1
|
Guan S, Li Y, Xin Y, Wang D, Lu P, Han F, Xu H. Deciphering the dual role of N-methyl-D-Aspartate receptor in postoperative cognitive dysfunction: A comprehensive review. Eur J Pharmacol 2024; 971:176520. [PMID: 38527701 DOI: 10.1016/j.ejphar.2024.176520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/03/2024] [Accepted: 03/20/2024] [Indexed: 03/27/2024]
Abstract
Postoperative cognitive dysfunction (POCD) is a common complication following surgery, adversely impacting patients' recovery, increasing the risk of negative outcomes, prolonged hospitalization, and higher mortality rates. The N-methyl-D-aspartate (NMDA) receptor, crucial for learning, memory, and synaptic plasticity, plays a significant role in the development of POCD. Various perioperative factors, including age and anesthetic use, can reduce NMDA receptor function, while surgical stress, inflammation, and pain may lead to its excessive activation. This review consolidates preclinical and clinical research to explore the intricate relationship between perioperative factors affecting NMDA receptor functionality and the onset of POCD. It discusses the influence of aging, anesthetic administration, perioperative injury, pain, and inflammation on the NMDA receptor-related pathophysiology of POCD. The comprehensive analysis presented aims to identify effective treatment targets for POCD, contributing to the improvement of patient outcomes post-surgery.
Collapse
Affiliation(s)
- Shaodi Guan
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yali Li
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Yueyang Xin
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Danning Wang
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Pei Lu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Fanglong Han
- Department of Anesthesiology, Xiangyang Maternal and Child Health Hospital, Xiangyang, 441003, China
| | - Hui Xu
- Department of Anesthesiology and Pain Medicine, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
| |
Collapse
|
2
|
de la Monte SM, Tong M. Agent Orange Herbicidal Toxin-Initiation of Alzheimer-Type Neurodegeneration. J Alzheimers Dis 2024; 97:1703-1726. [PMID: 38306038 PMCID: PMC10979462 DOI: 10.3233/jad-230881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2024]
Abstract
Background Agent Orange (AO) is a Vietnam War-era herbicide that contains a 1 : 1 ratio of 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). Emerging evidence suggests that AO exposures cause toxic and degenerative pathologies that may increase the risk for Alzheimer's disease (AD). Objective This study investigates the effects of the two main AO constituents on key molecular and biochemical indices of AD-type neurodegeneration. Methods Long Evans rat frontal lobe slice cultures treated with 250μg/ml of 2,4-D, 2,4,5-T, or both (D + T) were evaluated for cytotoxicity, oxidative injury, mitochondrial function, and AD biomarker expression. Results Treatment with the AO constituents caused histopathological changes corresponding to neuronal, white matter, and endothelial cell degeneration, and molecular/biochemical abnormalities indicative of cytotoxic injury, lipid peroxidation, DNA damage, and increased immunoreactivity to activated Caspase 3, glial fibrillary acidic protein, ubiquitin, tau, paired-helical filament phosphorylated tau, AβPP, Aβ, and choline acetyltransferase. Nearly all indices of cellular injury and degeneration were more pronounced in the D + T compared with 2,4-D or 2,4,5-T treated cultures. Conclusions Exposures to AO herbicidal chemicals damage frontal lobe brain tissue with molecular and biochemical abnormalities that mimic pathologies associated with early-stage AD-type neurodegeneration. Additional research is needed to evaluate the long-term effects of AO exposures in relation to aging and progressive neurodegeneration in Vietnam War Veterans.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Departments of Pathology and Laboratory Medicine, Neurology, and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
3
|
de la Monte SM, Goel A, Tong M, Delikkaya B. Agent Orange Causes Metabolic Dysfunction and Molecular Pathology Reminiscent of Alzheimer's Disease. J Alzheimers Dis Rep 2023; 7:751-766. [PMID: 37662613 PMCID: PMC10473158 DOI: 10.3233/adr-230046] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 06/19/2023] [Indexed: 09/05/2023] Open
Abstract
Background Agent Orange, an herbicide used during the Vietnam War, contains 2,4-dichlorophenoxyacetic acid (2,4-D) and 2,4,5-trichlorophenoxyacetic acid (2,4,5-T). Agent Orange has teratogenic and carcinogenic effects, and population-based studies suggest Agent Orange exposures lead to higher rates of toxic and degenerative pathologies in the peripheral and central nervous system (CNS). Objective This study examines the potential contribution of Agent Orange exposures to neurodegeneration. Methods Human CNS-derived neuroepithelial cells (PNET2) treated with 2,4-D and 2,4,5-T were evaluated for viability, mitochondrial function, and Alzheimer's disease (AD)-related proteins. Results Treatment with 250μg/ml 2,4-D or 2,4,5-T significantly impaired mitochondrial function, caused degenerative morphological changes, and reduced viability in PNET2 cells. Correspondingly, glyceraldehyde-3-phosphate dehydrogenase expression which is insulin-regulated and marks the integrity of carbohydrate metabolism, was significantly inhibited while 4-hydroxy-2-nonenal, a marker of lipid peroxidation, was increased. Tau neuronal cytoskeletal protein was significantly reduced by 2,4,5-T, and relative tau phosphorylation was progressively elevated by 2,4,5-T followed by 2,4-D treatment relative to control. Amyloid-β protein precursor (AβPP) was increased by 2,4,5-T and 2,4-D, and 2,4,5-T caused a statistical trend (0.05 < p<0.10) increase in Aβ. Finally, altered cholinergic function due to 2,4,5-T and 2,4-D exposures was marked by significantly increased choline acetyltransferase and decreased acetylcholinesterase expression, corresponding with responses in early-stage AD. Conclusion Exposures to Agent Orange herbicidal chemicals rapidly damage CNS neurons, initiating a path toward AD-type neurodegeneration. Additional research is needed to understand the permanency of these neuropathologic processes and the added risks of developing AD in Agent Orange-exposed aging Vietnam Veterans.
Collapse
Affiliation(s)
- Suzanne M. de la Monte
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
- Department of Neurology and Neurosurgery, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Anuva Goel
- Department of Neuroscience, Brown University, Providence, RI, USA
| | - Ming Tong
- Department of Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Busra Delikkaya
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Lifespan Academic Institutions, and the Warren Alpert Medical School of Brown University, Providence, RI, USA
| |
Collapse
|
4
|
Therapeutic Potential and Limitation of Serotonin Type 7 Receptor Modulation. Int J Mol Sci 2023; 24:ijms24032070. [PMID: 36768393 PMCID: PMC9916679 DOI: 10.3390/ijms24032070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/15/2023] [Accepted: 01/18/2023] [Indexed: 01/21/2023] Open
Abstract
Although a number of mood-stabilising atypical antipsychotics and antidepressants modulate serotonin type 7 receptor (5-HT7), the detailed contributions of 5-HT7 function to clinical efficacy and pathophysiology have not been fully understood. The mood-stabilising antipsychotic agent, lurasidone, and the serotonin partial agonist reuptake inhibitor, vortioxetine, exhibit higher binding affinity to 5-HT7 than other conventional antipsychotics and antidepressants. To date, the initially expected rapid onset of antidepressant effects-in comparison with conventional antidepressants or mood-stabilising antipsychotics-due to 5-HT7 inhibition has not been observed with lurasidone and vortioxetine; however, several clinical studies suggest that 5-HT7 inhibition likely contributes to quality of life of patients with schizophrenia and mood disorders via the improvement of cognition. Furthermore, recent preclinical studies reported that 5-HT7 inhibition might mitigate antipsychotic-induced weight gain and metabolic complication by blocking other monoamine receptors. Further preclinical studies for the development of 5-HT7 modulation against neurodevelopmental disorders and neurodegenerative diseases have been ongoing. To date, various findings from various preclinical studies indicate the possibility that 5-HT7 modifications can provide two independent strategies. The first is that 5-HT7 inhibition ameliorates the dysfunction of inter-neuronal transmission in mature networks. The other is that activation of 5-HT7 can improve transmission dysfunction due to microstructure abnormality in the neurotransmission network-which could be unaffected by conventional therapeutic agents-via modulating intracellular signalling during the neurodevelopmental stage or via loss of neural networks with aging. This review attempts to describe the current and novel clinical applications of 5-HT7 modulation based on preclinical findings.
Collapse
|
5
|
Mosilhy EA, Alshial EE, Eltaras MM, Rahman MMA, Helmy HI, Elazoul AH, Hamdy O, Mohammed HS. Non-invasive transcranial brain modulation for neurological disorders treatment: A narrative review. Life Sci 2022; 307:120869. [DOI: 10.1016/j.lfs.2022.120869] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Revised: 08/01/2022] [Accepted: 08/03/2022] [Indexed: 11/30/2022]
|
6
|
Perez SD, Phillips JS, Norise C, Kinney NG, Vaddi P, Halpin A, Rascovsky K, Irwin DJ, McMillan CT, Xie L, Wisse LE, Yushkevich PA, Kallogjeri D, Grossman M, Cousins KA. Neuropsychological and Neuroanatomical Features of Patients with Behavioral/Dysexecutive Variant Alzheimer’s disease (AD): A Comparison to Behavioral Variant Frontotemporal Dementia and Amnestic AD Groups. J Alzheimers Dis 2022; 89:641-658. [PMID: 35938245 PMCID: PMC10117623 DOI: 10.3233/jad-215728] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: An understudied variant of Alzheimer’s disease (AD), the behavioral/dysexecutive variant of AD (bvAD), is associated with progressive personality, behavior, and/or executive dysfunction and frontal atrophy. Objective: This study characterizes the neuropsychological and neuroanatomical features associated with bvAD by comparing it to behavioral variant frontotemporal dementia (bvFTD), amnestic AD (aAD), and subjects with normal cognition. Methods: Subjects included 16 bvAD, 67 bvFTD, and 18 aAD patients, and 26 healthy controls. Neuropsychological assessment and MRI data were compared between these groups. Results: Compared to bvFTD, bvAD showed more significant visuospatial impairments (Rey Figure copy and recall), more irritability (Neuropsychological Inventory), and equivalent verbal memory (Philadelphia Verbal Learning Test). Compared to aAD, bvAD indicated more executive dysfunction (F-letter fluency) and better visuospatial performance. Neuroimaging analysis found that bvAD showed cortical thinning relative to bvFTD posteriorly in left temporal-occipital regions; bvFTD had cortical thinning relative to bvAD in left inferior frontal cortex. bvAD had cortical thinning relative to aAD in prefrontal and anterior temporal regions. All patient groups had lower volumes than controls in both anterior and posterior hippocampus. However, bvAD patients had higher average volume than aAD patients in posterior hippocampus and higher volume than bvFTD patients in anterior hippocampus after adjustment for age and intracranial volume. Conclusion: Findings demonstrated that underlying pathology mediates disease presentation in bvAD and bvFTD.
Collapse
Affiliation(s)
- Sophia Dominguez Perez
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychological Sciences, University of Connecticut, Storrs, CT, USA
| | - Jeffrey S. Phillips
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Catherine Norise
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nikolas G. Kinney
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Prerana Vaddi
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Amy Halpin
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Psychology, University of Maine, Orono, ME, USA
| | - Katya Rascovsky
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - David J. Irwin
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Center for Neurodegenerative Disease Research, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Corey T. McMillan
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Long Xie
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Image Computing and Science Lab & Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura E.M. Wisse
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Image Computing and Science Lab & Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Diagnostic Radiology, Lund University, Lund, Sweden
| | - Paul A. Yushkevich
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Penn Image Computing and Science Lab & Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Dorina Kallogjeri
- Department of Otolaryngology, Washington University, St. Louis, MO, USA
| | - Murray Grossman
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Katheryn A.Q. Cousins
- Penn Frontotemporal Degeneration Center (FTDC), University of Pennsylvania, Philadelphia, PA, USA
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
7
|
Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies. STRESSES 2022. [DOI: 10.3390/stresses2030019] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Hydrogen peroxide (H2O2) is a compound involved in some mammalian reactions and processes. It modulates and signals the redox metabolism of cells by acting as a messenger together with hydrogen sulfide (H2S) and the nitric oxide radical (•NO), activating specific oxidations that determine the metabolic response. The reaction triggered determines cell survival or apoptosis, depending on which downstream metabolic pathways are activated. There are several ways to produce H2O2 in cells, and cellular systems tightly control its concentration. At the cellular level, the accumulation of hydrogen peroxide can trigger inflammation and even apoptosis, and when its concentration in the blood reaches toxic levels, it can lead to bioenergetic failure. This review summarizes existing research from a chemical perspective on the role of H2O2 in various enzymatic pathways and how this biochemistry leads to physiological or pathological responses.
Collapse
|
8
|
Quintero-Villegas A, Valdés-Ferrer SI. Central nervous system effects of 5-HT 7 receptors: a potential target for neurodegenerative diseases. Mol Med 2022; 28:70. [PMID: 35725396 PMCID: PMC9208181 DOI: 10.1186/s10020-022-00497-2] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 06/09/2022] [Indexed: 12/21/2022] Open
Abstract
5-HT7 receptors (5-HT7R) are the most recently identified among the family of serotonin receptors. Their role in health and disease, particularly as mediators of, and druggable targets for, neurodegenerative diseases, is incompletely understood. Unlike other serotonin receptors, for which abundant preclinical and clinical data evaluating their effect on neurodegenerative conditions exist, the available information on the role of the 5-HT7R receptor is limited. In this review, we describe the signaling pathways and cellular mechanisms implicated in the activation of the 5-HT7R; also, we analyze different mechanisms of neurodegeneration and the potential therapeutic implications of pharmacological interventions for 5-HT7R signaling.
Collapse
Affiliation(s)
- Alejandro Quintero-Villegas
- Department of Neurology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico.,Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico
| | - Sergio Iván Valdés-Ferrer
- Department of Neurology, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico. .,Department of Infectious Diseases, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán, Mexico City, Mexico. .,Institute of Bioelectronic Medicine, Feinstein Institutes for Medical Research, Manhasset, NY, USA.
| |
Collapse
|
9
|
Hashimoto M, Ho G, Takamatsu Y, Wada R, Sugama S, Takenouchi T, Waragai M, Masliah E. Possible Role of Amyloid Cross-Seeding in Evolvability and Neurodegenerative Disease. JOURNAL OF PARKINSONS DISEASE 2020; 9:793-802. [PMID: 31524179 PMCID: PMC6839461 DOI: 10.3233/jpd-191675] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Aging-related neurodegenerative disorders are frequently associated with the aggregation of multiple amyloidogenic proteins (APs), although the reason why such detrimental phenomena have emerged in the post-reproductive human brain across evolution is unclear. Speculatively, APs might provide physiological benefits for the human brain during developmental/reproductive stages. Of relevance, it is noteworthy that cross-seeding (CS) of APs has recently been characterized in cellular and animal models of neurodegenerative disease, and that normal physiological CS of multiple APs has also been observed in lower organisms, including yeast and bacteria. In this context, our main objective is to discuss a possible involvement of the CS of APs in promoting evolvability, a hypothetical view regarding the function of APs as an inheritance of acquired characteristics against human brain stressors, which are transgenerationally transmitted to offspring via germ cells. Mechanistically, the protofibrils formed by the CS of multiple APs might confer hormesis more potently than individual APs. By virtue of greater encoded stress information in parental brains being available, the brains of offspring can cope more efficiently with forth-coming stressors. On the other hand, subsequent neurodegeneration caused by APs in parental brain through the antagonistic pleiotropy mechanism in aging, may suggest that synergistically, multiple APs might be more detrimental compared to singular AP in neurodegeneration. Taken together, we suggest that the CS of multiple APs might be involved in both evolvability and neurodegenerative disease in human brain, which may be mechanistically and therapeutically important.
Collapse
Affiliation(s)
- Makoto Hashimoto
- Tokyo Metropolitan Institute of Medical Science, Setagaya-Ku, Tokyo, Japan
| | - Gilbert Ho
- PCND Neuroscience Research Institute, Poway, CA, USA
| | - Yoshiki Takamatsu
- Tokyo Metropolitan Institute of Medical Science, Setagaya-Ku, Tokyo, Japan
| | - Ryoko Wada
- Tokyo Metropolitan Institute of Medical Science, Setagaya-Ku, Tokyo, Japan
| | - Shuei Sugama
- Department of Physiology, Nippon Medical School, Tokyo, Japan
| | - Takato Takenouchi
- Institute of Agrobiological Sciences, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan
| | - Masaaki Waragai
- Tokyo Metropolitan Institute of Medical Science, Setagaya-Ku, Tokyo, Japan
| | - Eliezer Masliah
- Division of Neurosciences, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
10
|
Honma N, Saji S, Mikami T, Yoshimura N, Mori S, Saito Y, Murayama S, Harada N. Estrogen-Related Factors in the Frontal Lobe of Alzheimer's Disease Patients and Importance of Body Mass Index. Sci Rep 2017; 7:726. [PMID: 28389656 PMCID: PMC5429714 DOI: 10.1038/s41598-017-00815-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 03/16/2017] [Indexed: 01/22/2023] Open
Abstract
Estrogens play a physiologically important role in the brain, but controversies exist regarding the association between Alzheimer’s disease (AD) and estrogens. Estrogen-related factors were comprehensively examined in frontal lobe tissues from autopsied AD patients, and compared with controls. Concentrations of estrogens, expression of estrogen receptors (ERs), and estrogen-metabolizing enzymes (EMEs) which are important for determining the peripheral estrogen concentrations, were examined using liquid chromatography tandem mass spectrometry, immunohistochemistry, and quantitative real-time PCR, respectively. Body mass index (BMI), known to correlate with the serum estrogen concentrations, was also taken into consideration. There were no significant differences in estrogen concentrations or each EME level between the two groups in both the cortex and white matter, whereas glial nuclear ER-β expression was significantly lower in white matter from the AD group than the control group (Allred score, 3.2 ± 0.3 and 6.5 ± 0.3, respectively. P < 0.0001). Estrogen concentrations were found to closely correlate with BMI, particularly in controls. ER-β loss in the white matter from the AD group suggests the necessity of studying the effects of estrogens on glias as well as neurons in the etiology of AD. The correlation between BMI and estrogen concentrations in the frontal lobe suggests the importance of non-brain sources of estrogens.
Collapse
Affiliation(s)
- Naoko Honma
- Department of Pathology, Toho University School of Medicine, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan.
| | - Shigehira Saji
- Department of Medical Oncology, Fukushima Medical University, Hikariga-oka 1, Fukushima City, Fukushima, 960-1295, Japan
| | - Tetuo Mikami
- Department of Pathology, Toho University School of Medicine, Omori-Nishi 5-21-16, Ota-ku, Tokyo, 143-8540, Japan
| | - Noriko Yoshimura
- Department of Biochemistry, Fujita Health University School of Medicine, Dengakugakubo 1-98, Kutsukake-cho, Toyoake, 470-1192, Japan
| | - Seijiro Mori
- Department of Internal Medicine, Tokyo Metropolitan Geriatric Hospital, Sakaecho 35-2, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Yuko Saito
- Department of Pathology and Laboratory Medicine, National Center Hospital, National Center of Neurology and Psychiatry, Ogawa-Higashi 4-1-1, Kodaira, Tokyo, 187-8551, Japan
| | - Shigeo Murayama
- Department of Neuropathology, Tokyo Metropolitan Institute of Gerontology, Sakaecho 35-2, Itabashi-ku, Tokyo, 173-0015, Japan
| | - Nobuhiro Harada
- Department of Biochemistry, Fujita Health University School of Medicine, Dengakugakubo 1-98, Kutsukake-cho, Toyoake, 470-1192, Japan
| |
Collapse
|
11
|
Rombouts FJR, Alexander R, Cleiren E, De Groot A, Carpentier M, Dijkmans J, Fierens K, Masure S, Moechars D, Palomino-Schätzlein M, Pineda-Lucena A, Trabanco A, Van Glabbeek D, Vos A, Tresadern G. Fragment Binding to β-Secretase 1 without Catalytic Aspartate Interactions Identified via Orthogonal Screening Approaches. ACS OMEGA 2017; 2:685-697. [PMID: 28626832 PMCID: PMC5472370 DOI: 10.1021/acsomega.6b00482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 02/10/2017] [Indexed: 03/04/2025]
Abstract
An approach to identify β-secretase 1 (BACE1) fragment binders that do not interact with the catalytic aspartate dyad is presented. A ThermoFluor (thermal shift) and a fluorescence resonance energy transfer enzymatic screen on the soluble domain of BACE1, together with a surface plasmon resonance (SPR) screen on the soluble domain of BACE1 and a mutant of one catalytic Asp (D32N), were run in parallel. Fragments that were active in at least two of these assays were further confirmed using one-dimensional NMR (WaterLOGSY) and SPR binding competition studies with peptidic inhibitor OM99-2. Protein-observed NMR (two-dimensional 15N heteronuclear single-quantum coherence spectroscopy) and crystallographic studies with the soluble domain of BACE1 identified a unique and novel binding mode for compound 12, a fragment that still occupies the active site while not making any interactions with catalytic Asps. This novel approach of combining orthogonal fragment screening techniques, for both wild-type and mutant enzymes, as well as binding competition studies could be generalized to other targets to overcome undesired interaction motifs and as a hit-generation approach in highly constrained intellectual property space.
Collapse
Affiliation(s)
- Frederik J. R. Rombouts
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Richard Alexander
- Discovery
Sciences, Janssen Research & Development, 1400 McKean Road, Spring House, Pennsylvania 19477, United States
| | - Erna Cleiren
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Alex De Groot
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Michel Carpentier
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Joyce Dijkmans
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Katleen Fierens
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Stefan Masure
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Diederik Moechars
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Martina Palomino-Schätzlein
- Structural
Biochemistry Laboratory, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Antonio Pineda-Lucena
- Structural
Biochemistry Laboratory, Centro de Investigación Príncipe Felipe, Eduardo Primo Yúfera, 3, 46012 Valencia, Spain
| | - Andrés
A. Trabanco
- Neuroscience
Medicinal Chemistry, Janssen Research &
Development, C/ Jarama
75A, 45007 Toledo, Spain
| | - Daan Van Glabbeek
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Ann Vos
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - Gary Tresadern
- Neuroscience
Medicinal Chemistry, Discovery Sciences, and Neuroscience Biology, Janssen
Research & Development, Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
12
|
Natural products against Alzheimer's disease: Pharmaco-therapeutics and biotechnological interventions. Biotechnol Adv 2016; 35:178-216. [PMID: 28043897 DOI: 10.1016/j.biotechadv.2016.12.005] [Citation(s) in RCA: 154] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Revised: 12/19/2016] [Accepted: 12/23/2016] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is a severe, chronic and progressive neurodegenerative disease associated with memory and cognition impairment ultimately leading to death. It is the commonest reason of dementia in elderly populations mostly affecting beyond the age of 65. The pathogenesis is indicated by accumulation of the amyloid-beta (Aβ) plaques and neurofibrillary tangles (NFT) in brain tissues and hyperphosphorylation of tau protein in neurons. The main cause is considered to be the formation of reactive oxygen species (ROS) due to oxidative stress. The current treatment provides only symptomatic relief by offering temporary palliative therapy which declines the rate of cognitive impairment associated with AD. Inhibition of the enzyme acetylcholinesterase (AChE) is considered as one of the major therapeutic strategies offering only symptomatic relief and moderate disease-modifying effect. Other non-cholinergic therapeutic approaches include antioxidant and vitamin therapy, stem cell therapy, hormonal therapy, use of antihypertensive or lipid-lowering medications and selective phosphodiesterase (PDE) inhibitors, inhibition of β-secretase and γ-secretase and Aβ aggregation, inhibition of tau hyperphosphorylation and intracellular NFT, use of nonsteroidal anti-inflammatory drugs (NSAIDs), transition metal chelators, insulin resistance drugs, etanercept, brain-derived neurotrophic factor (BDNF) etc. Medicinal plants have been reported for possible anti-AD activity in a number of preclinical and clinical trials. Ethnobotany, being popular in China and in the Far East and possibly less emphasized in Europe, plays a substantial role in the discovery of anti-AD agents from botanicals. Chinese Material Medica (CMM) involving Chinese medicinal plants has been used traditionally in China in the treatment of AD. Ayurveda has already provided numerous lead compounds in drug discovery and many of these are also undergoing clinical investigations. A number of medicinal plants either in their crude forms or as isolated compounds have exhibited to reduce the pathological features associated with AD. In this present review, an attempt has been made to elucidate the molecular mode of action of various plant extracts, phytochemicals and traditional herbal formulations investigated against AD as reported in various preclinical and clinical tests. Herbal synergism often found in polyherbal formulations were found effective to combat disease heterogeneity as found in complex pathogenesis of AD. Finally a note has been added to describe biotechnological improvement, genetic and genomic resources and mathematical and statistical techniques for empirical model building associated with anti-AD plant secondary metabolites and their source botanicals.
Collapse
|
13
|
Ciordia M, Pérez-Benito L, Delgado F, Trabanco AA, Tresadern G. Application of Free Energy Perturbation for the Design of BACE1 Inhibitors. J Chem Inf Model 2016; 56:1856-71. [PMID: 27500414 DOI: 10.1021/acs.jcim.6b00220] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Novel spiroaminodihydropyrroles probing for optimized interactions at the P3 pocket of β-secretase 1 (BACE1) were designed with the use of free energy perturbation (FEP) calculations. The resulting molecules showed pIC50 potencies in enzymatic BACE1 inhibition assays ranging from approximately 5 to 7. Good correlation was observed between the predicted activity from the FEP calculations and experimental activity. Simulations run with a default 5 ns approach delivered a mean unsigned error (MUE) between prediction and experiment of 0.58 and 0.91 kcal/mol for retrospective and prospective applications, respectively. With longer simulations of 10 and 20 ns, the MUE was in both cases 0.57 kcal/mol for the retrospective application, and 0.69 and 0.59 kcal/mol for the prospective application. Other considerations that impact the quality of the calculations are discussed. This work provides an example of the value of FEP as a computational tool for drug discovery.
Collapse
Affiliation(s)
- Myriam Ciordia
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain.,Departamento de Química y Bioquímica, Facultad de Farmacia, Universidad CEU San Pablo , Urbanización Montepríncipe Ctra., Boadilla del Monte Km 5.3, 28668 Madrid, Spain
| | - Laura Pérez-Benito
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain.,Laboratori de Medicina Computacional Unitat de Bioestadistica, Facultat de Medicina, Universitat Autonoma de Barcelona , 08193 Bellaterra, Spain
| | - Francisca Delgado
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain
| | - Andrés A Trabanco
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain
| | - Gary Tresadern
- Janssen Research and Development , c/Jarama 75A, 45007 Toledo, Spain
| |
Collapse
|
14
|
Wilson GD, Marples B. A New Use for an Old Treatment: Radiation Therapy and Alzheimer's Disease. Radiat Res 2016; 185:443-8. [PMID: 27092764 DOI: 10.1667/rr14367.1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Affiliation(s)
- George D Wilson
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| | - Brian Marples
- Department of Radiation Oncology, William Beaumont Hospital, Royal Oak, Michigan
| |
Collapse
|
15
|
Minjarez B, Calderón-González KG, Rustarazo MLV, Herrera-Aguirre ME, Labra-Barrios ML, Rincon-Limas DE, Del Pino MMS, Mena R, Luna-Arias JP. Identification of proteins that are differentially expressed in brains with Alzheimer's disease using iTRAQ labeling and tandem mass spectrometry. J Proteomics 2016; 139:103-21. [PMID: 27012543 DOI: 10.1016/j.jprot.2016.03.022] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Revised: 02/26/2016] [Accepted: 03/11/2016] [Indexed: 10/22/2022]
Abstract
UNLABELLED Alzheimer's disease is one of the leading causes of dementia in the elderly. It is considered the result of complex events involving both genetic and environmental factors. To gain further insights into this complexity, we quantitatively analyzed the proteome of cortex region of brains from patients diagnosed with Alzheimer's disease, using a bottom-up proteomics approach. We identified 721 isobaric-tagged polypeptides. From this universe, 61 were found overexpressed and 69 subexpressed in three brains with Alzheimer's disease in comparison to a normal brain. We determined that the most affected processes involving the overexpressed polypeptides corresponded to ROS and stress responses. For the subexpressed polypeptides, the main processes affected were oxidative phosphorylation, organellar acidification and cytoskeleton. We used Drosophila to validate some of the hits, particularly those non-previously described as connected with the disease, such as Sideroflexin and Phosphoglucomutase-1. We manipulated their homolog genes in Drosophila models of Aβ- and Tau-induced pathology. We found proteins that can either modify Aβ toxicity, Tau toxicity or both, suggesting specific interactions with different pathways. This approach illustrates the potential of Drosophila to validate hits after MS studies and suggest that model organisms should be included in the pipeline to identify relevant targets for Alzheimer's disease. BIOLOGICAL SIGNIFICANCE We report a set of differentially expressed proteins in three Alzheimer's disease brains in comparison to a normal brain. Our analyses allowed us to identify that the main affected pathways were ROS and stress responses, oxidative phosphorylation, organellar acidification and cytoskeleton. We validated some identified proteins using genetic models of Amyloid-β and Tau-induced pathology in Drosophila melanogaster. With this approach, Sideroflexin and Phosphoglucomutase-1 were identified as novel proteins connected with Alzheimer's disease.
Collapse
Affiliation(s)
- Benito Minjarez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360 Ciudad de México, México.
| | - Karla Grisel Calderón-González
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360 Ciudad de México, México.
| | - Ma Luz Valero Rustarazo
- Unidad de Proteómica, Centro de Investigación Príncipe Felipe, C/Rambla del Saler 16, 46012 Valencia, España.
| | - María Esther Herrera-Aguirre
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360 Ciudad de México, México.
| | - María Luisa Labra-Barrios
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360 Ciudad de México, México.
| | - Diego E Rincon-Limas
- Department of Neurology, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA; Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL 32611, USA.
| | - Manuel M Sánchez Del Pino
- Unidad de Proteómica, Centro de Investigación Príncipe Felipe, C/Rambla del Saler 16, 46012 Valencia, España.
| | - Raul Mena
- Departamento de Fisiología, Biofísica y Neurociencias, Cinvestav-IPN, Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360 Ciudad de México, México
| | - Juan Pedro Luna-Arias
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav-IPN), Av. Instituto Politécnico Nacional 2508, Col. San Pedro Zacatenco, Gustavo A. Madero, C.P. 07360 Ciudad de México, México.
| |
Collapse
|
16
|
Marples B, McGee M, Callan S, Bowen SE, Thibodeau BJ, Michael DB, Wilson GD, Maddens ME, Fontanesi J, Martinez AA. Cranial irradiation significantly reduces beta amyloid plaques in the brain and improves cognition in a murine model of Alzheimer’s Disease (AD). Radiother Oncol 2016; 118:43-51. [DOI: 10.1016/j.radonc.2015.10.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 09/29/2015] [Accepted: 10/19/2015] [Indexed: 10/22/2022]
|
17
|
Shelkovnikova TA, Ustiugov AA, Kokhan VS, Tarasova TV, Medvedeva VK, Khritankova IV, Bachurin SO, Ninkina NN. [Study into molecular targets of a neuroprotective compound dimebon using a transgenic mice line]. BIOMEDIT︠S︡INSKAI︠A︡ KHIMII︠A︡ 2014; 60:354-63. [PMID: 25019398 DOI: 10.18097/pbmc20146003354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
In the present study we have used a transgenic mice overexpressing an amyloidogenic protein, gamma-synuclein, in the nervous system to address the effect of dimebon on proteinopathy progression. Neuroprotective effect of chronic dimebon administration in these mice at organismal level was confirmed by the increased lifespan. Using histological and biochemical approaches we have demonstrated that dimebon reduced the number of amyloid inclusions in spinal cord of transgenic animals and decreased the content of ubiquitinated proteins in detergent-insoluble fractions. These effects are likely to occur at the level of aggregated protein species, since transgene expression was not altered. Thus, pathological protein aggregation serves as one of dimebon targets in neurodegeneration.
Collapse
|
18
|
Hitz S, Habekost C, Fürst S, Delso G, Förster S, Ziegler S, Nekolla SG, Souvatzoglou M, Beer AJ, Grimmer T, Eiber M, Schwaiger M, Drzezga A. Systematic Comparison of the Performance of Integrated Whole-Body PET/MR Imaging to Conventional PET/CT for ¹⁸F-FDG Brain Imaging in Patients Examined for Suspected Dementia. J Nucl Med 2014; 55:923-31. [PMID: 24833495 DOI: 10.2967/jnumed.113.126813] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2013] [Accepted: 02/10/2014] [Indexed: 12/25/2022] Open
Abstract
UNLABELLED Technologic specifications of recently introduced integrated PET/MR instrumentation, such as MR-based attenuation correction, may particularly affect brain imaging procedures. To evaluate the qualitative performance of PET/MR in clinical neuroimaging, we systematically compared results obtained with integrated PET/MR with conventional PET/CT in the same patients examined for assessment of cognitive impairment. METHODS Thirty patients underwent a single-injection ((18)F-FDG), dual-imaging protocol including PET/CT and integrated PET/MR imaging in randomized order. Attenuation and scatter correction were performed using low-dose CT for the PET/CT and segmented Dixon MR imaging data for the PET/MR. Differences between PET/MR and PET/CT were assessed via region-of-interest (ROI)-based and voxel-based statistical group comparison. Analyses involved attenuation-corrected (AC) and non-attenuation-corrected (NAC) data. Individual PET/MR and PET/CT datasets were compared versus a predefined independent control population, using 3-dimensional stereotactic surface projections. RESULTS Generally, lower measured PET signal values were obtained throughout the brain in ROI-based quantification of the PET signal for PET/MR as compared with PET/CT in AC and NAC data, independently of the scan order. After elimination of global effects, voxel-based and ROI-based group comparison still revealed significantly lower relative tracer signal in PET/MR images in frontoparietal portions of the neocortex but significantly higher relative signal in subcortical and basal regions of the brain than the corresponding PET/CT images of the AC data. In the corresponding NAC images, the discrepancies in frontoparietal portions of the neocortex were diminished, but the subcortical overestimation of tracer intensity by PET/MR persisted. CONCLUSION Considerable region-dependent differences were observed between brain imaging data acquired on the PET/MR, compared with corresponding PET/CT images, in patients evaluated for neurodegenerative disorders. These findings may only in part be explained by inconsistencies in the attenuation-correction procedures. The observed differences may interfere with semiquantitative evaluation and with individual qualitative clinical assessment and they need to be considered, for example, for clinical trials. Improved attenuation-correction algorithms and a PET/MR-specific healthy control database are recommended for reliable and consistent application of PET/MR for clinical neuroimaging.
Collapse
Affiliation(s)
- Stefan Hitz
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Cornelia Habekost
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Sebastian Fürst
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Gaspar Delso
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Stefan Förster
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany TUM Neuroimaging Center (TUM-NIC), Technische Universität München, Munich, Germany
| | - Sibylle Ziegler
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Stephan G Nekolla
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | | | - Ambros J Beer
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Timo Grimmer
- Department of Psychiatry and Psychotherapy, Technische Universität München, Munich, Germany
| | - Matthias Eiber
- Department of Radiology, Technische Universität München, Munich, Germany; and
| | - Markus Schwaiger
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, Technische Universität München, Munich, Germany Department of Nuclear Medicine, University Hospital of Cologne, Cologne, Germany
| |
Collapse
|
19
|
Cai Z. Monoamine oxidase inhibitors: promising therapeutic agents for Alzheimer's disease (Review). Mol Med Rep 2014; 9:1533-41. [PMID: 24626484 DOI: 10.3892/mmr.2014.2040] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2013] [Accepted: 02/10/2014] [Indexed: 11/06/2022] Open
Abstract
Activated monoamine oxidase (MAO) has a critical role in the pathogenesis of Alzheimer's disease (AD), including the formation of amyloid plaques from amyloid β peptide (Aβ) production and accumulation, formation of neurofibrillary tangles, and cognitive impairment via the destruction of cholinergic neurons and disorder of the cholinergic system. Several studies have indicated that MAO inhibitors improve cognitive deficits and reverse Aβ pathology by modulating proteolytic cleavage of amyloid precursor protein and decreasing Aβ protein fragments. Thus, MAO inhibitors may be considered as promising therapeutic agents for AD.
Collapse
Affiliation(s)
- Zhiyou Cai
- Department of Neurology, The Lu'an Affiliated Hospital of Anhui Medical University, Lu'an People's Hospital, Lu'an, Anhui 237005, P.R. China
| |
Collapse
|
20
|
Abstract
AbstractAlzheimer’s disease (AD) is a neurodegenerative disorder that is characterized by normal memory loss and cognitive impairment in humans. Many drug targets and disease-modulating therapies are available for treatment of AD, but none of these are effective enough in reducing problems associated with recognition and memory. Potential drug targets so far reported for AD are β-secretase, Γ-secretase, amyloid beta (Aβ) and Aβ fibrils, glycogen synthase kinase-3 (GSK-3), acyl-coenzyme A: cholesterol acyl-transferase (ACAT) and acetylcholinesterase (AChE). Herbal remedies (antioxidants) and natural metal-chelators have shown a very significant role in reducing the risk of AD, as well as lowering the effect of Aβ in AD patients. Researchers are working in the direction of antisense and stem cell-based therapies for a cure for AD, which mainly depends on the clearance of misfolded protein deposits — including Aβ, tau, and alpha-synuclein. Computational approaches for inhibitor designing, interaction analysis, principal descriptors and an absorption, distribution, metabolism, excretion and toxicity (ADMET) study could speed up the process of drug development with higher efficacy and less chance of failure. This paper reviews the known drugs, drug targets, and existing and future therapies for the treatment of AD.
Collapse
|
21
|
Evidences for B6C3-Tg (APPswe/PSEN1dE9) double-transgenic mice between 3 and 10 months as an age-related Alzheimer's disease model. J Mol Neurosci 2013; 53:370-6. [PMID: 24362678 DOI: 10.1007/s12031-013-0203-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 12/03/2013] [Indexed: 01/30/2023]
Abstract
Transgenic mouse has shown great advantages in the study of Alzheimer's disease (AD) and drug screening as AD develops rapidly resent years, while more detail information of these transgenic mice and experience of application are needed. To obtain the basic background information of the B6C3-Tg (APPswe/PSEN1dE9) double-transgenic mouse, which was reported with early onset AD, three- to ten-month-old B6C3-Tg AD mice and normal C57BL/6 mice were selected randomly to test the ability of learning memory by Morris water maze, the brain acetylcholinesterase (AChE) activity by AChE kit, and beta amyloid protein level by immunohistochemistry staining. Compared with the control group, the escape latency time of B6C3-Tg AD mice at 9 and 10 months of age is significantly longer (P < 0.05) in Morris maze test, and the activity of brain AChE is higher. β-Amyloid plaques were observed at 3 months of age and developed rapidly. Statistical analysis showed a positive correlation between the area of these plaques and the ages of B6C3-Tg AD mouse (y = 0.0355e(0.5557x), R = 0.9557). The model's behavior is conformed to simulate behaviors of human Alzheimer's disease at the early stage and may provide detail background information a new choice when transgenic mice are needed in the research of AD.
Collapse
|
22
|
Lockau H, Jessen F, Fellgiebel A, Drzezga A. Structural and Functional Magnetic Resonance Imaging: Mild Cognitive Impairment and Alzheimer Disease. PET Clin 2013; 8:407-30. [PMID: 27156470 DOI: 10.1016/j.cpet.2013.08.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Magnetic resonance (MR) imaging is playing an increasingly pivotal role in the clinical management of dementia, including Alzheimer disease (AD). In addition to established MR imaging procedures, the introduction of advanced instrumentation such as 7-T MR imaging, as well as novel MR imaging sequences such as arterial spin labeling, MR spectroscopy, diffusion tensor imaging, and resting-state functional MR imaging, may open new pathways toward improved diagnosis of AD even in early stages of disease such as mild cognitive impairment (MCI). This article describes the typical findings of established and new MR imaging procedures in healthy aging, MCI, and AD.
Collapse
Affiliation(s)
- Hannah Lockau
- Department of Radiology, University Hospital Cologne, Kerpener Street 62, Cologne 50937, Germany
| | - Frank Jessen
- Department of Psychiatry, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Sigmund-Freud-Straße 25, Bonn 53105, Germany
| | - Andreas Fellgiebel
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, Untere Zahlbacher Street 8, Mainz 55131, Germany
| | - Alexander Drzezga
- Department of Nuclear Medicine, University Hospital Cologne, Kerpener Street 62, Cologne 50937, Germany.
| |
Collapse
|
23
|
Mandal M, Zhu Z, Cumming JN, Liu X, Strickland C, Mazzola RD, Caldwell JP, Leach P, Grzelak M, Hyde L, Zhang Q, Terracina G, Zhang L, Chen X, Kuvelkar R, Kennedy ME, Favreau L, Cox K, Orth P, Buevich A, Voigt J, Wang H, Kazakevich I, McKittrick BA, Greenlee W, Parker EM, Stamford AW. Design and Validation of Bicyclic Iminopyrimidinones As Beta Amyloid Cleaving Enzyme-1 (BACE1) Inhibitors: Conformational Constraint to Favor a Bioactive Conformation. J Med Chem 2012; 55:9331-45. [DOI: 10.1021/jm301039c] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Mihirbaran Mandal
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jared N. Cumming
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Corey Strickland
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert D. Mazzola
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - John P. Caldwell
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Prescott Leach
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Grzelak
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn Hyde
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xia Chen
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Reshma Kuvelkar
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Matthew E. Kennedy
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Leonard Favreau
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Kathleen Cox
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Peter Orth
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Alexei Buevich
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongwu Wang
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Brian A. McKittrick
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Andrew W. Stamford
- Department
of Medicinal Chemistry, ‡Department of Neuroscience, §Global Structural Chemistry, ∥Department of Analytical
Chemistry, ⊥Department of Basic Pharmaceutical Sciences, and #Department of Exploratory Drug Metabolism, Merck Research Laboratories, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
24
|
Insulin receptor expression and activity in the brains of nondiabetic sporadic Alzheimer's disease cases. Int J Alzheimers Dis 2012; 2012:321280. [PMID: 22666619 PMCID: PMC3362009 DOI: 10.1155/2012/321280] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2011] [Revised: 02/10/2012] [Accepted: 02/13/2012] [Indexed: 11/18/2022] Open
Abstract
We investigated the contents of the insulin receptor-beta subunit (IRβ) and [Tyr1162/1163]-phosphorylated IRβ as surrogate indices of total IR content and IR activation in postmortem hippocampal formation brain specimens from nondiabetic sporadic Alzheimer's disease (AD) cases. We found no significant changes in the brain contents of total IRβ or [Tyr1162/1163]-phosphorylated IRβ, suggesting normal IR content and activation in the brains of nondiabetic sporadic AD cases. Moreover, total IRβ and [Tyr1162/1163]-phosphorylated IRβ levels in the hippocampal formation are not correlated with the severity of amyloid or tau-neuropathology. Exploring the regulation of glycogen synthase kinase 3 (GSK3) α/β, key IR-signaling components, we observed significantly lower levels of total GSK3 α/β in brain specimens from nondiabetic AD cases, suggesting that impaired IR signaling mechanisms might contribute to the onset and/or progression of AD dementia. Outcomes from our study support the development of insulin-sensitizing therapeutic strategies to stimulate downstream IR signaling in nondiabetic AD cases.
Collapse
|
25
|
de la Monte SM. Contributions of brain insulin resistance and deficiency in amyloid-related neurodegeneration in Alzheimer's disease. Drugs 2012; 72:49-66. [PMID: 22191795 PMCID: PMC4550303 DOI: 10.2165/11597760-000000000-00000] [Citation(s) in RCA: 182] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in North America. Growing evidence supports the concept that AD is fundamentally a metabolic disease that results in progressive impairment in the brain's capacity to utilize glucose and respond to insulin and insulin-like growth factor (IGF) stimulation. Moreover, the heterogeneous nature of AD is only partly explained by the brain's propensity to accumulate aberrantly processed, misfolded and aggregated oligomeric structural proteins, including amyloid-β peptides and hyperphosphorylated tau. Evidence suggests that other factors, including impaired energy metabolism, oxidative stress, neuroinflammation, insulin and IGF resistance, and insulin/IGF deficiency in the brain should be incorporated into an overarching hypothesis to develop more realistic diagnostic and therapeutic approaches to AD. In this review, the interrelationship between impaired insulin and IGF signalling and amyloid-β pathology is discussed along with potential therapeutic approaches. Impairments in brain insulin/IGF signalling lead to increased expression of amyloid-β precursor protein (AβPP) and accumulation of AβPP-Aβ. In addition, they promote oxidative stress and deficits in energy metabolism, leading to the activation of pro-AβPP-Aβ-mediated neurodegeneration cascades. Although brain insulin/IGF resistance and deficiency can be induced by primary or secondary disease processes, the soaring rates of peripheral insulin resistance associated with obesity, diabetes mellitus and metabolic syndrome quite likely play major roles in the current AD epidemic. Both clinical and experimental data have linked chronic hyperinsulinaemia to cognitive impairment and neurodegeneration with increased AβPP-Aβ accumulation/reduced clearance in the CNS. Correspondingly, both the restoration of insulin responsiveness and the use of insulin therapy can lead to improved cognitive performance, although with variable effects on brain AβPP-Aβ load. On the other hand, experimental evidence supports the concept that the toxic effects of AβPP-Aβ can promote insulin resistance. Together, these findings suggest that a positive feedback loop of progressive neurodegeneration can develop whereby insulin resistance drives AβPP-Aβ accumulation, and AβPP-Aβ fibril toxicity drives brain insulin resistance. This phenomenon could explain why measuring AβPP-Aβ levels in cerebrospinal fluid or imaging of the brain has proven to be inadequate as a stand-alone biomarker for diagnosing AD, and why the clinical trial results of anti-AβPP-Aβ monotherapy have been disappointing. Instead, the aggregate data suggest that brain insulin resistance and deficiency must also be therapeutically targeted to halt AD progression or reverse its natural course. The positive therapeutic effects of different treatments that address the role of brain insulin/IGF resistance and deficiency, including the use of intranasal insulin delivery, incretins and insulin sensitizer agents are discussed along with potential benefits of lifestyle changes to modify risk for developing mild cognitive impairment or AD. Altogether, the data strongly support the notion that we must shift toward the implementation of multimodal rather than unimodal diagnostic and therapeutic strategies for AD.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Pathology, Rhode Island Hospital and the Warren Alpert Medical School of Brown University, Providence, RI 02903, USA.
| |
Collapse
|
26
|
Characterization of oligomerization-aggregation products of neurodegenerative target proteins by ion mobility mass spectrometry. Methods Mol Biol 2012; 896:399-412. [PMID: 22821540 DOI: 10.1007/978-1-4614-3704-8_27] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Protein amyloidogenesis is generally considered to be a major cause of two most severe neurodegenerative disorders, Parkinson's disease (PD) and Alzheimer's disease (AD). Formation and accumulation of fibrillar aggregates and plaques derived from α-synuclein (α-Syn) and ß-amyloid (Aß) polypeptide in brain have been recognized as characteristics of Parkinson's disease and Alzheimer's disease. Oligomeric aggregates of α-Syn and Aß are considered as neurotoxic intermediate products leading to progressive neurodegeneration. However, molecular details of the oligomerization and aggregation pathway(s) and the molecular structure details are still unclear. We describe here the application of ion-mobility mass spectrometry (IMS-MS) to the identification of α-Syn and Aß oligomerization-aggregation products, and to the characterization of different conformational forms. IMS-MS is an analytical technique capable of separating gaseous ions based on their size, shape, and topography. IMS-MS studies of soluble α-Syn and Aß-aggregates prepared by in vitro incubation over several days were performed on a quadrupole time of flight mass spectrometer equipped with a "travelling wave" ion mobility cell, and revealed the presence of different conformational states and, remarkably, truncation and proteolytic products of high aggregating reactivity. These results suggest that different polypeptide sequences may contribute to the formation of oligomeric aggregates of heterogeneous composition and distinct biochemical properties.
Collapse
|
27
|
de la Monte SM. Therapeutic targets of brain insulin resistance in sporadic Alzheimer's disease. Front Biosci (Elite Ed) 2012. [PMID: 22201977 DOI: 10.2741/482] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Growing evidence supports roles for brain insulin and insulin-like growth factor (IGF) resistance and metabolic dysfunction in the pathogenesis of Alzheimer's disease (AD). Whether the underlying problem stems from a primary disorder of central nervous system (CNS) neurons and glia, or secondary effects of systemic diseases such as obesity, Type 2 diabetes, or metabolic syndrome, the end-results include impaired glucose utilization, mitochondrial dysfunction, increased oxidative stress, neuroinflammation, and the propagation of cascades that result in the accumulation of neurotoxic misfolded, aggregated, and ubiquitinated fibrillar proteins. This article reviews the roles of impaired insulin and IGF signaling to AD-associated neuronal loss, synaptic disconnection, tau hyperphosphorylation, amyloid-beta accumulation, and impaired energy metabolism, and discusses therapeutic strategies and lifestyle approaches that could be used to prevent, delay the onset, or reduce the severity of AD. Finally, it is critical to recognize that AD is heterogeneous and has a clinical course that fully develops over a period of several decades. Therefore, early and multi-modal preventive and treatment approaches should be regarded as essential.
Collapse
Affiliation(s)
- Suzanne M de la Monte
- Department of Neurology, Rhode Island Hospital and Alpert Medical School of Brown University, Providence, RI, USA.
| |
Collapse
|
28
|
Sun ZY, Asberom T, Bara T, Bennett C, Burnett D, Chu I, Clader J, Cohen-Williams M, Cole D, Czarniecki M, Durkin J, Gallo G, Greenlee W, Josien H, Huang X, Hyde L, Jones N, Kazakevich I, Li H, Liu X, Lee J, MacCoss M, Mandal MB, McCracken T, Nomeir A, Mazzola R, Palani A, Parker EM, Pissarnitski DA, Qin J, Song L, Terracina G, Vicarel M, Voigt J, Xu R, Zhang L, Zhang Q, Zhao Z, Zhu X, Zhu Z. Cyclic Hydroxyamidines as Amide Isosteres: Discovery of Oxadiazolines and Oxadiazines as Potent and Highly Efficacious γ-Secretase Modulators in Vivo. J Med Chem 2011; 55:489-502. [DOI: 10.1021/jm201407j] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Zhong-Yue Sun
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Theodros Asberom
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Thomas Bara
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Chad Bennett
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Duane Burnett
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Inhou Chu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - John Clader
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Mary Cohen-Williams
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - David Cole
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Michael Czarniecki
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - James Durkin
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Gioconda Gallo
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - William Greenlee
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hubert Josien
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xianhai Huang
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lynn Hyde
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Nicholas Jones
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Irina Kazakevich
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Hongmei Li
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaoxiang Liu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Julie Lee
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Malcolm MacCoss
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Mihir B. Mandal
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Troy McCracken
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Amin Nomeir
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Robert Mazzola
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Anandan Palani
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Eric M. Parker
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Dmitri A. Pissarnitski
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Jun Qin
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lixin Song
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Giuseppe Terracina
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Monica Vicarel
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Johannes Voigt
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Ruo Xu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Lili Zhang
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Qi Zhang
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhiqiang Zhao
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Xiaohong Zhu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| | - Zhaoning Zhu
- Department
of Medicinal Chemistry, §Department of Neuroscience, ∥Department of Structural Chemistry, ⊥Department of Pharmaceutical
Sciences, and #Department of Drug Metabolism and Pharmacokinetics, Schering Plough Research Institute, 2015 Galloping
Hill Road, Kenilworth, New Jersey 07033, United States
| |
Collapse
|
29
|
Montgomery SL, Mastrangelo MA, Habib D, Narrow WC, Knowlden SA, Wright TW, Bowers WJ. Ablation of TNF-RI/RII expression in Alzheimer's disease mice leads to an unexpected enhancement of pathology: implications for chronic pan-TNF-α suppressive therapeutic strategies in the brain. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2053-70. [PMID: 21835156 DOI: 10.1016/j.ajpath.2011.07.001] [Citation(s) in RCA: 77] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Received: 04/06/2011] [Revised: 06/28/2011] [Accepted: 07/01/2011] [Indexed: 11/15/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by severe memory loss and cognitive impairment. Neuroinflammation, including the extensive production of pro-inflammatory molecules and the activation of microglia, has been implicated in the disease process. Tumor necrosis factor (TNF)-α, a prototypic pro-inflammatory cytokine, is elevated in AD, is neurotoxic, and colocalizes with amyloid plaques in AD animal models and human brains. We previously demonstrated that the expression of TNF-α is increased in AD mice at ages preceding the development of hallmark amyloid and tau pathological features and that long-term expression of this cytokine in these mice leads to marked neuronal death. Such observations suggest that TNF-α signaling promotes AD pathogenesis and that therapeutics suppressing this cytokine's activity may be beneficial. To dissect TNF-α receptor signaling requirements in AD, we generated triple-transgenic AD mice (3xTg-AD) lacking both TNF-α receptor 1 (TNF-RI) and 2 (TNF-RII), 3xTg-ADxTNF-RI/RII knock out, the cognate receptors of TNF-α. These mice exhibit enhanced amyloid and tau-related pathological features by the age of 15 months, in stark contrast to age-matched 3xTg-AD counterparts. Moreover, 3xTg-ADxTNF-RI/RII knock out-derived primary microglia reveal reduced amyloid-β phagocytic marker expression and phagocytosis activity, indicating that intact TNF-α receptor signaling is critical for microglial-mediated uptake of extracellular amyloid-β peptide pools. Overall, our results demonstrate that globally ablated TNF receptor signaling exacerbates pathogenesis and argues against long-term use of pan-anti-TNF-α inhibitors for the treatment of AD.
Collapse
MESH Headings
- Adaptor Proteins, Signal Transducing/metabolism
- Aging/pathology
- Alzheimer Disease/drug therapy
- Alzheimer Disease/metabolism
- Alzheimer Disease/pathology
- Alzheimer Disease/physiopathology
- Amyloid/metabolism
- Amyloid beta-Protein Precursor/genetics
- Animals
- Brain/metabolism
- Brain/pathology
- CA1 Region, Hippocampal/pathology
- CA1 Region, Hippocampal/physiopathology
- CA3 Region, Hippocampal/pathology
- CA3 Region, Hippocampal/physiopathology
- Calcium-Binding Proteins/metabolism
- Crosses, Genetic
- Female
- Humans
- Lipopolysaccharide Receptors/metabolism
- Long-Term Potentiation
- Male
- Mice
- Mice, Knockout
- Microfilament Proteins/metabolism
- Microglia/metabolism
- Microglia/pathology
- Phagocytosis
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type II/deficiency
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Synapses/metabolism
- Transgenes/genetics
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/metabolism
- tau Proteins/genetics
Collapse
Affiliation(s)
- Sara L Montgomery
- Department of Pathology and Laboratory Medicine, University of Rochester Medical Center, Rochester, NY14642, USA.
| | | | | | | | | | | | | |
Collapse
|
30
|
León R, Garcia AG, Marco-Contelles J. Recent advances in the multitarget-directed ligands approach for the treatment of Alzheimer's disease. Med Res Rev 2011; 33:139-89. [PMID: 21793014 DOI: 10.1002/med.20248] [Citation(s) in RCA: 354] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
With 27 million cases worldwide documented in 2006, Alzheimer's disease (AD) constitutes an overwhelming health, social, economic, and political problem to nations. Unless a new medicine capable to delay disease progression is found, the number of cases will reach 107 million in 2050. So far, the therapeutic paradigm one-compound-one-target has failed. This could be due to the multiple pathogenic mechanisms involved in AD including amyloid β (Aβ) aggregation to form plaques, τ hyperphosphorylation to disrupt microtubule to form neurofibrillary tangles, calcium imbalance, enhanced oxidative stress, impaired mitochondrial function, apoptotic neuronal death, and deterioration of synaptic transmission, particularly at cholinergic neurons. Approximately 100 compounds are presently been investigated directed to single targets, namely inhibitors of β and γ secretase, vaccines or antibodies that clear Aβ, metal chelators to inhibit Aβ aggregation, blockers of glycogen synthase kinase 3β, enhancers of mitochondrial function, antioxidants, modulators of calcium-permeable channels such as voltage-dependent calcium channels, N-methyl-D-aspartate receptors for glutamate, or enhancers of cholinergic neurotransmission such as inhibitors of acetylcholinesterase or butyrylcholinesterase. In view of this complex pathogenic mechanisms, and the successful treatment of chronic diseases such as HIV or cancer, with multiple drugs having complementary mechanisms of action, the concern is growing that AD could better be treated with a single compound targeting two or more of the pathogenic mechanisms leading to neuronal death. This review summarizes the current therapeutic strategies based on the paradigm one-compound-various targets to treat AD. A treatment that delays disease onset and/or progression by 5 years could halve the number of people requiring institutionalization and/or dying from AD.
Collapse
Affiliation(s)
- Rafael León
- Department of Chemistry, University of Cambridge, Cambridge, Lensfield road, Cambridge CB2 1EW, United Kingdom.
| | | | | |
Collapse
|
31
|
Fuentealba J, Dibarrart AJ, Fuentes-Fuentes MC, Saez-Orellana F, Quiñones K, Guzmán L, Perez C, Becerra J, Aguayo LG. Synaptic failure and adenosine triphosphate imbalance induced by amyloid-β aggregates are prevented by blueberry-enriched polyphenols extract. J Neurosci Res 2011; 89:1499-508. [PMID: 21647937 DOI: 10.1002/jnr.22679] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2010] [Revised: 03/23/2011] [Accepted: 03/31/2011] [Indexed: 12/29/2022]
Abstract
The potential neuroprotective properties of fruits have been widely recognized. In this study, we evaluated the protective properties of a blueberry extract (BB-4), rich in polyphenols, in a neurodegenerative model induced by amyloid-β peptide (Aβ). Chronic treatment with Aβ drastically reduced synaptic transmission and the extent of secretory vesicles, which were recovered partially with BB-4. Also, the extract recovered Ca(2+) transients in hippocampal neurons preincubated with Aβ (0.5 and 5 μM) by about 25% ± 3% and 30% ± 2, respectively. In this work, we demonstrate a novel effect of the BB-4 extract on Aβ-induced ATP leakage, in which this extract was able to antagonize the acute ATP leakage but not chronic ATP depletion. On the other hand, BB-4 prevented the uncoupling of mitochondrial function induced by FCCP by about 85%, but it was unable to modify the uncoupling induced by Aβ. The present results strongly indicate that BB-4 plays a role in the process of Aβ aggregation by reducing the toxic species (i.e., 40 kDa). These findings suggest that a blueberry extract can protect neuronal tissue from Aβ toxicity mainly through its antiaggregation property, and its antioxidant properties and mitochondrial membrane potential capacities are secondary mechanisms important in chronic stages. Our work suggests that BB-4 could be an important nutritional complement to neuronal health as well as a potential nutraceutical formulation useful as a dietary supplement in the elderly.
Collapse
Affiliation(s)
- Jorge Fuentealba
- Neuroactive Drugs Screening Unit, Department of Physiology, Faculty of Biological Sciences, University of Concepción, Concepcion, Chile.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Kim JH, Ee SM, Jittiwat J, Ong ES, Farooqui AA, Jenner AM, Ong WY. Increased expression of acyl-coenzyme A: cholesterol acyltransferase-1 and elevated cholesteryl esters in the hippocampus after excitotoxic injury. Neuroscience 2011; 185:125-34. [PMID: 21514367 DOI: 10.1016/j.neuroscience.2011.04.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2010] [Revised: 04/05/2011] [Accepted: 04/08/2011] [Indexed: 10/18/2022]
Abstract
Significant increases in levels of cholesterol and cholesterol oxidation products are detected in the hippocampus undergoing degeneration after excitotoxicity induced by the potent glutamate analog, kainate (KA), but until now, it is unclear whether the cholesterol is in the free or esterified form. The present study was carried out to examine the expression of the enzyme involved in cholesteryl ester biosynthesis, acyl-coenzyme A: cholesterol acyltransferase (ACAT) and cholesteryl esters after KA excitotoxicity. A 1000-fold greater basal mRNA level of ACAT1 than ACAT2 was detected in the normal brain. ACAT1 mRNA and protein were upregulated in the hippocampus at 1 and 2 weeks after KA injections, at a time of glial reaction. Immunohistochemistry showed ACAT1 labeling of oligodendrocytes in the white matter and axon terminals in hippocampal CA fields of normal rats, and loss of staining in neurons but increased immunoreactivity of oligodendrocytes, in areas affected by KA. Gas chromatography-mass spectrometry analyses confirmed previous observations of a marked increase in level of total cholesterol and cholesterol oxidation products, whilst nuclear magnetic resonance spectroscopy showed significant increases in cholesteryl ester species in the degenerating hippocampus. Upregulation of ACAT1 expression was detected in OLN93 oligodendrocytes after KA treatment, and increased expression was prevented by an antioxidant or free radical scavenger in vitro. This suggests that ACAT1 expression may be induced by oxidative stress. Together, our results show elevated ACAT1 expression and increased cholesteryl esters after KA excitotoxicity. Further studies are necessary to determine a possible role of ACAT1 in acute and chronic neurodegenerative diseases.
Collapse
Affiliation(s)
- J-H Kim
- Department of Anatomy, National University of Singapore, Singapore 119260
| | | | | | | | | | | | | |
Collapse
|
33
|
Abstract
The hippocampus is a vulnerable and plastic brain structure that is damaged by a variety of stimuli, e.g. hypoxia, hypoperfusion, hypoglycaemia, stress and seizures. Alzheimer's disease is a common and important disorder in which hippocampal atrophy is reported. Indeed, the available evidence suggests that hippocampal atrophy is the starting point of the pathogenesis of Alzheimer's disease and a significant number of patients with hippocampal atrophy will develop Alzheimer's disease. Studies indicate that hippocampal atrophy has functional consequences, e.g. cognitive impairment. Deposition of tau protein, formation of neurofibrillary tangles and accumulation of β-amyloid (Aβ) contributes to hippocampal atrophy together with damage caused by several other factors. Some of the factors associated with the development of hippocampal atrophy in Alzheimer's disease have been identified, e.g. hypertension, diabetes mellitus, hyperlipidaemia, seizures, affective disturbances and stress, and more is being learnt about other factors. Hypertension can potentially damage the hippocampus through ischaemia caused by atherosclerosis and cerebral amyloid angiopathy. Diabetes can produce hippocampal lesions via both vascular and non-vascular pathologies and can reduce the threshold for hippocampal damage. Carriers of the apolipoprotein E (ApoE)-ε4 genotype have been shown to have greater mesial temporal atrophy and poorer memory functions than non-carriers. In addition to giving rise to abnormal lipid metabolism, the ApoE-ε4 allele can affect the course of Alzheimer's disease via both Aβ-dependent and -independent pathways. Repetitive seizures can increase Aβ-peptide production and cause neurotransmission dysfunction and cytoskeletal abnormalities or a combination of these. Affective disturbances and stress are proposed to increase corticosteroid-induced hippocampal damage in many different ways. In the absence of any specific markers for predicting Alzheimer's disease progression, it seems appropriate to learn more about the various predictors of hippocampal atrophy that determine the progression of Alzheimer's disease from mild cognitive impairment (MCI), and then attempt to address these. It would be interesting to know to what extent these predictors play a role in the development of MCI or hasten the conversion of MCI to full-blown Alzheimer's disease. Finally, it would be useful to know the extent to which these predictors can worsen or aggravate existing Alzheimer's disease. Of the clinically used drugs in Alzheimer's disease, anticholinesterases have been shown to slow down the rate of progression of hippocampal atrophy. One study observed that the neuroprotective effect of these agents is possibly due to an anti-Aβ effect produced by cholinergic stimulation. Similarly, antihypertensive and antihyperglycaemic drugs (pioglitazone and insulin) have been shown to reduce the risk of Alzheimer's disease or disease progression. Currently, there are no disease-modifying therapies available for Alzheimer's disease. It has been suggested that for treatment to be most effective, the regimen must be started before significant downstream damage has occurred (i.e. before the clinical diagnosis of Alzheimer's disease, at the stage of MCI or earlier). Since the hippocampus is a plastic structure and atrophy of this structure is closely related to the pathophysiology of Alzheimer's disease, if we could control blood pressure, regulate blood sugar, treat behavioural and psychological symptoms, achieve satisfactory lipid lowering and maintain a seizure-free state in patients with Alzheimer's disease, this may not only improve disease control but could also potentially affect the rate of disease progression.
Collapse
Affiliation(s)
- Vikas Dhikav
- Department of Pharmacology, All India Institute of Medical Sciences, New Delhi, India.
| | | |
Collapse
|
34
|
Forette F, Hauw JJ. [Treatment of Alzheimer's disease and future approaches]. Therapie 2010; 65:429-37. [PMID: 21144478 DOI: 10.2515/therapie/2010055] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2010] [Accepted: 07/15/2010] [Indexed: 01/28/2023]
Abstract
The progressive neuronal loss in Alzheimer's disease leads to neurochemical abnormalities which provide the basis for symptomatic treatments. Four cholinesterase inhibitors were released in this indication. Meta-analyses have confirmed a beneficial effect on cognitive functioning and activities of daily living. The NMDA receptor antagonist, memantine, was also approved for the treatment of moderate to severe and may be associated. Progress in the patho-physiology of the disease offers some hope of new treatments acting on the cerebral lesions. The amyloid hypothesis allowed the emergence of active or passive immunotherapies, and of secretase inhibitors or modulators. Recent studies have targeted the P tau protein. The brain plasticity and the uses of stem cells offer more distant hope.
Collapse
Affiliation(s)
- Françoise Forette
- AP-HP, Hôpital Broca, Université Paris V, Fondation Nationale de Gérontologie-ILC, Paris, France.
| | | |
Collapse
|
35
|
Panza F, Frisardi V, Imbimbo BP, D’Onofrio G, Pietrarossa G, Seripa D, Pilotto A, Solfrizzi V. Bapineuzumab: anti-β-amyloid monoclonal antibodies for the treatment of Alzheimer’s disease. Immunotherapy 2010; 2:767-82. [DOI: 10.2217/imt.10.80] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In the last decade, new therapeutic approaches targeting β-amyloid (Aβ) have been discovered and developed with the hope of modifying the natural history of Alzheimer’s disease (AD). The most revolutionary of these approaches consists in the removal of brain Aβ via anti-Aβ antibodies. After an active vaccine (AN1792) was discontinued in 2002 due to occurrence of meningoencephalitis in approximately 6% of patients, several other second-generation active Aβ vaccines and passive Aβ immunotherapies have been developed and are under clinical investigation with the aim of accelerating Aβ clearance from the brain of AD patients. The most advanced of these immunological approaches is bapineuzumab, which is composed of humanized anti-Aβ monoclonal antibodies that has been tested in two Phase II trials. Bapineuzumab has been shown to reduce Aβ burden in the brain of AD patients. However, its preliminary cognitive efficacy appears uncertain, particularly in ApoE ε4 carriers, and vasogenic edema may limit its clinical use. The results of four ongoing large Phase III trials on bapineuzumab will provide answers regarding whether passive anti-Aβ immunization is able to alter the course of this devastating disease.
Collapse
Affiliation(s)
| | - Vincenza Frisardi
- Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, Bari, Italy
| | - Bruno P Imbimbo
- Research & Development Department, Chiesi Farmaceutici, Parma, Italy
| | - Grazia D’Onofrio
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | | | - Davide Seripa
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Alberto Pilotto
- Geriatric Unit & Gerontology-Geriatric Research Laboratory, IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy
| | - Vincenzo Solfrizzi
- Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, Bari, Italy
| |
Collapse
|
36
|
Mathura VS, Patel N, Bachmeier C, Mullan M, Paris D. A 3D-QSAR model based screen for dihydropyridine-like compound library to identify inhibitors of amyloid beta (Aβ) production. Bioinformation 2010; 5:122-7. [PMID: 21364791 PMCID: PMC3041004 DOI: 10.6026/97320630005122] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2010] [Accepted: 08/11/2010] [Indexed: 12/28/2022] Open
Abstract
Abnormal accumulation of amyloid beta peptide (Aβ) is one of the hallmarks of Alzheimer's disease progression. Practical limitations such as cost , poor hit
rates and a lack of well characterized targets are a major bottle neck in the in vitro screening of a large number of chemical libraries and profiling them to
identify Aβ inhibitors. We used a limited set of 1,4 dihydropyridine (DHP)-like compounds from our model set (MS) of 24 compounds which inhibit Aβ as a
training set and built 3D-QSAR (Three-dimensional Quantitative Structure-Activity Relationship) models using the Phase program (SchrÖdinger, USA). We
developed a 3D-QSAR model that showed the best prediction for Aβ inhibition in the test set of compounds and used this model to screen a 1,043 DHP-like
small library set of (LS) compounds. We found that our model can effectively predict potent hits at a very high rate and result in significant cost savings when
screening larger libraries. We describe here our in silico model building strategy, model selection parameters and the chemical features that are useful for
successful screening of DHP and DHP-like chemical libraries for Aβ inhibitors.
Collapse
|
37
|
Toda N, Kaneko T, Kogen H. Development of an efficient therapeutic agent for Alzheimer's disease: design and synthesis of dual inhibitors of acetylcholinesterase and serotonin transporter. Chem Pharm Bull (Tokyo) 2010; 58:273-87. [PMID: 20190429 DOI: 10.1248/cpb.58.273] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
To date, acetylcholinesterase (AChE) inhibitors have been clinically effective drugs for the palliative treatment of Alzheimer's disease, but their clinical efficacy is limited, mainly due to their adverse effects on peripheral organs. Since patients of Alzheimer's disease often exhibit depression as well as memory impairment, dual inhibitors of AChE and serotonin transporter (SERT) would be a better therapeutic method. Anti-depressive effects based on SERT inhibition would reduce the dose-related side effects of AChE inhibitors. Such dual inhibitors were designed by the hybridization of rivastigmine and fluoxetine based on a hypothetical model of the AChE active site. Various derivatives were synthesized and evaluated for their in vitro inhibition, and then (S)-5j (RS-1259), which possessed balanced inhibitory activities of AChE (IC(50)=101 nM) and SERT (IC(50)=42 nM), was successfully obtained. An ex vivo experiment in mice indicated that (S)-5j (RS-1259) simultaneously inhibited AChE and SERT in the brain following an oral administration. The simultaneous elevation of extracellular levels of acetylcholine and serotonin in the rat hippocampus was actually confirmed by microdialysis.
Collapse
Affiliation(s)
- Narihiro Toda
- R&D Division, Daiichi Sankyo Co., Shinagawa-ku, Tokyo, Japan
| | | | | |
Collapse
|
38
|
Abstract
Presenilins form the catalytic part of the gamma-secretases, protein complexes that are responsible for the intramembranous cleavage of transmembrane proteins. The presenilins are involved in several biological functions, but are best known for their role in the generation of the beta-amyloid (Abeta) peptide in Alzheimer's disease and are therefore thought to be important drug targets for this disorder. Mutations in the presenilin genes cause early-onset familial Alzheimer's disease, but mutation carriers have substantial phenotypic heterogeneity. Recent evidence implicating presenilin mutations in non-Alzheimer's dementias, including frontotemporal dementia and Lewy body dementia, warrants further investigation. An increased understanding of the diversity of the molecular cell biology of the gamma-secretase complex and the effects of clinical mutations in the presenilin genes might help pave the way for improved development of drugs that are designed to target gamma-secretase enzymatic activity in Alzheimer's disease and potentially in other neurological diseases.
Collapse
Affiliation(s)
- Bruno A Bergmans
- Department of Molecular and Developmental Genetics, VIB, Leuven, Belgium; Center for Human Genetics, Katholieke Universiteit Leuven, Leuven, Belgium
| | | |
Collapse
|
39
|
Lewczuk P, Kamrowski-Kruck H, Peters O, Heuser I, Jessen F, Popp J, Bürger K, Hampel H, Frölich L, Wolf S, Prinz B, Jahn H, Luckhaus C, Perneczky R, Hüll M, Schröder J, Kessler H, Pantel J, Gertz HJ, Klafki HW, Kölsch H, Reulbach U, Esselmann H, Maler JM, Bibl M, Kornhuber J, Wiltfang J. Soluble amyloid precursor proteins in the cerebrospinal fluid as novel potential biomarkers of Alzheimer's disease: a multicenter study. Mol Psychiatry 2010; 15:138-45. [PMID: 18663368 DOI: 10.1038/mp.2008.84] [Citation(s) in RCA: 121] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In this report, we present the results of a multicenter study to test analytic and diagnostic performance of soluble forms of amyloid precursor proteins alpha and beta (sAPP alpha and sAPP beta) in the cerebrospinal fluid (CSF) of patients with different forms of dementing conditions. CSF samples were collected from 188 patients with early dementia (mini-mental state examination >or=20 in majority of cases) and mild cognitive impairment (MCI) in 12 gerontopsychiatric centers, and the clinical diagnoses were supported by neurochemical dementia diagnostic (NDD) tools: CSF amyloid beta peptides, Tau and phospho-Tau. sAPP alpha and sAPP beta were measured with multiplexing method based on electrochemiluminescence. sAPP alpha and sAPP beta CSF concentrations correlated with each other with very high correlation ratio (R=0.96, P<0.001). We observed highly significantly increased sAPP alpha and sAPP beta CSF concentrations in patients with NDD characteristic for Alzheimer's disease (AD) compared to those with NDD negative results. sAPP alpha and sAPP beta highly significantly separated patients with AD, whose diagnosis was supported by NDD findings (sAPP alpha: cutoff, 117.4 ng ml(-1), sensitivity, 68%, specificity, 85%, P<0.001; sAPP beta: cutoff, 181.8 ng ml(-1), sensitivity, 75%, specificity, 85%, P<0.001), from the patients clinically assessed as having other dementias and supported by NDD untypical for AD. We conclude sAPP alpha and sAPP beta might be regarded as novel promising biomarkers supporting the clinical diagnosis of AD.
Collapse
Affiliation(s)
- P Lewczuk
- Department of Psychiatry and Psychotherapy, University of Erlangen-Nuremberg, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Abstract
Cerebrolysin is a parenterally administered, porcine brain-derived peptide preparation that has pharmacodynamic properties similar to those of endogenous neurotrophic factors. In several randomized, double-blind trials of up to 28 weeks' duration in patients with Alzheimer's disease, Cerebrolysin was superior to placebo in improving global outcome measures and cognitive ability. A large, randomized comparison of Cerebrolysin, donepezil or combination therapy showed beneficial effects on global measures and cognition for all three treatment groups compared with baseline. Although not as extensively studied in patients with vascular dementia, Cerebrolysin has also shown beneficial effects on global measures and cognition in this patient population. Cerebrolysin was generally well tolerated in clinical trials, with dizziness (or vertigo) being the most frequently reported adverse event. Although further studies with Cerebrolysin, including longer term trials and further exploration of its use in combination with cholinesterase inhibitors, are needed to more clearly determine its place in the management of Alzheimer's disease and vascular dementia, available data suggest that Cerebrolysin is a useful addition to the treatment options available for dementia.
Collapse
|
41
|
Drzezga A. Amyloid-plaque imaging in early and differential diagnosis of dementia. Ann Nucl Med 2010; 24:55-66. [DOI: 10.1007/s12149-009-0330-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2009] [Accepted: 11/20/2009] [Indexed: 12/12/2022]
|
42
|
Panza F, Solfrizzi V, Frisardi V, Capurso C, D'Introno A, Colacicco AM, Vendemiale G, Capurso A, Imbimbo BP. Disease-modifying approach to the treatment of Alzheimer's disease: from alpha-secretase activators to gamma-secretase inhibitors and modulators. Drugs Aging 2010; 26:537-55. [PMID: 19655822 DOI: 10.2165/11315770-000000000-00000] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In the last decade, advances in understanding the neurobiology of Alzheimer's disease (AD) have translated into an increase in clinical trials assessing various potential AD treatments. At present, drugs used for the treatment of AD only slightly delay the inevitable symptomatic progression of the disease and do not affect the main neuropathological hallmarks of the disease, i.e. senile plaques and neurofibrillary tangles. Brain accumulation of oligomeric species of beta-amyloid (A beta) peptides, the principal components of senile plaques, is believed to play a crucial role in the development of AD. Based on this hypothesis, huge efforts are being made to identify drugs able to interfere with proteases regulating A beta formation from amyloid precursor protein (APP). Compounds that stimulate alpha-secretase, the enzyme responsible for non-amyloidogenic metabolism of APP, are being developed and one of these, EHT-0202, has recently commenced evaluation in a phase II study. The discovery of inhibitors of beta-secretase (memapsin-2, beta-amyloid cleaving enzyme-1 [BACE-1]), the enzyme that regulates the first step of amyloidogenic APP metabolism, has proved to be particularly difficult because of inherent medicinal chemistry issues and only one compound (CTS-21166) has proceeded to clinical testing. Conversely, several compounds that inhibit gamma-secretase, the pivotal enzyme that generates A beta, have been identified, the most advanced being LY-450139 (semagacestat), presently in phase III clinical development. There has been considerable disappointment over the failure of a phase III study of tarenflurbil, a compound believed to modulate the activity of gamma-secretase, after encouraging phase II findings. Nevertheless, other promising gamma-secretase modulators are being developed and are approaching clinical testing. All these therapeutic approaches increase the hope of slowing the rate of decline in patients with AD and modifying the natural history of this devastating disease within the next 5 years.
Collapse
Affiliation(s)
- Francesco Panza
- Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, Bari, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Liu GT, Volpe NJ, Galetta SL. Disorders of higher cortical visual function. Neuroophthalmology 2010. [DOI: 10.1016/b978-1-4160-2311-1.00009-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
44
|
Panza F, Solfrizzi V, Frisardi V, Imbimbo BP, Capurso C, D'Introno A, Colacicco AM, Seripa D, Vendemiale G, Capurso A, Pilotto A. Beyond the neurotransmitter-focused approach in treating Alzheimer's disease: drugs targeting beta-amyloid and tau protein. Aging Clin Exp Res 2009; 21:386-406. [PMID: 20154508 DOI: 10.1007/bf03327445] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Drugs currently used to treat Alzheimer's Disease (AD) have limited therapeutic value and do not affect the main neuropathological hallmarks of the disease, i.e., senile plaques and neurofibrillar tangles. Senile plaques are mainly formed of beta-amyloid (Abeta), a 42-aminoacid peptide. Neurofibrillar tangles are composed of paired helical filaments of hyperphosphorylated tau protein. New, potentially disease-modifying, therapeutic approaches are targeting Abeta and tau protein. Drugs directed against Abeta include active and passive immunization, that have been found to accelerate Abeta clearance from the brain. The most developmentally advanced monoclonal antibody directly targeting Abeta is bapineuzumab, now being studied in a large Phase III clinical trial. Compounds that interfere with proteases regulating Abeta formation from amyloid precursor protein (APP) are also actively pursued. The discovery of inhibitors of beta-secretase, the enzyme that regulates the first step of the amyloidogenic metabolism of APP, has been revealed to be particularly difficult due to inherent medicinal chemistry problems, and only one compound (CTS-21166) has reached clinical testing. Conversely, several compounds that inhibit gamma-secretase, the pivotal enzyme that generates Abeta, have been identified, the most advanced being LY-450139 (semagacestat), now in Phase III clinical development. Compounds that stimulate alpha-secretase, the enzyme responsible for the non-amyloidogenic metabolism of APP, are also being developed, and one of them, EHT-0202, has recently entered Phase II testing. Potent inhibitors of Abeta aggregation have also been identified, and one of such compounds, PBT-2, has provided encouraging neuropsychological results in a recently completed Phase II study. Therapeutic approaches directed against tau protein include inhibitors of glycogen synthase kinase- 3 (GSK-3), the enzyme responsible for tau phosphorylation and tau protein aggregation inhibitors. NP-12, a promising GSK-3 inhibitor, is being tested in a Phase II study, and methylthioninium chloride, a tau protein aggregation inhibitor, has given initial encouraging results in a 50-week study. With all these approaches on their way, the hope for disease-modifying therapy in this devastating disease may become a reality in the next 5 years.
Collapse
Affiliation(s)
- Francesco Panza
- Department of Geriatrics, Center for Aging Brain, Memory Unit, University of Bari, 70124, Bari, Italy.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Promising multifunctional anti-Alzheimer's dimer bis(7)-Cognitin acting as an activator of protein kinase C regulates activities of α-secretase and BACE-1 concurrently. Eur J Pharmacol 2009; 623:14-21. [DOI: 10.1016/j.ejphar.2009.09.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2009] [Revised: 08/26/2009] [Accepted: 09/08/2009] [Indexed: 11/19/2022]
|
46
|
Structural characterization of ß-amyloid oligomer-aggregates by ion mobility mass spectrometry and electron spin resonance spectroscopy. Anal Bioanal Chem 2009; 395:2509-19. [DOI: 10.1007/s00216-009-3164-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 09/15/2009] [Accepted: 09/16/2009] [Indexed: 11/30/2022]
|
47
|
Kontsekova E, Ivanovova N, Handzusova M, Novak M. Chaperone-like antibodies in neurodegenerative tauopathies: implication for immunotherapy. Cell Mol Neurobiol 2009; 29:793-8. [PMID: 19214739 PMCID: PMC11506202 DOI: 10.1007/s10571-009-9355-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2008] [Accepted: 01/22/2009] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD) belongs to the category of neurodegenerative tauopathies, which are characterized by intracellular and extracellular accumulation of misfolded tau. Structurally, tau belongs to the family of the intrinsically disordered proteins that are characterized by the absence of well-defined three-dimensional structure of the free protein. In the course of neurodegeneration, intrinsically disordered tau protein gains highly ordered misfolded structure. Currently it is widely accepted that misfolded tau proteins represent viable drug target for prospective therapeutic development. Until now several therapeutic approaches targeting misfolded tau were developed. Monoclonal antibodies with chaperone-like activities that would be able to neutralize the toxic gain of function of misfolded tau represent novel promising immunological concept in the treatment of AD. We suggest that antibodies as specific chaperones targeting misfolded proteins may serve as potent therapeutic drugs of AD as well as others conformational diseases.
Collapse
Affiliation(s)
- Eva Kontsekova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10 Slovak Republic
| | - Natalia Ivanovova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10 Slovak Republic
| | - Martina Handzusova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10 Slovak Republic
| | - Michal Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10 Slovak Republic
- Axon Neuroscience, Renweg 95b, Vienna, 1030 Austria
| |
Collapse
|
48
|
Kluger BM, Saunders LV, Hou W, Garvan CW, Kirli S, Efros DB, Chau QAN, Crucian GP, Finney GR, Meador KJ, Heilman KM. A brief computerized self-screen for dementia. J Clin Exp Neuropsychol 2008; 31:234-44. [PMID: 19051092 DOI: 10.1080/13803390802317559] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Among his many contributions to the field of neuropsychology, Arthur Benton recognized the broad public health significance and unique ability of focused neuropsychological tests to screen for dementia. The need for validated screening tests for the presence of dementia will continue to grow as the cumulative prevalence of dementia grows and as our ability to treat or slow the progression of these diseases improves. We have developed a brief, self-administered computerized screening test for dementia, which is user friendly to the majority of elderly participants, including those with dementia. This test demonstrates comparable discriminant validity to the Mini Mental State Examination (MMSE), and its subscales correlate well with the traditional neuropsychological tests upon which it is based. We discuss its relative merits and limitations in comparison to other current instruments as well as suggesting future directions for this field.
Collapse
|
49
|
Turner LN, Balasubramaniam R, Hersh EV, Stoopler ET. Drug therapy in Alzheimer disease: an update for the oral health care provider. ACTA ACUST UNITED AC 2008; 106:467-76. [PMID: 18928896 DOI: 10.1016/j.tripleo.2008.06.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2008] [Revised: 06/02/2008] [Accepted: 06/03/2008] [Indexed: 01/07/2023]
Abstract
Alzheimer disease (AD) is a progressive neurologic disorder that manifests as memory loss, personality changes, global cognitive dysfunction, and functional impairment. As the United States population continues to age, the prevalence of AD will rise. Accordingly, oral health care providers will be more likely to treat patients affected by this disease; therefore, it is necessary to understand the pharmacologic agents used for the management of AD. This article provides an update of the available drug therapies for AD and discusses their implications on the oral and dental health of patients.
Collapse
Affiliation(s)
- Lena N Turner
- Department of Oral Medicine, University of Pennsylvania School of Dental Medicine, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
50
|
van Straaten ECW, Harvey D, Scheltens P, Barkhof F, Petersen RC, Thal LJ, Jack CR, DeCarli C. Periventricular white matter hyperintensities increase the likelihood of progression from amnestic mild cognitive impairment to dementia. J Neurol 2008; 255:1302-8. [PMID: 18825439 DOI: 10.1007/s00415-008-0874-y] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2007] [Revised: 12/22/2007] [Accepted: 01/14/2008] [Indexed: 11/30/2022]
Abstract
BACKGROUND White matter hyperintensities (WMH) have an effect on cognition and are increased in severity among individuals with amnestic mild cognitive impairment (aMCI). The influence of WMH on progression of aMCI to Alzheimer's disease (AD) is less clear. METHODS Data were drawn from a three-year prospective, double blind, placebo controlled clinical trial that examined the effect of donepezil or vitamin E on progression from aMCI to AD. WMH from multiple brain regions were scored on MR images obtained at entry into the trial from a subset of 152 study participants using a standardized visual rating scale. Cox proportional hazards models adjusting for age, education and treatment arm were used to investigate the role of WMH on time to progression. RESULTS 55 of the 152 (36.2 %) aMCI subjects progressed to AD. Only periventricular hyperintensities (PVH) were related to an increased risk of AD within three years (HR = 1.59, 95 % CI = 1.24 - 2.05, p-value < 0.001). Correcting for medial temporal lobe atrophy or the presence of lacunes did not change statistical significance. CONCLUSION PVH are associated with an increased risk of progression from aMCI to AD. This suggests that PVH, an MRI finding thought to represent cerebrovascular damage, contributes to AD onset in vulnerable individuals independent of Alzheimer pathology.
Collapse
Affiliation(s)
- Elisabeth C W van Straaten
- Dept. of Neurology and Alzheimer Center, VU Medical Center, De Boelelaan 1117, 7057, 1007 MB, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|