. In addition, plasma of SI-administered animals contained substantial MI, suggesting a possible metabolic conversion of SI to MI. In the skeletal muscle fractions, glucose transporter type 4 (GLUT4) content in the plasma membrane increased, indicating that inositol stereoisomers stimulated GLUT4 translocation.
Collapse
Affiliation(s)
- Yoko Yamashita
- Department of Agrobioscience and ‡Organization of Advanced Science and Technology, Kobe University , 1-1 Rokkodai, Nada, Kobe 657-8501, Japan
| | | | | | | | | |
Collapse
|
143
|
Ng OTW, Wong Y, Chan HM, Cheng J, Qi X, Chan WH, Yung KKL, Li HW. N-Acetyl-l-cysteine capped quantum dots offer neuronal cell protection by inhibiting beta (1-40) amyloid fibrillation. Biomater Sci 2013; 1:577-580. [PMID: 32481833 DOI: 10.1039/c3bm60029g] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
This is the first work that revealed the neuro-protective effect of functionalized quantum dots against the cytotoxicity induced by beta-amyloid peptides. This study gives insight into the future treatment of Alzheimer's disease. It opens many avenues for the development of the next generation nanotechnology for biomedical and therapeutic applications.
Collapse
Affiliation(s)
- Olivia T W Ng
- Department of Biology, Hong Kong Baptist University, Kowloon Tong, Hong Kong, P. R. China.
| | | | | | | | | | | | | | | |
Collapse
|
144
|
Wang F, Blanchard AP, Elisma F, Granger M, Xu H, Bennett SAL, Figeys D, Zou H. Phosphoproteome analysis of an early onset mouse model (TgCRND8) of Alzheimer's disease reveals temporal changes in neuronal and glia signaling pathways. Proteomics 2013; 13:1292-305. [DOI: 10.1002/pmic.201200415] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2012] [Revised: 10/19/2012] [Accepted: 11/08/2012] [Indexed: 12/12/2022]
Affiliation(s)
- Fangjun Wang
- Key Lab of Separation Sciences for Analytical Chemistry; National Chromatographic R & A Center; Dalian Institute of Chemical Physics; Chinese Academy of Sciences; Dalian P. R. China
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
| | - Alexandre P. Blanchard
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
- Neural Regeneration Laboratory; Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa Canada
| | - Fred Elisma
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
| | - Matthew Granger
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
- Neural Regeneration Laboratory; Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa Canada
| | - Hongbin Xu
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
- Neural Regeneration Laboratory; Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa Canada
| | - Steffany A. L. Bennett
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
- Neural Regeneration Laboratory; Department of Biochemistry, Microbiology, and Immunology; University of Ottawa; Ottawa Canada
| | - Daniel Figeys
- Ottawa Institute of Systems Biology; University of Ottawa; Ottawa Canada
| | - Hanfa Zou
- Key Lab of Separation Sciences for Analytical Chemistry; National Chromatographic R & A Center; Dalian Institute of Chemical Physics; Chinese Academy of Sciences; Dalian P. R. China
| |
Collapse
|
145
|
O'Hare E, Scopes DI, Kim EM, Palmer P, Jones M, Whyment AD, Spanswick D, Amijee H, Nerou E, McMahon B, Treherne JM, Jeggo R. Orally bioavailable small molecule drug protects memory in Alzheimer's disease models. Neurobiol Aging 2013; 34:1116-25. [DOI: 10.1016/j.neurobiolaging.2012.10.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2012] [Revised: 09/21/2012] [Accepted: 10/19/2012] [Indexed: 12/19/2022]
|
146
|
Liang E, Garzone P, Cedarbaum JM, Koller M, Tran T, Xu V, Ross B, Jhee SS, Ereshefsky L, Pastrak A, Abushakra S. Pharmacokinetic Profile of Orally Administered Scyllo-Inositol (Elnd005) in Plasma, Cerebrospinal Fluid and Brain, and Corresponding Effect on Amyloid-Beta in Healthy Subjects. Clin Pharmacol Drug Dev 2013; 2:186-94. [DOI: 10.1002/cpdd.14] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Accepted: 11/30/2012] [Indexed: 11/06/2022]
Affiliation(s)
- Earvin Liang
- Elan Pharmaceuticals, Inc.; South San Francisco, CA; USA
| | - Pamela Garzone
- Elan Pharmaceuticals, Inc.; South San Francisco, CA; USA
| | | | - Martin Koller
- Elan Pharmaceuticals, Inc.; South San Francisco, CA; USA
| | - Thao Tran
- Huntington Medical Research Institutes; Pasadena, CA; USA
| | - Victor Xu
- Huntington Medical Research Institutes; Pasadena, CA; USA
| | - Brian Ross
- Huntington Medical Research Institutes; Pasadena, CA; USA
| | - Stanford S. Jhee
- PAREXEL International Early Phase (formerly California Clinical Trials); Glendale, CA; USA
| | - Larry Ereshefsky
- The University of Texas Health Science Center San Antonio; San Antonio, TX; USA
| | | | | |
Collapse
|
147
|
Wong HE, Irwin JA, Kwon I. Halogenation generates effective modulators of amyloid-Beta aggregation and neurotoxicity. PLoS One 2013; 8:e57288. [PMID: 23468958 PMCID: PMC3585355 DOI: 10.1371/journal.pone.0057288] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2012] [Accepted: 01/22/2013] [Indexed: 11/19/2022] Open
Abstract
Halogenation of organic compounds plays diverse roles in biochemistry, including selective chemical modification of proteins and improved oral absorption/blood-brain barrier permeability of drug candidates. Moreover, halogenation of aromatic molecules greatly affects aromatic interaction-mediated self-assembly processes, including amyloid fibril formation. Perturbation of the aromatic interaction caused by halogenation of peptide building blocks is known to affect the morphology and other physical properties of the fibrillar structure. Consequently, in this article, we investigated the ability of halogenated ligands to modulate the self-assembly of amyloidogenic peptide/protein. As a model system, we chose amyloid-beta peptide (Aβ), which is implicated in Alzheimer’s disease, and a novel modulator of Aβ aggregation, erythrosine B (ERB). Considering that four halogen atoms are attached to the xanthene benzoate group in ERB, we hypothesized that halogenation of the xanthene benzoate plays a critical role in modulating Aβ aggregation and cytotoxicity. Therefore, we evaluated the modulating capacities of four ERB analogs containing different types and numbers of halogen atoms as well as fluorescein as a negative control. We found that fluorescein is not an effective modulator of Aβ aggregation and cytotoxicity. However, halogenation of either the xanthenes or benzoate ring of fluorescein substantially enhanced the inhibitory capacity on Aβ aggregation. Such Aβ aggregation inhibition by ERB analogs except rose bengal correlated well to the inhibition of Aβ cytotoxicity. To our knowledge, this is the first report demonstrating that halogenation of aromatic rings substantially enhance inhibitory capacities of small molecules on Aβ-associated neurotoxicity via Aβ aggregation modulation.
Collapse
Affiliation(s)
- H. Edward Wong
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, Unites States of America
| | - Jacob A. Irwin
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, Unites States of America
| | - Inchan Kwon
- Department of Chemical Engineering, University of Virginia, Charlottesville, Virginia, Unites States of America
- Institutes on Aging, University of Virginia, Charlottesville, Virginia, Unites States of America
- * E-mail:
| |
Collapse
|
148
|
Belluti F, Rampa A, Gobbi S, Bisi A. Small-molecule inhibitors/modulators of amyloid-β peptide aggregation and toxicity for the treatment of Alzheimer's disease: a patent review (2010 - 2012). Expert Opin Ther Pat 2013; 23:581-96. [PMID: 23425062 DOI: 10.1517/13543776.2013.772983] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Genetic, physiological, and biochemical data indicate that agglomerates of the 42-amino acid form of the amyloid-β (Aβ(42)) peptide are strongly linked to Alzheimer's disease (AD) etiology and thus represent a particularly attractive target for the development of an effective disease-modifying approach for AD treatment. A plethora of chemical entities able to modulate Aβ(42) self-assembly have been developed in recent years, among them, several are in clinical or preclinical development. AREAS COVERED This review accounts for small-molecule inhibitors of Aβ peptide polymerization and toxicity, reported in the patent literature during the 2010 - 2012 period, and their potential use as disease-modifying therapeutics for AD cure. EXPERT OPINION The earliest pathogenic event is the formation of soluble Aβ oligomers that disrupt synaptic communication. Drug design strategies targeting these primary toxic agents could hold considerable promises for obtaining effective anti-AD drugs candidate. The heterogeneous aggregation of Aβ and the resulting difficulty to structurally characterize the peptide represent important drawbacks.
Collapse
Affiliation(s)
- Federica Belluti
- Alma MaterStudiorum-Bologna University, Department of Pharmacy and Biotechnologies, Bologna, Italy.
| | | | | | | |
Collapse
|
149
|
Comparative docking and ADMET study of some curcumin derivatives and herbal congeners targeting β-amyloid. ACTA ACUST UNITED AC 2013. [DOI: 10.1007/s13721-012-0021-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
150
|
Abstract
Alzheimer’s disease (AD), considered the commonest neurodegenerative cause of dementia, is associated with hallmark pathologies including extracellular amyloid-β protein (Aβ) deposition in extracellular senile plaques and vessels, and intraneuronal tau deposition as neurofibrillary tangles. Although AD is usually categorized as neurodegeneration distinct from cerebrovascular disease (CVD), studies have shown strong links between AD and CVD. There is evidence that vascular risk factors and CVD may accelerate Aβ 40-42 production/ aggregation/deposition and contribute to the pathology and symptomatology of AD. Aβ deposited along vessels also causes cerebral amyloid angiopathy. Amyloid imaging allows in vivo detection of AD pathology, opening the way for prevention and early treatment, if disease-modifying therapies in the pipeline show safety and efficacy. In this review, we review the role of vascular factors and Aβ, underlining that vascular risk factor management may be important for AD prevention and treatment.
Collapse
|