1
|
Wang T, Liu Y, Lu Y, Chi L. NTN-1 attenuates amyloid-β-mediated microglial neuroinflammation and memory impairment via the NF-κB pathway and NLRP3 inflammasome in a rat model of Alzheimer's disease. Front Aging Neurosci 2025; 17:1516399. [PMID: 40357234 PMCID: PMC12066592 DOI: 10.3389/fnagi.2025.1516399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 04/08/2025] [Indexed: 05/15/2025] Open
Abstract
Introduction Neuroinflammation driven by microglial activation represents a pivotal pathological mechanism underlying brain injury in Alzheimer's disease (AD), with NLRP3 inflammasome activation being a hallmark feature of this process. Netrin-1 (NTN-1) was recently shown to have potent anti-inflammatory and anti-apoptotic properties in a range of inflammatory diseases; however, its potential effect on neuroinflammation in AD treatment has not been well examined. Accordingly, this study aimed to investigate the effects of NTN-1 on cognitive impairment and to explore the anti-inflammatory properties related to the NLRP3 inflammasome and NF-κB signaling in Aβ1-42-induced rat models. Methods We assessed the effects of NTN-1 on neurobehavioral function, microglial activation and neuroinflammation mechanisms in Aβ1-42-treated rats using the Morris water maze test and Western blotting. Results Our results indicated that microinjections of NTN-1 attenuated Aβ1-42-induced memory and cognitive dysfunction and significantly inhibited microglial proliferation and NLRP3 inflammasome activation in the hippocampus and cortex of AD rats. Additionally, NTN-1 effectively prevented proinflammatory factor (IL1β and IL18) release and NF-κB signaling upstream activation. Discussion Overall, the results of the present study indicated that exogenous NTN-1 treatment prevented neuroinflammation and cognitive deficits by inhibiting microglial activation, which is possibly mediated by the NF-κB signaling pathway and NLRP3 inflammasome activation in Aβ1-42-simulated rat models. NTN-1 emerges as a promising therapeutic candidate for mitigating microglia-mediated neuropathology in AD through its anti-inflammatory properties.
Collapse
Affiliation(s)
| | | | | | - Lijun Chi
- Department of Neurology, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
2
|
Mashhadi-Sharif N, Soodi M, Mirnajafi-Zadeh J, Shadboorestan A, Ebadi-Ramsheh H, Bakhtiarzadeh F. Ferulago angulata Extract Protects against Beta-amyloid-induced Memory Impairment Through Modulation of Monoaminoxidase Enzymes in the Rat's Hippocampus. Neurochem Res 2025; 50:114. [PMID: 40042687 DOI: 10.1007/s11064-025-04365-3] [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: 12/06/2023] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 04/26/2025]
Abstract
Ferulago angulata, a medicinal plant of the Apiaceae family, is known for its antioxidative, anti-inflammatory, anti-apoptotic, and neuroprotective effects. This study investigated the protective potential of F. angulata total extract against memory impairment induced by amyloid β-peptide (Aβ25-35) in rats. Aβ25-35 was bilaterally administered into the CA1 region of the hippocampus in male Wistar rats for four consecutive days. Starting from the first surgery day, rats received oral treatment with F. angulata extract (400 mg/kg) or Rivastigmine (1.5 mg/kg) for 16 consecutive days. Morris water maze tests conducted from days 14 to 17 evaluated the learning and memory performance, followed by biochemical and molecular assays on isolated hippocampi. F. angulata extract significantly reversed Aβ-induced deficits in learning and memory. In the Aβ group, monoamine oxidase A and B enzyme activities and gene expression increased, along with elevated acetylcholinesterase gene expression but decreased enzyme activity compared to the control group. Treatment with F. angulata extract mitigated all these abnormalities. Safety assessments revealed no alterations in blood biochemistry or histology following F. angulata extract treatment. In conclusion, our findings suggest that F. angulata extract possesses neuroprotective properties, and ameliorates Aβ-induced memory deficits by modulating monoamine oxidase enzyme activity and acetylcholinesterase levels without significant adverse effects. Thus, this safe herbal extract shows promise for further exploration in Alzheimer's disease research and potential therapeutic interventions.
Collapse
Affiliation(s)
| | - Maliheh Soodi
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran.
- Institute for natural products and medicinal plants, Tarbiat Modares University, Tehran, Iran.
| | - Javad Mirnajafi-Zadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Shadboorestan
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hanieh Ebadi-Ramsheh
- Department of Toxicology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Fatemeh Bakhtiarzadeh
- Department of Physiology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| |
Collapse
|
3
|
Zellmer JC, Tarantino MB, Kim M, Lomoio S, Maesako M, Hajnóczky G, Bhattacharyya R. Stabilization of mitochondria-associated endoplasmic reticulum membranes regulates Aβ generation in a three-dimensional neural model of Alzheimer's disease. Alzheimers Dement 2025; 21:e14417. [PMID: 39713841 PMCID: PMC11848173 DOI: 10.1002/alz.14417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/24/2024] [Accepted: 10/21/2024] [Indexed: 12/24/2024]
Abstract
INTRODUCTION We previously demonstrated that regulating mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs) affects axonal Aβ generation in a well-characterized three-dimensional (3D) neural Alzheimer's disease (AD) model. MAMs vary in thickness and length, impacting their functions. Here, we examined the effect of MAM thickness on Aβ in our 3D neural model of AD. METHODS We employed fluorescence resonance energy transfer (FRET) or fluorescence-based MAM stabilizers, electron microscopy, Aβ enzyme-linked immunosorbent assay (ELISA), and live-cell imaging with kymography to assess how stabilizing MAMs of different gap widths influence Aβ production and MAM axonal mobility. RESULTS Stabilizing tight MAMs (∼6 nm gap width) significantly increased Aβ levels, whereas basal (∼25 nm) and loose MAMs (∼40 nm) maintained or reduced Aβ levels, respectively. Tight MAMs reduced mitochondrial axonal velocity compared to basal MAMs, while loose MAMs showed severely reduced axonal distribution. DISCUSSION Our findings suggest that stabilizing MAMs of specific gap widths, particularly in axons, without complete destabilization could be an effective therapeutic strategy for AD. HIGHLIGHTS The stabilization of MAMs exacerbates or ameliorates Aβ generation from AD neurons in a MAM gap width-dependent manner. A specific stabilization threshold within the MAM gap width spectrum shifts the amyloidogenic process to non-amyloidogenic. Tight MAMs slow down mitochondrial axonal transport compared to lose MAMs offering a quantitative method for measuring MAM stabilization.
Collapse
Affiliation(s)
- Jacob C. Zellmer
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Marina B. Tarantino
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Michelle Kim
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| | - Selene Lomoio
- Department of NeuroscienceTufts University School of MedicineBostonMassachusettsUSA
| | - Masato Maesako
- Alzheimer's Disease Research UnitMassGeneral Institute for Neurodegenerative DiseaseMassachusetts General Hospital/Harvard Medical SchoolCharlestownMassachusettsUSA
| | - György Hajnóczky
- MitoCare CenterDepartment of PathologyAnatomy & Cell BiologyThomas Jefferson UniversityPhiladelphiaPennsylvaniaUSA
| | - Raja Bhattacharyya
- Genetics and Aging Research UnitMassGeneral Institute for Neurodegenerative DiseaseHenry and Allison McCance Center for Brain HealthDepartment of NeurologyMassachusetts General Hospital, Harvard Medical SchoolCharlestownMassachusettsUSA
| |
Collapse
|
4
|
Reid GA, Darvesh S. Interaction of exogenous acetylcholinesterase and butyrylcholinesterase with amyloid-β plaques in human brain tissue. Chem Biol Interact 2024; 395:111012. [PMID: 38648920 DOI: 10.1016/j.cbi.2024.111012] [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: 03/14/2024] [Revised: 04/10/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) are associated with amyloid-β (Aβ) plaques and exhibit altered biochemical properties in human Alzheimer's disease (AD), as well as in the transgenic 5XFAD mouse model of AD amyloidosis. In the brains of the 5XFAD mouse model devoid of BChE enzyme (5XFAD/BChE-KO), incubation of tissue sections with exogenous BChE purified from human plasma (pl-BChE) leads to its association with Aβ plaques and its biochemical properties are comparable to those reported for endogenous BChE associated with plaques in both human AD and in 5XFAD mouse brain tissue. We sought to determine whether these observations in 5XFAD/BChE-KO mice also apply to human brain tissues. To do so, endogenous ChE activity in human AD brain tissue sections was quenched with 50 % aqueous acetonitrile (MeCNaq) leaving the tissue suitable for further studies. Quenched sections were then incubated with recombinant AChE (r-AChE) or pl-BChE and stained for each enzymes' activity. Exogenous r-AChE or pl-BChE became associated with Aβ plaques, and when bound, had properties that were comparable to the endogenous ChE enzymes associated with plaques in AD brain tissues without acetonitrile treatment. These findings in human AD brain tissue extend previous observations in the 5XFAD/BChE-KO mouse model and demonstrate that exogenously applied r-AChE and pl-BChE have high affinity for Aβ plaques in human brain tissues. This association alters the biochemical properties of these enzymes, most likely due a conformational change. If incorporation of AChE and BChE in Aβ plaques facilitates AD pathogenesis, blocking this association could lead to disease-modifying approaches to AD. This work provides a method to study the mechanism of AChE and BChE interaction with Aβ plaque pathology in post-mortem human brain tissue.
Collapse
Affiliation(s)
- G A Reid
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada
| | - S Darvesh
- Department of Medical Neuroscience, Dalhousie University, Halifax, NS, Canada; Department of Medicine (Geriatric Medicine and Neurology), Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
5
|
Gamit N, Patil M, B Sundrappa S, Sundaram SM, Sethi G, Dharmarajan A, Warrier S. Mesenchymal stem cell-derived rapid drug screening system for Alzheimer's disease for the identification of novel drugs. Drug Dev Res 2023; 84:1496-1512. [PMID: 37571798 DOI: 10.1002/ddr.22102] [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: 02/14/2023] [Revised: 06/11/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
Abstract
A reliable and efficient in vitro model is needed to screen drugs for Alzheimer's disease (AD), as many drugs are currently in the developmental stage. To address this, we developed an in vitro model using amniotic membrane-derived mesenchymal stem cells (AM-MSCs) to screen novel drugs for AD. We differentiated AM-MSCs into neurons and degenerated them using beta amyloid1-42 (Aß). We then tested AD drugs, which are commercially available such as donepezil, rivastigmine, memantine, citicoline, and two novel drugs, that is, probucol, an anti-hyperlipidaemic drug, and NMJ-2, a cinnamic acid analogue for their potential to protect the cells against neurodegeneration. We used gene expression and immunofluorescence staining to assess the neuroprotective ability of these drugs. We also measured the ability of these drugs to reduce lactate dehydrogenase, reactive oxygen species, and nitric oxide levels, as well as their ability to stabilize the mitochondrial membrane potential and increase acetylcholine (ACh) levels. The AD drugs and novel drugs reduced cytotoxicity and oxidative stress, stabilized mitochondrial membrane potential, and restored ACh levels. Furthermore, they reduced BACE1 expression, with a concomitant increase in the expression of cholinergic markers. This AM-MSCs-based AD-like model has immense potential to be an accurate human model and an alternative to animal models for testing a large number of lead compounds in a short time. Our results also suggest that the novel drugs probucol and NMJ-2 may protect against Aß-induced neurodegeneration.
Collapse
Affiliation(s)
- Naisarg Gamit
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - Manasi Patil
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - Soumya B Sundrappa
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - S Mohana Sundaram
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Arun Dharmarajan
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| | - Sudha Warrier
- Division of Cancer Stem Cells and Cardiovascular Regeneration, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
- Cuor Stem Cellutions Pvt Ltd, Manipal Institute of Regenerative Medicine, Manipal Academy of Higher Education (MAHE), Bangalore, India
- Department of Biotechnology, Faculty of Biomedical Sciences and Technology, Sri Ramachandra Institute of Higher Education and Research, Chennai, India
| |
Collapse
|
6
|
Neha, Parvez S. Emerging therapeutics agents and recent advances in drug repurposing for Alzheimer's disease. Ageing Res Rev 2023; 85:101815. [PMID: 36529440 DOI: 10.1016/j.arr.2022.101815] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 11/29/2022] [Accepted: 12/01/2022] [Indexed: 12/23/2022]
Abstract
Alzheimer's disease (AD) is a multivariate and diversified disease and affects the most sensitive areas of the brain, the cerebral cortex, and the hippocampus. AD is a progressive age-related neurodegenerative disease most often associated with memory deficits and cognition that get more worsen over time. The central theory on the pathophysiological hallmark features of AD is characterized by the accumulation of amyloid β (Aβ) peptides, also associated with tau proteins (τ) dysfunctioning which leads to distorted microtubular structure, affects the cholinergic system, and mitochondrial biogenesis. This review emphasizes how simple it is to find novel treatments for AD and focuses on several recently developed medications through repurposing that can speed up traditional drug development.
Collapse
Affiliation(s)
- Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
7
|
Moreira NCDS, Lima JEBDF, Marchiori MF, Carvalho I, Sakamoto-Hojo ET. Neuroprotective Effects of Cholinesterase Inhibitors: Current Scenario in Therapies for Alzheimer's Disease and Future Perspectives. J Alzheimers Dis Rep 2022; 6:177-193. [PMID: 35591949 PMCID: PMC9108627 DOI: 10.3233/adr-210061] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/16/2022] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a slowly progressive neurodegenerative disease conceptualized as a continuous process, ranging from mild cognitive impairment (MCI), to the mild, moderate, and severe clinical stages of AD dementia. AD is considered a complex multifactorial disease. Currently, the use of cholinesterase inhibitors (ChEI), such as tacrine, donepezil, rivastigmine, and galantamine, has been the main treatment for AD patients. Interestingly, there is evidence that ChEI also promotes neuroprotective effects, bringing some benefits to AD patients. The mechanisms by which the ChEI act have been investigated in AD. ChEI can modulate the PI3K/AKT pathway, which is an important signaling cascade that is capable of causing a significant functional impact on neurons by activating cell survival pathways to promote neuroprotective effects. However, there is still a huge challenge in the field of neuroprotection, but in the context of unravelling the details of the PI3K/AKT pathway, a new scenario has emerged for the development of more efficient drugs that act on multiple protein targets. Thus, the mechanisms by which ChEI can promote neuroprotective effects and prospects for the development of new drug candidates for the treatment of AD are discussed in this review.
Collapse
Affiliation(s)
| | | | - Marcelo Fiori Marchiori
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Ivone Carvalho
- School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| | - Elza Tiemi Sakamoto-Hojo
- Department of Genetics, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Department of Biology, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Ribeirão Preto, SP, Brazil
| |
Collapse
|
8
|
Dulla BS, S. B, K. LP. A Study on the Effect of Valeric Acid in Alzheimer's Induced Rats by the Estimation of Aβ 1-42 Biomarker. JOURNAL OF HEALTH AND ALLIED SCIENCES NU 2021. [DOI: 10.1055/s-0041-1736274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
AbstractThe effect of valeric acid on the behavior of Alzheimer's disease (AD)-induced rats by aluminum chloride (100 mg/kg body weight) was assessed using elevated plus maze (EPM) and the Hebb Williams maze (HWM). Amyloid β 1–42(Aβ1–42) biomarker was estimated by ELISA. In this study, valeric acid-treated rats were compared with those treated with piracetam (200 mg/kg), rivastigmine (0.5 mg/kg), and the results showed the rats treated with valeric acid had a very less transfer latency of EPM and HWM when compared with other standard drugs. In addition, valeric acid-treated rats showed reduced levels of amyloid β1–42 biomarker in the plasma. Hence, this study found that valeric acid may be suggested as a better drug for Alzheimer's disease.
Collapse
Affiliation(s)
- Blessina Sugandhi Dulla
- Department of Anatomy, Yenepoya Medical College, Yenepoya (deemed to be) University, Mangalore, Karnataka, India
| | - Bindhu S.
- Department of Anatomy, Yenepoya Medical College, Yenepoya (deemed to be) University, Mangalore, Karnataka, India
| | - Leena Pramod K.
- Department of Forensic Medicine and Toxicology, Yenepoya Medical College, Yenepoya (deemed to be) University, Mangalore, Karnataka, India
| |
Collapse
|
9
|
Walczak-Nowicka ŁJ, Herbet M. Acetylcholinesterase Inhibitors in the Treatment of Neurodegenerative Diseases and the Role of Acetylcholinesterase in their Pathogenesis. Int J Mol Sci 2021; 22:9290. [PMID: 34502198 PMCID: PMC8430571 DOI: 10.3390/ijms22179290] [Citation(s) in RCA: 79] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/21/2022] Open
Abstract
Acetylcholinesterase (AChE) plays an important role in the pathogenesis of neurodegenerative diseases by influencing the inflammatory response, apoptosis, oxidative stress and aggregation of pathological proteins. There is a search for new compounds that can prevent the occurrence of neurodegenerative diseases and slow down their course. The aim of this review is to present the role of AChE in the pathomechanism of neurodegenerative diseases. In addition, this review aims to reveal the benefits of using AChE inhibitors to treat these diseases. The selected new AChE inhibitors were also assessed in terms of their potential use in the described disease entities. Designing and searching for new drugs targeting AChE may in the future allow the discovery of therapies that will be effective in the treatment of neurodegenerative diseases.
Collapse
Affiliation(s)
| | - Mariola Herbet
- Chair and Department of Toxicology, Faculty of Pharmacy, Medical University of Lublin, Jaczewskiego 8bStreet, 20-090 Lublin, Poland;
| |
Collapse
|
10
|
Reinhardt F, Scarmeas N, Karan R, Veldandi UK, Modali S, Duvvuri K, Pathan RK. Real-world Utilisation of the Rivastigmine Transdermal Patches Accompanying the use of Risk Minimisation Tools in Patients with Dementia. Curr Alzheimer Res 2021; 18:273-282. [PMID: 34279198 DOI: 10.2174/1567205018666210716120540] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 01/14/2021] [Accepted: 03/17/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Transdermal patches are convenient to use, especially in patients with Alzheimer's disease (AD)-associated dementia. However, various identified risks of errors in ad- ministering the patches cannot be disregarded. Patient Reminder Cards (PRCs, included a Medica- tion record sheet [MRS]) have been recently introduced as a risk minimisation tool to prevent incor- rect patch use (IU). OBJECTIVES This study aimed to assess the effectiveness of PRCs to prevent IU and to investigate the dose titration pattern of rivastigmine patches in a real-world setting. METHODS This multinational, observational, 11-month study included patients with AD currently using rivastigmine patches (4.6 mg/day, 9.5 mg/day, 13.3 mg/day) accompanied by a caregiver. Study outcomes were IU, including multiple patch use (MPU), incorrect patch placement, other IUs, perceived usefulness of the PRCs, and titration patterns of the patches. RESULTS Of the total 614 patients included, most were aged ≥65 years and had mild-to-moderate AD. Before and during the study, 27.7% and 18.0% of patients reported IU, respectively. Most pa- tients used MRS, and 73.5% rated it 'helpful' and reported lower rates of IU than those who report- ed it 'not helpful' (13.9%-16.5% vs. 20.2%). Overall, 141 patients had dose titrations, with 75.8% being up-titrated from 4.6 mg/day to 9.5 mg/day after a mean duration of 58 days. Safety findings were consistent with the established profile for the rivastigmine patch. CONCLUSION PRC was effective as a risk minimisation tool in limiting the inappropriate use of ri- vastigmine patches. The majority of patients requiring dose-change were up-titrated to 9.5 mg/day patches.
Collapse
Affiliation(s)
| | - Nikolaos Scarmeas
- 1st Department of Neurology, Aiginition Hospital, National and Kapodistrian University of Athens Medical School, Athens. Greece
| | - Rajesh Karan
- Global Program Clinical Head, Novartis Pharma AG. Switzerland
| | | | | | | | | |
Collapse
|
11
|
Perez FP, Maloney B, Chopra N, Morisaki JJ, Lahiri DK. Repeated electromagnetic field stimulation lowers amyloid-β peptide levels in primary human mixed brain tissue cultures. Sci Rep 2021; 11:621. [PMID: 33436686 PMCID: PMC7804462 DOI: 10.1038/s41598-020-77808-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 11/17/2020] [Indexed: 02/07/2023] Open
Abstract
Late Onset Alzheimer's Disease is the most common cause of dementia, characterized by extracellular deposition of plaques primarily of amyloid-β (Aβ) peptide and tangles primarily of hyperphosphorylated tau protein. We present data to suggest a noninvasive strategy to decrease potentially toxic Aβ levels, using repeated electromagnetic field stimulation (REMFS) in primary human brain (PHB) cultures. We examined effects of REMFS on Aβ levels (Aβ40 and Aβ42, that are 40 or 42 amino acid residues in length, respectively) in PHB cultures at different frequencies, powers, and specific absorption rates (SAR). PHB cultures at day in vitro 7 (DIV7) treated with 64 MHz, and 1 hour daily for 14 days (DIV 21) had significantly reduced levels of secreted Aβ40 (p = 001) and Aβ42 (p = 0.029) peptides, compared to untreated cultures. PHB cultures (DIV7) treated at 64 MHz, for 1 or 2 hour during 14 days also produced significantly lower Aβ levels. PHB cultures (DIV28) treated with 64 MHz 1 hour/day during 4 or 8 days produced a similar significant reduction in Aβ40 levels. 0.4 W/kg was the minimum SAR required to produce a biological effect. Exposure did not result in cellular toxicity nor significant changes in secreted Aβ precursor protein-α (sAPPα) levels, suggesting the decrease in Aβ did not likely result from redirection toward the α-secretase pathway. EMF frequency and power used in our work is utilized in human magnetic resonance imaging (MRI, thus suggesting REMFS can be further developed in clinical settings to modulate Aβ deposition.
Collapse
Affiliation(s)
- Felipe P Perez
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Division of General Internal Medicine and Geriatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan Maloney
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA
| | - Nipun Chopra
- Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA
| | - Jorge J Morisaki
- Department of Bioengineering, University of Illinois at Chicago, Chicago, IL, USA
| | - Debomoy K Lahiri
- Indiana University School of Medicine, Indianapolis, IN, USA.
- Department of Psychiatry, Institute of Psychiatric Research, Neuroscience Research Center, Indiana University School of Medicine, 320 W. 15th St, Indianapolis, IN, 46201, USA.
- Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
12
|
Rodd ZA, Hauser SR, Swartzwelder HS, Waeiss RA, Lahiri DK, Bell RL. Regulation of the deleterious effects of binge-like exposure to alcohol during adolescence by α7 nicotinic acetylcholine receptor agents: prevention by pretreatment with a α7 negative allosteric modulator and emulation by a α7 agonist in alcohol-preferring (P) male and female rats. Psychopharmacology (Berl) 2020; 237:2601-2611. [PMID: 32607619 PMCID: PMC7502519 DOI: 10.1007/s00213-020-05557-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Accepted: 05/18/2020] [Indexed: 12/23/2022]
Abstract
RATIONALE AND OBJECTIVES Binge-like alcohol consumption during adolescence associates with several deleterious consequences during adulthood including an increased risk for developing alcohol use disorder (AUD) and other addictions. Replicated preclinical data has indicated that adolescent exposure to binge-like levels of alcohol results in a reduction of choline acetyltransferase (ChAT) and an upregulation in the α7 nicotinic receptor (α7). From this information, we hypothesized that the α7 plays a critical role in mediating the effects of adolescent alcohol exposure. METHODS Male and female P rats were injected with the α7 agonist AR-R17779 (AR) once during 6 time points between post-natal days (PND) 29-37. Separate groups were injected with the α7 negative allosteric modulator (NAM) dehydronorketamine (DHNK) 2 h before administration of 4 g/kg EtOH (14 total exposures) during PND 28-48. On PND 75, all rats were given access to water and ethanol (15 and 30%) for 6 consecutive weeks (acquisition). All rats were then deprived of EtOH for 2 weeks and then, alcohol was returned (relapse). RESULTS Administration of AR during adolescence significantly increased acquisition of alcohol consumption during adulthood and prolonged relapse drinking in P rats. In contrast, administration of DHNK prior to binge-like EtOH exposure during adolescence prevented the increase in alcohol consumption observed during acquisition of alcohol consumption and the enhancement of relapse drinking observed during adulthood. DISCUSSION The data indicate that α7 mediates the effects of alcohol during adolescence. The data also indicate that α7 NAMs are potential prophylactic agents to reduce the deleterious effects of adolescent alcohol abuse.
Collapse
Affiliation(s)
- Zachary A Rodd
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA.
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA.
| | - Sheketha R Hauser
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - H Scott Swartzwelder
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - R Aaron Waeiss
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Debomoy K Lahiri
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| | - Richard L Bell
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Neuroscience Research Building, 320 W. 15th Street, Suite 300B, Indianapolis, IN, 46202-2266, USA
- Department of Psychiatry and Behavioral Sciences, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
13
|
Ray B, Maloney B, Sambamurti K, Karnati HK, Nelson PT, Greig NH, Lahiri DK. Rivastigmine modifies the α-secretase pathway and potentially early Alzheimer's disease. Transl Psychiatry 2020; 10:47. [PMID: 32066688 PMCID: PMC7026402 DOI: 10.1038/s41398-020-0709-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 11/25/2019] [Accepted: 12/19/2019] [Indexed: 12/12/2022] Open
Abstract
Rivastigmine (or Exelon) is a cholinesterase inhibitor, currently used as a symptomatic treatment for mild-to-moderate Alzheimer's disease (AD). Amyloid-β peptide (Aβ) generated from its precursor protein (APP) by β-secretase (or BACE1) and γ-secretase endoproteolysis. Alternative APP cleavage by α-secretase (a family of membrane-bound metalloproteases- Adamalysins) precludes the generation of toxic Aβ and yields a neuroprotective and neurotrophic secreted sAPPα fragment. Several signal transduction pathways, including protein kinase C and MAP kinase, stimulate α-secretase. We present data to suggest that rivastigmine, in addition to anticholinesterase activity, directs APP processing away from BACE1 and towards α-secretases. We treated rat neuronal PC12 cells and primary human brain (PHB) cultures with rivastigmine and the α-secretase inhibitor TAPI and assayed for levels of APP processing products and α-secretases. We subsequently treated 3×Tg (transgenic) mice with rivastigmine and harvested hippocampi to assay for levels of APP processing products. We also assayed postmortem human control, AD, and AD brains from subjects treated with rivastigmine for levels of APP metabolites. Rivastigmine dose-dependently promoted α-secretase activity by upregulating levels of ADAM-9, -10, and -17 α-secretases in PHB cultures. Co-treatment with TAPI eliminated rivastigmine-induced sAPPα elevation. Rivastigmine treatment elevated levels of sAPPα in 3×Tg mice. Consistent with these results, we also found elevated sAPPα in postmortem brain samples from AD patients treated with rivastigmine. Rivastigmine can modify the levels of several shedding proteins and directs APP processing toward the non-amyloidogenic pathway. This novel property of rivastigmine can be therapeutically exploited for disease-modifying intervention that goes beyond symptomatic treatment for AD.
Collapse
Affiliation(s)
- Balmiki Ray
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Bryan Maloney
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,grid.257413.60000 0001 2287 3919Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Kumar Sambamurti
- grid.259828.c0000 0001 2189 3475Department of Neurosciences, Medical University of South Carolina, Charleston, 29425 SC USA
| | - Hanuma K. Karnati
- grid.419475.a0000 0000 9372 4913National Institute on Aging, Drug Design and Development Section, Bethesda, MD 20892 USA
| | - Peter T. Nelson
- grid.266539.d0000 0004 1936 8438Sanders-Brown Center on Aging, University of Kentucky, Lexington, KY 40536 USA
| | - Nigel H. Greig
- grid.419475.a0000 0000 9372 4913National Institute on Aging, Drug Design and Development Section, Bethesda, MD 20892 USA
| | - Debomoy K. Lahiri
- grid.257413.60000 0001 2287 3919Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,grid.257413.60000 0001 2287 3919Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,grid.257413.60000 0001 2287 3919Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
14
|
Saito Y, Sakata M, Kobayakawa M, Kawachi H, Kawaguchi K, Hiki Y, Kato M, Mori M, Hasegawa M, Ohashi N, Yuzawa Y, Kitaguchi N. Removal of Aβ Oligomers from the Blood: A Potential Therapeutic System for Alzheimer's Disease. Neuropsychiatr Dis Treat 2020; 16:607-627. [PMID: 32210564 PMCID: PMC7064818 DOI: 10.2147/ndt.s241074] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Accepted: 02/14/2020] [Indexed: 01/12/2023] Open
Abstract
PURPOSE Amyloid-β protein (Aβ) is one of the causative proteins of Alzheimer's disease. We have been developing extracorporeal blood Aβ-removal systems as a method for enhancing Aβ clearance from the brain. We reported previously that medical adsorbents and hemodialyzers removed Aβ monomers from peripheral blood, which was associated with influx of Aβ monomers from the brain into the bloodstream. Our intent here was to develop a method to promote clearance of Aβ oligomers and to provide an estimate of the molecular size of intact Aβ oligomers in plasma. METHODS Two hollow-fiber devices with different pore sizes (Membranes A and B) were evaluated as removers of Aβ oligomers with human plasma in vitro. The concomitant removal of Aβ oligomers and monomers was investigated by using Membrane B and hexadecyl alkylated cellulose beads or polysulfone hemodialyzers. Double-filtration plasmapheresis with Membrane A was investigated as an approach for the removal of plasma Aβ oligomers in humans. RESULTS Aβ oligomers were effectively removed by both Membranes A and B. The increase of Aβ oligomers in plasma was observed just after the removal of plasma Aβ oligomers in humans. The intact molecular size of major Aβ oligomers in the plasma was estimated to be larger than albumin at approximately 60 kDa or more. Additionally, the concomitant removal of Aβ monomers and oligomers evoked dissociation of larger Aβ oligomers into smaller ones and monomers. CONCLUSION Aβ oligomers were cleared from plasma both in vitro and in human subjects by using hollow-fiber membranes with large pores, indicating that their intact sizes were mostly larger than 60 kDa. Aβ oligomers in peripheral circulation were increased after some clearances in human. Further investigation will determine whether the Aβ oligomers detected in circulation after clearance were via influx from the brain.
Collapse
Affiliation(s)
- Yuta Saito
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.,Graduate School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan.,Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Miwa Sakata
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Moe Kobayakawa
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Kazunori Kawaguchi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiyuki Hiki
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Masao Kato
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Mayuko Mori
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan.,Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| | - Midori Hasegawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | | | - Yukio Yuzawa
- Department of Nephrology, School of Medicine, Fujita Health University, Toyoake, Aichi, Japan
| | - Nobuya Kitaguchi
- Faculty of Clinical Engineering, School of Medical Sciences, Fujita Health University, Toyoake, Aichi, Japan
| |
Collapse
|
15
|
Donepezil modulates amyloid precursor protein endocytosis and reduction by up-regulation of SNX33 expression in primary cortical neurons. Sci Rep 2019; 9:11922. [PMID: 31417133 PMCID: PMC6695423 DOI: 10.1038/s41598-019-47462-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/16/2019] [Indexed: 11/09/2022] Open
Abstract
Donepezil, a therapeutic drug for Alzheimer’s disease, ameliorates cognitive dysfunction through selective inhibition of acetylcholinesterase. However, recent studies have also reported off-target effects of donepezil that likely contribute to its therapeutic effects. In this study, we investigated the (i) role of donepezil in amyloid precursor protein (APP) processing and (ii) involvement of sorting nexin protein 33 (SNX33), a member of the sorting nexin protein family, in this processing. Results showed that donepezil induces an increase in SNX33 expression in primary cortical neurons. The secretion of sAPPα in culture media increased, whereas the expression of full-length APP in the cell lysate remained unchanged. Exposure of cortical cultures to donepezil led to a decrease in amyloid β (Aβ) protein levels in a concentration- and time-dependent manner. This decrease was not affected by concomitant treatment with acetylcholine receptor antagonists. SNX33 knockdown by target-specific morpholino oligos inhibited the effects of donepezil. Donepezil treatment increased cell membrane surface expression of APP in SNX33 expression-dependent manner. These results suggested that donepezil decreases the level of Aβ by increasing SNX33 expression and APP cleavage by α-secretase in cortical neurons.
Collapse
|
16
|
Li K, Li J, Zheng J, Qin S. Reactive Astrocytes in Neurodegenerative Diseases. Aging Dis 2019; 10:664-675. [PMID: 31165009 PMCID: PMC6538217 DOI: 10.14336/ad.2018.0720] [Citation(s) in RCA: 264] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Accepted: 07/20/2018] [Indexed: 12/17/2022] Open
Abstract
Astrocytes, the largest and most numerous glial cells in the central nervous system (CNS), play a variety of important roles in regulating homeostasis, increasing synaptic plasticity and providing neuroprotection, thus helping to maintain normal brain function. At the same time, astrocytes can participate in the inflammatory response and play a key role in the progression of neurodegenerative diseases. Reactive astrocytes are strongly induced by numerous pathological conditions in the CNS. Astrocyte reactivity is initially characterized by hypertrophy of soma and processes, triggered by different molecules. Recent studies have demonstrated that neuroinflammation and ischemia can elicit two different types of reactive astrocytes, termed A1s and A2s. However, in the case of astrocyte reactivity in different neurodegenerative diseases, the recently published research issues remain a high level of conflict and controversy. So far, we still know very little about whether and how the function or reactivity of astrocytes changes in the progression of different neurodegenerative diseases. In this review, we aimed to briefly discuss recent studies highlighting the complex contribution of astrocytes in the process of various neurodegenerative diseases, which may provide us with new prospects for the development of an excellent therapeutic target for neurodegenerative diseases.
Collapse
Affiliation(s)
- Kunyu Li
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jiatong Li
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Jialin Zheng
- 2Center for Translational Neurodegeneration and Regenerative Therapy, Shanghai Tenth People's Hospital affiliated to Tongji University School of Medicine, Shanghai, China
| | - Song Qin
- 1Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| |
Collapse
|
17
|
Azure B affects amyloid precursor protein metabolism in PS70 cells. Chem Biol Interact 2019; 299:88-93. [PMID: 30500345 DOI: 10.1016/j.cbi.2018.11.023] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Revised: 10/22/2018] [Accepted: 11/27/2018] [Indexed: 01/01/2023]
Abstract
Alzheimer's disease (AD), the most common form of dementia, is characterized by abundant deposition of amyloid-β (Aβ) peptide that is the result of sequential cleavage of amyloid precursor protein (APP) by β-secretase and γ-secretase. Several studies have documented that inhibition of Aβ peptide synthesis or facilitating its degradation is one of the attractive therapeutic strategies in AD. Methylene blue (MethB), which has recently been investigated in Phase II clinical trials, is a prominent inhibitor in reducing Aβ oligomers. Herein, we wonder whether the mitigating effects of MethB on amyloid metabolism are related to the activity of its major metabolite, azure B. The goal of this study was to investigate the effects of azure B, which is also a cholinesterase inhibitor, on APP processing by using Chinese hamster ovary cells stably expressing human wild-type APP and presenilin 1 (PS70). Azure B significantly decreased the levels of secreted APPα (sAPPα) and Aβ40/42 in culture medium with a dose-dependent manner. A significant decrease was also observed in the levels of intracellular APP without affecting the cell viability. In parallel with the decrease of APP and APP metabolites, the activity of β-secretase 1 (BACE1) was significantly attenuated compared to control. Overall, our results show that azure B has a large contribution for the pharmacological profile of MethB in APP metabolism.
Collapse
|
18
|
Yuksel M, Biberoglu K, Onder S, Akbulut KG, Tacal O. Toluidine blue O modifies hippocampal amyloid pathology in a transgenic mouse model of Alzheimer's disease. Biochimie 2018; 146:105-112. [DOI: 10.1016/j.biochi.2017.12.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 12/12/2017] [Indexed: 01/04/2023]
|
19
|
Muñoz-Moreno JA, Prats A, Moltó J, Garolera M, Pérez-Álvarez N, Díez-Quevedo C, Miranda C, Fumaz CR, Ferrer MJ, Clotet B. Transdermal rivastigmine for HIV-associated cognitive impairment: A randomized pilot study. PLoS One 2017; 12:e0182547. [PMID: 28854283 PMCID: PMC5576750 DOI: 10.1371/journal.pone.0182547] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Accepted: 07/11/2017] [Indexed: 11/19/2022] Open
Abstract
Objective To assess the efficacy and safety of transdermal rivastigmine for the treatment of HIV-associated cognitive impairment. Methods We recruited HIV-infected patients with cognitive impairment on stable antiretroviral therapy in a randomized controlled pilot trial with a 48-week follow-up. An additional assessment was held at 12 weeks. Participants received transdermal rivastigmine (9.5 mg daily), lithium (400 mg twice daily, titrated progressively), or remained in a control group (no new medication). The primary efficacy endpoint was change in a global cognitive score (NPZ-7). Secondary endpoints included change in specific cognitive measures, domains, and functional parameters. Safety covered the frequency of adverse events and changes in laboratory results. Results Seventy-six subjects were screened, and 29 were finally enrolled. Better cognitive outcomes were observed in all groups, although there were no significant differences between the arms (mean NPZ-7 change [SD]): rivastigmine, 0.35 (0.14); lithium, 0.25 (0.40); control, 0.20 (0.44) (p = 0.78). The rivastigmine group showed the highest positive trend (mean NPZ-7 [SD], baseline vs week 48): rivastigmine, –0.47 (0.22) vs –0.11 (0.29), p = 0.06; lithium, –0.50 (0.40) vs –0.26 (0.21), p = 0.22; control, –0.52 (0.34) vs –0.32 (0.52), p = 0.44. The cognitive domains with the highest positive trends were information processing speed at week 12 and executive function at week 48 (rivastigmine vs control): information processing speed, 0.35 (0.64) vs –0.13 (0.25), p = 0.17, d = 0.96; and executive functioning, 0.73 (0.33) vs 0.03 (0.74), p = 0.09, d = 1.18. No relevant changes were observed regarding functional outcomes. A total of 12 (41%) individuals dropped out of the study: 2 (20%) were due to medication-related effects in the rivastigmine group and 4 (36%) in the lithium group. No severe adverse events were reported. Conclusions The results from this small randomized trial indicate that transdermal rivastigmine did not provide significant cognitive benefits in people with HAND on stable antiretroviral therapy, even though positive trends were found in specific cognitive domains. Relevant tolerability issues were not observed.
Collapse
Affiliation(s)
- Jose A. Muñoz-Moreno
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- Facultat de Psicologia i Ciències de l'Educació, Universitat Oberta de Catalunya (UOC), Barcelona, Catalonia, Spain
- * E-mail:
| | - Anna Prats
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Catalonia, Spain
| | - José Moltó
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Maite Garolera
- Clinical Research Group for Brain, Cognition and Behavior, Consorci Sanitari Hospital de Terrassa, Terrassa, Catalonia, Spain
- Grup de Recerca Consolidat en Neuropsicologia, Universitat de Barcelona (UB), Barcelona, Catalonia, Spain
| | - Núria Pérez-Álvarez
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- Departament d'Estadística i Investigació Operativa, Universitat Politècnica de Catalunya (UPC), Barcelona, Catalonia, Spain
| | - Crisanto Díez-Quevedo
- Departament de Psiquiatria i Medicina Legal, Universitat Autònoma de Barcelona (UAB), Cerdanyola del Vallès, Catalonia, Spain
- Servei de Psiquiatria, Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Cristina Miranda
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Carmina R. Fumaz
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Maria J. Ferrer
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
| | - Bonaventura Clotet
- Fundació Lluita contra la SIDA (FLS), Hospital Universitari Germans Trias i Pujol, Badalona, Catalonia, Spain
- Institut per la Recerca de la SIDA IrsiCaixa, Badalona, Catalonia, Spain
- Càtedra de la SIDA i Malalties Relacionades, Universitat de Vic—Universitat Central de Catalunya (UVic), Vic, Catalonia, Spain
| | | |
Collapse
|
20
|
Yuksel M, Biberoglu K, Onder S, Akbulut KG, Tacal O. Effects of phenothiazine-structured compounds on APP processing in Alzheimer's disease cellular model. Biochimie 2017; 138:82-89. [DOI: 10.1016/j.biochi.2017.04.012] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/25/2017] [Indexed: 01/04/2023]
|
21
|
Mockett BG, Richter M, Abraham WC, Müller UC. Therapeutic Potential of Secreted Amyloid Precursor Protein APPsα. Front Mol Neurosci 2017; 10:30. [PMID: 28223920 PMCID: PMC5293819 DOI: 10.3389/fnmol.2017.00030] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 01/25/2017] [Indexed: 11/26/2022] Open
Abstract
Cleavage of the amyloid precursor protein (APP) by α-secretase generates an extracellularly released fragment termed secreted APP-alpha (APPsα). Not only is this process of interest due to the cleavage of APP within the amyloid-beta sequence, but APPsα itself has many physiological properties that suggest its great potential as a therapeutic target. For example, APPsα is neurotrophic, neuroprotective, neurogenic, a stimulator of protein synthesis and gene expression, and enhances long-term potentiation (LTP) and memory. While most early studies have been conducted in vitro, effectiveness in animal models is now being confirmed. These studies have revealed that either upregulating α-secretase activity, acutely administering APPsα or chronic delivery of APPsα via a gene therapy approach can effectively treat mouse models of Alzheimer's disease (AD) and other disorders such as traumatic head injury. Together these findings suggest the need for intensifying research efforts to harness the therapeutic potential of this multifunctional protein.
Collapse
Affiliation(s)
- Bruce G. Mockett
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of OtagoOtago, New Zealand
| | - Max Richter
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg UniversityHeidelberg, Germany
| | - Wickliffe C. Abraham
- Department of Psychology, Brain Health Research Centre, Brain Research New Zealand, University of OtagoOtago, New Zealand
| | - Ulrike C. Müller
- Department of Functional Genomics, Institute for Pharmacy and Molecular Biotechnology, Heidelberg UniversityHeidelberg, Germany
| |
Collapse
|
22
|
Multi-target-directed therapeutic potential of 7-methoxytacrine-adamantylamine heterodimers in the Alzheimer's disease treatment. Biochim Biophys Acta Mol Basis Dis 2017; 1863:607-619. [DOI: 10.1016/j.bbadis.2016.11.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 11/04/2016] [Accepted: 11/15/2016] [Indexed: 12/30/2022]
|
23
|
Soukup O, Winder M, Killi UK, Wsol V, Jun D, Kuca K, Tobin G. Acetylcholinesterase Inhibitors and Drugs Acting on Muscarinic Receptors- Potential Crosstalk of Cholinergic Mechanisms During Pharmacological Treatment. Curr Neuropharmacol 2017; 15:637-653. [PMID: 27281175 PMCID: PMC5543679 DOI: 10.2174/1570159x14666160607212615] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Revised: 04/28/2016] [Accepted: 05/31/2016] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Pharmaceuticals with targets in the cholinergic transmission have been used for decades and are still fundamental treatments in many diseases and conditions today. Both the transmission and the effects of the somatomotoric and the parasympathetic nervous systems may be targeted by such treatments. Irrespective of the knowledge that the effects of neuronal signalling in the nervous systems may include a number of different receptor subtypes of both the nicotinic and the muscarinic receptors, this complexity is generally overlooked when assessing the mechanisms of action of pharmaceuticals. METHODS We have search of bibliographic databases for peer-reviewed research literature focused on the cholinergic system. Also, we have taken advantage of our expertise in this field to deduce the conclusions of this study. RESULTS Presently, the life cycle of acetylcholine, muscarinic receptors and their effects are reviewed in the major organ systems of the body. Neuronal and non-neuronal sources of acetylcholine are elucidated. Examples of pharmaceuticals, in particular cholinesterase inhibitors, affecting these systems are discussed. The review focuses on salivary glands, the respiratory tract and the lower urinary tract, since the complexity of the interplay of different muscarinic receptor subtypes is of significance for physiological, pharmacological and toxicological effects in these organs. CONCLUSION Most pharmaceuticals targeting muscarinic receptors are employed at such large doses that no selectivity can be expected. However, some differences in the adverse effect profile of muscarinic antagonists may still be explained by the variation of expression of muscarinic receptor subtypes in different organs. However, a complex pattern of interactions between muscarinic receptor subtypes occurs and needs to be considered when searching for selective pharmaceuticals. In the development of new entities for the treatment of for instance pesticide intoxication, the muscarinic receptor selectivity needs to be considered. Reactivators generally have a muscarinic M2 receptor acting profile. Such a blockade may engrave the situation since it may enlarge the effect of the muscarinic M3 receptor effect. This may explain why respiratory arrest is the major cause for deaths by esterase blocking.
Collapse
Affiliation(s)
- Ondrej Soukup
- Department of Toxicology and Military Pharmacy, Faculty of Military Health Sciences, University of Defence, Hradec Kralove, Czech Republic
| | - Michael Winder
- Institute of Neuroscience and Physiology, Department of Pharmacology, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| | - Uday Kumar Killi
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Hradec Kralove, Czech Republic
| | - Vladimir Wsol
- Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Hradec Kralove, Czech Republic
| | - Daniel Jun
- Biomedical Research Centre, University Hospital Hradec Kralove, Hradec Kralove, Czech Republic
| | - Kamil Kuca
- Biomedical Research Centre, University Hospital Hradec Kralove, Czech Republic
| | - Gunnar Tobin
- Institute of Neuroscience and Physiology, Department of Pharmacology, the Sahlgrenska Academy at the University of Gothenburg, Sweden
| |
Collapse
|
24
|
Novel Nuclear Factor-KappaB Targeting Peptide Suppresses β-Amyloid Induced Inflammatory and Apoptotic Responses in Neuronal Cells. PLoS One 2016; 11:e0160314. [PMID: 27764084 PMCID: PMC5072831 DOI: 10.1371/journal.pone.0160314] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Accepted: 07/18/2016] [Indexed: 11/19/2022] Open
Abstract
In the central nervous system (CNS), activation of the transcription factor nuclear factor-kappa B (NF-κβ) is associated with both neuronal survival and increased vulnerability to apoptosis. The mechanisms underlying these dichotomous effects are attributed to the composition of NF-κΒ dimers. In Alzheimer’s disease (AD), β-amyloid (Aβ) and other aggregates upregulate activation of p65:p50 dimers in CNS cells and enhance transactivation of pathological mediators that cause neuroinflammation and neurodegeneration. Hence selective targeting of activated p65 is an attractive therapeutic strategy for AD. Here we report the design, structural and functional characterization of peptide analogs of a p65 interacting protein, the glucocorticoid induced leucine zipper (GILZ). By virtue of binding the transactivation domain of p65 exposed after release from the inhibitory IκΒ proteins in activated cells, the GILZ analogs can act as highly selective inhibitors of activated p65 with minimal potential for off-target effects.
Collapse
|
25
|
Progress in drug development for Alzheimer's disease: An overview in relation to mitochondrial energy metabolism. Eur J Med Chem 2016; 121:774-784. [DOI: 10.1016/j.ejmech.2016.03.084] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2015] [Revised: 01/21/2016] [Accepted: 03/26/2016] [Indexed: 12/27/2022]
|
26
|
Mohamed LA, Keller JN, Kaddoumi A. Role of P-glycoprotein in mediating rivastigmine effect on amyloid-β brain load and related pathology in Alzheimer's disease mouse model. BIOCHIMICA ET BIOPHYSICA ACTA 2016; 1862:778-787. [PMID: 26780497 PMCID: PMC4788561 DOI: 10.1016/j.bbadis.2016.01.013] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2015] [Revised: 01/13/2016] [Accepted: 01/14/2016] [Indexed: 01/01/2023]
Abstract
Recently, we showed that rivastigmine decreased amyloid-β (Aβ) brain load in aged rats by enhancing its clearance across the blood-brain barrier (BBB) via upregulation of P-glycoprotein (P-gp) and low-density lipoprotein receptor-related protein 1 (LRP1). Here, we extend our previous work to clarify P-gp role in mediating rivastigmine effect on Aβ brain levels and neuroprotection in a mouse model of Alzheimer's disease (AD) that expresses different levels of P-gp. APPSWE mice were bred with mdr1a/b knockout mice to produce littermates that were divided into three groups; APP(+)/mdr1(+/+), APP(+)/mdr1(+/-) and APP(+)/mdr1(-/-). Animals received rivastigmine treatment (0.3mg/kg/day) or vehicle for 8weeks using Alzet osmotic mini-pumps. ELISA analysis of brain homogenates for Aβ showed rivastigmine treatment to significantly decrease Aβ brain load in APP(+)/mdr1(+/+) by 25% and in APP(+)/mdr1(+/-) mice by 21% compared to their vehicle treated littermates, but not in APP(+)/mdr1(-/-) mice. In addition, rivastigmine reduced GFAP immunostaining of astrocytes by 50% and IL-1β brain level by 43% in APP(+)/mdr1(+/+) mice, however its effect was less pronounced in P-gp knockout mice. Moreover, rivastigmine demonstrated a P-gp expression dependent neuroprotective effect that was highest in APP(+)/mdr1(+/+)>APP(+)/mdr1(+/-)>APP(+)/mdr1(-/-) as determined by expression of synaptic markers PSD-95 and SNAP-25 using Western blot analysis. Collectively, our results suggest that P-gp plays important role in mediating rivastigmine non-cholinergic beneficial effects, including Aβ brain load reduction, neuroprotective and anti-inflammatory effects in the AD mouse models.
Collapse
Affiliation(s)
- Loqman A Mohamed
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, LA 71201, United States
| | - Jeffrey N Keller
- Pennington Biomedical Research Center, Louisiana State University, Baton Rouge, LA 70808, United States
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, LA 71201, United States.
| |
Collapse
|
27
|
Mohamed LA, Qosa H, Kaddoumi A. Age-Related Decline in Brain and Hepatic Clearance of Amyloid-Beta is Rectified by the Cholinesterase Inhibitors Donepezil and Rivastigmine in Rats. ACS Chem Neurosci 2015; 6:725-36. [PMID: 25782004 PMCID: PMC5248655 DOI: 10.1021/acschemneuro.5b00040] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In Alzheimer's disease (AD), accumulation of brain amyloid-β (Aβ) depends on imbalance between production and clearance of Aβ. Several pathways for Aβ clearance have been reported including transport across the blood-brain barrier (BBB) and hepatic clearance. The incidence of AD increases with age and failure of Aβ clearance correlates with AD. The cholinesterase inhibitors (ChEIs) donepezil and rivastigmine are used to ease the symptoms of dementia associated with AD. Besides, both drugs have been reported to provide neuroprotective and disease-modifying effects. Here, we investigated the effect of ChEIs on age-related reduced Aβ clearance. Findings from in vitro and in vivo studies demonstrated donepezil and rivastigmine to enhance (125)I-Aβ40 clearance. Also, the increase in brain and hepatic clearance of (125)I-Aβ40 was more pronounced in aged compared to young rats, and was associated with significant reduction in brain Aβ endogenous levels determined by ELISA. Furthermore, the enhanced clearance was concomitant with up-regulation in the expression of Aβ major transport proteins P-glycoprotein and LRP1. Collectively, our findings that donepezil and rivastigmine enhance Aβ clearance across the BBB and liver are novel and introduce an additional mechanism by which both drugs could affect AD pathology. Thus, optimizing their clinical use could help future drug development by providing new drug targets and possible mechanisms involved in AD pathology.
Collapse
Affiliation(s)
- Loqman A. Mohamed
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, Louisiana 71201, United States
| | - Hisham Qosa
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, Louisiana 71201, United States
| | - Amal Kaddoumi
- Department of Basic Pharmaceutical Science, School of Pharmacy, University of Louisiana at Monroe, 1800 Bienville Dr., Monroe, Louisiana 71201, United States
| |
Collapse
|
28
|
Abstract
Antiretroviral therapy extends the lifespan of human immunodeficiency virus (HIV)-infected patients, but many survivors develop premature impairments in cognition. These residual cognitive impairments may involve aberrant deposition of amyloid β-peptides (Aβ). By unknown mechanisms, Aβ accumulates in the lysosomal and autophagic compartments of neurons in the HIV-infected brain. Here we identify the molecular events evoked by the HIV coat protein gp120 that facilitate the intraneuronal accumulation of Aβ. We created a triple transgenic gp120/APP/PS1 mouse that recapitulates intraneuronal deposition of Aβ in a manner reminiscent of the HIV-infected brain. In cultured neurons, we found that the HIV coat protein gp120 increased the transcriptional expression of BACE1 through repression of PPARγ, and increased APP expression by promoting interaction of the translation-activating RBP heterogeneous nuclear ribonucleoprotein C with APP mRNA. APP and BACE1 were colocalized into stabilized membrane microdomains, where the β-cleavage of APP and Aβ formation were enhanced. Aβ-peptides became localized to lysosomes that were engorged with sphingomyelin and calcium. Stimulating calcium efflux from lysosomes with a TRPM1 agonist promoted calcium efflux, luminal acidification, and cleared both sphingomyelin and Aβ from lysosomes. These findings suggest that therapeutics targeted to reduce lysosomal pH in neurodegenerative conditions may protect neurons by facilitating the clearance of accumulated sphingolipids and Aβ-peptides.
Collapse
|
29
|
Erickson CA, Ray B, Maloney B, Wink LK, Bowers K, Schaefer TL, McDougle CJ, Sokol DK, Lahiri DK. Impact of acamprosate on plasma amyloid-β precursor protein in youth: a pilot analysis in fragile X syndrome-associated and idiopathic autism spectrum disorder suggests a pharmacodynamic protein marker. J Psychiatr Res 2014; 59:220-8. [PMID: 25300441 PMCID: PMC4253657 DOI: 10.1016/j.jpsychires.2014.07.011] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/09/2014] [Accepted: 07/14/2014] [Indexed: 12/20/2022]
Abstract
BACKGROUND Understanding of the pathophysiology of autism spectrum disorder (ASD) remains limited. Brain overgrowth has been hypothesized to be associated with the development of ASD. A derivative of amyloid-β precursor protein (APP), secreted APPα (sAPPα), has neuroproliferative effects and has been shown to be elevated in the plasma of persons with ASD compared to control subjects. Reduction in sAPPα holds promise as a novel molecular target of treatment in ASD. Research into the neurochemistry of ASD has repeatedly implicated excessive glutamatergic and deficient GABAergic neurotransmission in the disorder. With this in mind, acamprosate, a novel modulator of glutamate and GABA function, has been studied in ASD. No data is available on the impact of glutamate or GABA modulation on sAPPα function. METHODS Plasma APP derivative levels pre- and post-treatment with acamprosate were determined in two pilot studies involving youth with idiopathic and fragile X syndrome (FXS)-associated ASD. We additionally compared baseline APP derivative levels between youth with FXS-associated or idiopathic ASD. RESULTS Acamprosate use was associated with a significant reduction in plasma sAPP(total) and sAPPα levels but no change occurred in Aβ40 or Aβ42 levels in 15 youth with ASD (mean age: 11.1 years). Youth with FXS-associated ASD (n = 12) showed increased sAPPα processing compared to age-, gender- and IQ-match youth with idiopathic ASD (n = 11). CONCLUSIONS Plasma APP derivative analysis holds promise as a potential biomarker for use in ASD targeted treatment. Reduction in sAPP (total) and sAPPα may be a novel pharmacodynamic property of acamprosate. Future study is required to address limitations of the current study to determine if baseline APP derivative analysis may predict subgroups of persons with idiopathic or FXS-associated ASD who may respond best to acamprosate or to potentially other modulators of glutamate and/or GABA neurotransmission.
Collapse
Affiliation(s)
| | - Balmiki Ray
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Bryan Maloney
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Logan K. Wink
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Katherine Bowers
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Tori L. Schaefer
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Christopher J. McDougle
- Lurie Center for Autism, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Deborah K. Sokol
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Debomoy K. Lahiri
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA,Corresponding Author: Debomoy K. Lahiri, Ph.D., Professor, Departments of Psychiatry and of Medical & Molecular Genetics, Indiana University School of Medicine, Institute of Psychiatric Research, Neuroscience Research Building, 320 West 15th Street, NB 200C, Indianapolis, IN 46202-2266, USA, Tel: (317) 274-2706; Fax: (317) 231-0200
| |
Collapse
|
30
|
Grothe MJ, Ewers M, Krause B, Heinsen H, Teipel SJ. Basal forebrain atrophy and cortical amyloid deposition in nondemented elderly subjects. Alzheimers Dement 2014; 10:S344-53. [PMID: 24418052 PMCID: PMC4092050 DOI: 10.1016/j.jalz.2013.09.011] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2013] [Revised: 08/09/2013] [Accepted: 09/04/2013] [Indexed: 02/05/2023]
Abstract
BACKGROUND Both neurodegeneration of the cholinergic basal forebrain (BF) and deposition of β-amyloid are early events in the course of Alzheimer's disease (AD). Associations between increased amyloid pathology and cholinergic atrophy have been described in autopsy studies. METHODS We used structural MRI and AV45-PET amyloid imaging data of 225 cognitively normal or mildly impaired elderly subjects from the Alzheimer's Disease Neuroimaging Initiative to assess in vivo associations between BF atrophy and cortical amyloid deposition. Associations were examined using region-of-interest (ROI) and voxel-based approaches with reference to cytoarchitectonic mappings of the cholinergic BF nuclei. RESULTS ROI- and voxel-based approaches yielded complementary evidence for an association between BF volume and cortical amyloid deposition in presymptomatic and predementia stages of AD, irrespective of age, gender, and APOE genotype. CONCLUSIONS The observed correlations between BF atrophy and cortical amyloid load likely reflect associations between cholinergic degeneration and amyloid pathology as reported in neuropathologic examination studies.
Collapse
Affiliation(s)
- Michel J Grothe
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany.
| | - Michael Ewers
- Institute for Stroke and Dementia Research, Klinikum Grosshadern, Ludwig-Maximilians University, Munich, Germany
| | - Bernd Krause
- Department of Nuclear Medicine, University of Rostock, Rostock, Germany
| | - Helmut Heinsen
- Laboratory of Morphological Brain Research, Department of Psychiatry, University of Würzburg, Würtzburg, Germany
| | - Stefan J Teipel
- German Center for Neurodegenerative Diseases (DZNE), Rostock, Germany; Department of Psychosomatic Medicine, University of Rostock, Rostock, Germany
| |
Collapse
|
31
|
Ray B, Chopra N, Long JM, Lahiri DK. Human primary mixed brain cultures: preparation, differentiation, characterization and application to neuroscience research. Mol Brain 2014; 7:63. [PMID: 25223359 PMCID: PMC4181361 DOI: 10.1186/s13041-014-0063-0] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2014] [Accepted: 08/15/2014] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Culturing primary cortical neurons is an essential neuroscience technique. However, most cultures are derived from rodent brains and standard protocols for human brain cultures are sparse. Herein, we describe preparation, maintenance and major characteristics of a primary human mixed brain culture, including neurons, obtained from legally aborted fetal brain tissue. This approach employs standard materials and techniques used in the preparation of rodent neuron cultures, with critical modifications. RESULTS This culture has distinct differences from rodent cultures. Specifically, a significant numbers of cells in the human culture are derived from progenitor cells, and the yield and survival of the cells grossly depend on the presence of bFGF. In the presence of bFGF, this culture can be maintained for an extended period. Abundant productions of amyloid-β, tau and proteins make this a powerful model for Alzheimer's research. The culture also produces glia and different sub-types of neurons. CONCLUSION We provide a well-characterized methodology for human mixed brain cultures useful to test therapeutic agents under various conditions, and to carry forward mechanistic and translational studies for several brain disorders.
Collapse
|
32
|
Counts SE, Ray B, Mufson EJ, Perez SE, He B, Lahiri DK. Intravenous immunoglobulin (IVIG) treatment exerts antioxidant and neuropreservatory effects in preclinical models of Alzheimer's disease. J Clin Immunol 2014; 34 Suppl 1:S80-5. [PMID: 24760109 PMCID: PMC4293701 DOI: 10.1007/s10875-014-0020-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 03/19/2014] [Indexed: 12/23/2022]
Abstract
Intravenous immunoglobulin (IVIG) has shown limited promise so far in human clinical studies on Alzheimer's disease (AD), yet overwhelmingly positive preclinical work in animals and human brain cultures support the notion that the therapy remains potentially efficacious. Here, we elaborate on IVIG neuropreservation by demonstrating that IVIG protects human primary neurons against oxidative stress in vitro and that IVIG preserves antioxidant defense mechanisms in vivo. Based on these results, we propose the following translational impact: If the dosage and treatment conditions are adequately optimized, then IVIG treatment could play a significant role in preventing and/or delaying the progression of neurodegenerative diseases, such as AD. We suggest that IVIG warrants further investigation to fully exploit its potential as an anti-oxidant, neuroprotective and synapto-protecting agent.
Collapse
Affiliation(s)
- Scott E Counts
- Department of Translational Science and Molecular Medicine, Michigan State University, 333 Bostwick Ave NE, Grand Rapids, MI, 49503, USA,
| | | | | | | | | | | |
Collapse
|
33
|
Durand D, Carniglia L, Beauquis J, Caruso C, Saravia F, Lasaga M. Astroglial mGlu3 receptors promote alpha-secretase-mediated amyloid precursor protein cleavage. Neuropharmacology 2014; 79:180-9. [DOI: 10.1016/j.neuropharm.2013.11.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/26/2013] [Accepted: 11/20/2013] [Indexed: 12/21/2022]
|
34
|
Weyer SW, Zagrebelsky M, Herrmann U, Hick M, Ganss L, Gobbert J, Gruber M, Altmann C, Korte M, Deller T, Müller UC. Comparative analysis of single and combined APP/APLP knockouts reveals reduced spine density in APP-KO mice that is prevented by APPsα expression. Acta Neuropathol Commun 2014; 2:36. [PMID: 24684730 PMCID: PMC4023627 DOI: 10.1186/2051-5960-2-36] [Citation(s) in RCA: 70] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Accepted: 03/07/2014] [Indexed: 11/21/2022] Open
Abstract
Synaptic dysfunction and synapse loss are key features of Alzheimer's pathogenesis. Previously, we showed an essential function of APP and APLP2 for synaptic plasticity, learning and memory. Here, we used organotypic hippocampal cultures to investigate the specific role(s) of APP family members and their fragments for dendritic complexity and spine formation of principal neurons within the hippocampus. Whereas CA1 neurons from APLP1-KO or APLP2-KO mice showed normal neuronal morphology and spine density, APP-KO mice revealed a highly reduced dendritic complexity in mid-apical dendrites. Despite unaltered morphology of APLP2-KO neurons, combined APP/APLP2-DKO mutants showed an additional branching defect in proximal apical dendrites, indicating redundancy and a combined function of APP and APLP2 for dendritic architecture. Remarkably, APP-KO neurons showed a pronounced decrease in spine density and reductions in the number of mushroom spines. No further decrease in spine density, however, was detectable in APP/APLP2-DKO mice. Mechanistically, using APPsα-KI mice lacking transmembrane APP and expressing solely the secreted APPsα fragment we demonstrate that APPsα expression alone is sufficient to prevent the defects in spine density observed in APP-KO mice. Collectively, these studies reveal a combined role of APP and APLP2 for dendritic architecture and a unique function of secreted APPs for spine density.
Collapse
Affiliation(s)
- Sascha W Weyer
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| | - Marta Zagrebelsky
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Spielmannstr. 7, Braunschweig D-38106, Germany
| | - Ulrike Herrmann
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Spielmannstr. 7, Braunschweig D-38106, Germany
| | - Meike Hick
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| | - Lennard Ganss
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
- Present address: Department of Applied Tumor Biology, Ruprecht-Karls University Heidelberg, Institute of Pathology, University of Heidelberg, Heidelberg D-69120, Germany
| | - Julia Gobbert
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| | - Morna Gruber
- Goethe University Frankfurt, Institute of Clinical Neuroanatomy, Neuroscience Center, Theodor-Stern-Kai 7, Frankfurt am Main D-60596, Germany
| | - Christine Altmann
- Goethe University Frankfurt, Institute of Clinical Neuroanatomy, Neuroscience Center, Theodor-Stern-Kai 7, Frankfurt am Main D-60596, Germany
| | - Martin Korte
- TU Braunschweig, Zoological Institute, Cellular Neurobiology, Spielmannstr. 7, Braunschweig D-38106, Germany
| | - Thomas Deller
- Goethe University Frankfurt, Institute of Clinical Neuroanatomy, Neuroscience Center, Theodor-Stern-Kai 7, Frankfurt am Main D-60596, Germany
| | - Ulrike C Müller
- Department of Bioinformatics and Functional Genomics, Ruprecht-Karls University Heidelberg, Institute of Pharmacy and Molecular Biotechnology, Im Neuenheimer Feld 364, Heidelberg D-69120, Germany
| |
Collapse
|
35
|
Spalletta G, Caltagirone C, Padovani A, Sorbi S, Attar M, Colombo D, Cravello L. Cognitive and affective changes in mild to moderate Alzheimer's disease patients undergoing switch of cholinesterase inhibitors: a 6-month observational study. PLoS One 2014; 9:e89216. [PMID: 24586603 PMCID: PMC3929703 DOI: 10.1371/journal.pone.0089216] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 01/17/2014] [Indexed: 01/23/2023] Open
Abstract
Patients with Alzheimer’s disease after an initial response to cholinesterase inhibitors may complain a later lack of efficacy. This, in association with incident neuropsychiatric symptoms, may worsen patient quality of life. Thus, the switch to another cholinesterase inhibitor could represent a valid therapeutic strategy. The aim of this study was to investigate the effectiveness of the switch from one to another cholinesterase inhibitor on cognitive and affective symptoms in mild to moderate Alzheimer disease patients. Four hundred twenty-three subjects were included from the EVOLUTION study, an observational, longitudinal, multicentre study conducted on Alzheimer disease patients who switched to different cholinesterase inhibitor due either to lack/loss of efficacy or response, reduced tolerability or poor compliance. All patients underwent cognitive and neuropsychiatric assessments, carried out before the switch (baseline), and at 3 and 6-month follow-up. A significant effect of the different switch types was found on Mini-Mental State Examination score during time, with best effectiveness on mild Alzheimer’s disease patients switching from oral cholinesterase inhibitors to rivastigmine patch. Depressive symptoms, when measured using continuous Neuropsychiatric Inventory values, decreased significantly, while apathy symptoms remained stable over the 6 months after the switch. However, frequency of both depression and apathy, when measured categorically using Neuropsychiatric Inventory cut-off scores, did not change significantly during time. In mild to moderate Alzheimer disease patients with loss of efficacy and tolerability during cholinesterase inhibitor treatment, the switch to another cholinesterase inhibitor may represent an important option for slowing cognitive deterioration. The evidence of apathy stabilization and the positive tendency of depressive symptom improvement should definitively be confirmed in double-blind controlled studies.
Collapse
Affiliation(s)
- Gianfranco Spalletta
- Neuropsychiatry Laboratory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | - Carlo Caltagirone
- Neuropsychiatry Laboratory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy ; Department of System Medicine, University of Rome "Tor Vergata", Rome, Italy
| | - Alessandro Padovani
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Sandro Sorbi
- Department of Neurological and Psychiatric Sciences, University of Florence, Florence, Italy
| | | | | | - Luca Cravello
- Neuropsychiatry Laboratory, Department of Clinical and Behavioral Neurology, IRCCS Santa Lucia Foundation, Rome, Italy
| | | |
Collapse
|
36
|
Lahiri DK, Maloney B, Long JM, Greig NH. Lessons from a BACE1 inhibitor trial: off-site but not off base. Alzheimers Dement 2014; 10:S411-9. [PMID: 24530026 DOI: 10.1016/j.jalz.2013.11.004] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2013] [Revised: 11/04/2013] [Accepted: 11/25/2013] [Indexed: 11/19/2022]
Abstract
Alzheimer's disease (AD) is characterized by formation of neuritic plaque primarily composed of a small filamentous protein called amyloid-β peptide (Aβ). The rate-limiting step in the production of Aβ is the processing of Aβ precursor protein (APP) by β-site APP-cleaving enzyme (BACE1). Hence, BACE1 activity plausibly plays a rate-limiting role in the generation of potentially toxic Aβ within brain and the development of AD, thereby making it an interesting drug target. A phase II trial of the promising LY2886721 inhibitor of BACE1 was suspended in June 2013 by Eli Lilly and Co., due to possible liver toxicity. This outcome was apparently a surprise to the study's team, particularly since BACE1 knockout mice and mice treated with the drug did not show such liver toxicity. Lilly proposed that the problem was not due to LY2886721 anti-BACE1 activity. We offer an alternative hypothesis, whereby anti-BACE1 activity may induce apparent hepatotoxicity through inhibiting BACE1's processing of β-galactoside α-2,6-sialyltransferase I (STGal6 I). In knockout mice, paralogues, such as BACE2 or cathepsin D, could partially compensate. Furthermore, the short duration of animal studies and short lifespan of study animals could mask effects that would require several decades to accumulate in humans. Inhibition of hepatic BACE1 activity in middle-aged humans would produce effects not detectable in mice. We present a testable model to explain the off-target effects of LY2886721 and highlight more broadly that so-called off-target drug effects might actually represent off-site effects that are not necessarily off-target. Consideration of this concept in forthcoming drug design, screening, and testing programs may prevent such failures in the future.
Collapse
Affiliation(s)
- Debomoy K Lahiri
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN, USA.
| | - Bryan Maloney
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Justin M Long
- Laboratory of Molecular Neurogenetics, Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nigel H Greig
- Laboratory of Translational Gerontology, Intramural Research Program, National Institute of Aging, National Institutes of Health, Baltimore, MD, USA
| |
Collapse
|
37
|
Léger GC, Massoud F. Novel disease-modifying therapeutics for the treatment of Alzheimer’s disease. Expert Rev Clin Pharmacol 2014; 6:423-42. [DOI: 10.1586/17512433.2013.811237] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
|
38
|
Cognitive impairments accompanying rodent mild traumatic brain injury involve p53-dependent neuronal cell death and are ameliorated by the tetrahydrobenzothiazole PFT-α. PLoS One 2013; 8:e79837. [PMID: 24312187 PMCID: PMC3842915 DOI: 10.1371/journal.pone.0079837] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Accepted: 10/02/2013] [Indexed: 01/21/2023] Open
Abstract
With parallels to concussive mild traumatic brain injury (mTBI) occurring in humans, anesthetized mice subjected to a single 30 g weight drop mTBI event to the right parietal cortex exhibited significant diffuse neuronal degeneration that was accompanied by delayed impairments in recognition and spatial memory. To elucidate the involvement of reversible p53-dependent apoptosis in this neuronal loss and associated cognitive deficits, mice were subjected to experimental mTBI followed by the systemic administration of the tetrahydrobenzothiazole p53 inactivator, PFT-α, or vehicle. Neuronal loss was quantified immunohistochemically at 72 hr. post-injury by the use of fluoro-Jade B and NeuN within the dentate gyrus on both sides of the brain, and recognition and spatial memory were assessed by novel object recognition and Y-maze paradigms at 7 and 30 days post injury. Systemic administration of a single dose of PFT-α 1 hr. post-injury significantly ameliorated both neuronal cell death and cognitive impairments, which were no different from sham control animals. Cellular studies on human SH-SY5Y cells and rat primary neurons challenged with glutamate excitotoxicity and H2O2 induced oxidative stress, confirmed the ability of PFT-α and a close analog to protect against these TBI associated mechanisms mediating neuronal loss. These studies suggest that p53-dependent apoptotic mechanisms underpin the neuronal and cognitive losses accompanying mTBI, and that these are potentially reversible by p53 inactivation.
Collapse
|
39
|
Yang HQ, Sun ZK, Yang WM, Han HM, Ma JJ, Li W. Effects of rivastigmine on secreted amyloid precursor protein and beta-amyloid secretion in neuroblastoma SK-N-SH cells. NEUROCHEM J+ 2013. [DOI: 10.1134/s181971241303015x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
40
|
Lilja AM, Luo Y, Yu QS, Röjdner J, Li Y, Marini AM, Marutle A, Nordberg A, Greig NH. Neurotrophic and neuroprotective actions of (-)- and (+)-phenserine, candidate drugs for Alzheimer's disease. PLoS One 2013; 8:e54887. [PMID: 23382994 PMCID: PMC3559887 DOI: 10.1371/journal.pone.0054887] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2012] [Accepted: 12/17/2012] [Indexed: 12/11/2022] Open
Abstract
Neuronal dysfunction and demise together with a reduction in neurogenesis are cardinal features of Alzheimer’s disease (AD) induced by a combination of oxidative stress, toxic amyloid-β peptide (Aβ) and a loss of trophic factor support. Amelioration of these was assessed with the Aβ lowering AD experimental drugs (+)-phenserine and (−)-phenserine in neuronal cultures, and actions in mice were evaluated with (+)-phenserine. Both experimental drugs together with the metabolite N1-norphenserine induced neurotrophic actions in human SH-SY5Y cells that were mediated by the protein kinase C (PKC) and extracellular signal–regulated kinases (ERK) pathways, were evident in cells expressing amyloid precursor protein Swedish mutation (APPSWE), and retained in the presence of Aβ and oxidative stress challenge. (+)-Phenserine, together with its (−) enantiomer as well as its N1- and N8-norphenserine and N1,N8-bisnorphenserine metabolites, likewise provided neuroprotective activity against oxidative stress and glutamate toxicity via the PKC and ERK pathways. These neurotrophic and neuroprotective actions were evident in primary cultures of subventricular zone (SVZ) neural progenitor cells, whose neurosphere size and survival were augmented by (+)-phenserine. Translation of these effects in vivo was assessed in wild type and AD APPswe transgenic (Tg2576) mice by doublecortin (DCX) immunohistochemical analysis of neurogenesis in the SVZ, which was significantly elevated by 16 day systemic (+)-phenserine treatment, in the presence of a (+)-phenserine-induced elevation in brain- derived neurotrophic factor (BDNF).
Collapse
Affiliation(s)
- Anna M. Lilja
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail: (AL); (NHG)
| | - Yu Luo
- Department of Neurological Surgery, Case Western Reserve University School of Medicine, Cleveland, Ohio, United States of America
| | - Qian-sheng Yu
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Jennie Röjdner
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yazhou Li
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
| | - Ann M. Marini
- Department of Neurology and Neuroscience Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Amelia Marutle
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Agneta Nordberg
- Alzheimer Neurobiology Center, Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
- Department of Geriatric Medicine, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Nigel H. Greig
- Drug Design and Development Section, Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States of America
- * E-mail: (AL); (NHG)
| |
Collapse
|
41
|
Ismail MF, Elmeshad AN, Salem NAH. Potential therapeutic effect of nanobased formulation of rivastigmine on rat model of Alzheimer's disease. Int J Nanomedicine 2013; 8:393-406. [PMID: 23378761 PMCID: PMC3558309 DOI: 10.2147/ijn.s39232] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Background To sustain the effect of rivastigmine, a hydrophilic cholinesterase inhibitor, nanobased formulations were prepared. The efficacy of the prepared rivastigmine liposomes (RLs) in comparison to rivastigmine solution (RS) was assessed in an aluminium chloride (AlCl3)-induced Alzheimer’s model. Methods Liposomes were prepared by lipid hydration (F1) and heating (F2) methods. Rats were treated with either RS or RLs (1 mg/kg/day) concomitantly with AlCl3 (50 mg/kg/day). Results The study showed that the F1 method produced smaller liposomes (67.51 ± 14.2 nm) than F2 (528.7 ± 15.5 nm), but both entrapped the same amount of the drug (92.1% ± 1.4%). After 6 hours, 74.2% ± 1.5% and 60.8% ± 2.3% of rivastigmine were released from F1 and F2, respectively. Both RLs and RS improved the deterioration of spatial memory induced by AlCl3, with RLs having a superior effect. Further biochemical measurements proved that RS and RLs were able to lower plasma C-reactive protein, homocysteine and asymmetric dimethy-larginine levels. RS significantly attenuated acetylcholinesterase (AChE) activity, whereas Na+/K+-adenosine triphosphatase (ATPase) activity was enhanced compared to the AlCl3-treated animals; however, RLs succeeded in normalization of AChE and Na+/K+ ATPase activities. Gene-expression profile showed that cotreatment with RS to AlCl3-treated rats succeeded in exerting significant decreases in BACE1, AChE, and IL1B gene expression. Normalization of the expression of the aforementioned genes was achieved by coadministration of RLs to AlCl3-treated rats. The profound therapeutic effect of RLs over RS was evidenced by nearly preventing amyloid plaque formation, as shown in the histopathological examination of rat brain. Conclusion RLs could be a potential drug-delivery system for ameliorating Alzheimer’s disease.
Collapse
|
42
|
Alkalay A, Rabinovici GD, Zimmerman G, Agarwal N, Kaufer D, Miller BL, Jagust WJ, Soreq H. Plasma acetylcholinesterase activity correlates with intracerebral β-amyloid load. Curr Alzheimer Res 2013; 10:48-56. [PMID: 23157337 PMCID: PMC3768143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2012] [Revised: 06/13/2012] [Accepted: 06/15/2012] [Indexed: 06/01/2023]
Abstract
BACKGROUND Previous studies have demonstrated alterations in the peripheral cholinergic system in Alzheimer's disease (AD), though results have been inconsistent and not linked to in vivo biomarkers of pathology. We examined the relationship between amyloid-beta (Aβ) plaques and plasma cholinesterase activity in a heterogeneous dementia population. METHODS 29 participants with clinical AD and 35 with non-AD diagnoses underwent positron emission tomography (PET) with the amyloid ligand [11C] PIB and plasma measurements of acetylcholinesterase (AChE) and butyrylcholinesterase (BChE) activity. Multi-linear regression was used to evaluate the relationship between AChE or BChE activity and PIB binding (adjusted for age, sex, apolipoprotein E4 and vascular risk), applying voxel-wise and region of interest (ROI) approaches. AChE activity was further adjusted for cholinesterase inhibitor (ChE-I) use. Global amyloid load was measured using a PIB Index, representing mean tracer binding in frontal, parietal, lateral temporal and cingulate cortex. RESULTS AChE activity was correlated with PIB Index (β=0.39, p < 0.001) and with regional PIB binding in frontal, temporal, parietal and occipital lobes, precuneus and posterior cingulate on both voxel-wise (p < 0.001 uncorrected) and ROI (β=0.26-0.41, p < 0.005) analysis. Correlations remained significant after covarying clinical diagnosis (β=0.42, p=0.001), and among participants naive to ChE-I (β=0.51, p=0.005). No correlation was found between BChE activity and PIB. Among AD participants, disease severity was not correlated with AChE, BChE or PIB Index. CONCLUSION AChE activity in plasma is correlated with brain Aβ load. Activation of the 'anti-inflammatory cholinergic pathway' may provide the link between Aβ plaques and peripheral cholinergic measures.
Collapse
Affiliation(s)
- Adi Alkalay
- Helen Wills Neuroscience Institute, University of California Berkeley, CA, USA
| | | | | | | | | | | | | | | |
Collapse
|
43
|
Lahiri DK, Ray B. Abnormal cerebrospinal fluid (CSF) dynamics in Alzheimer's disease and normal pressure hydrocephalus: CSF-amyloid β precursor protein metabolites as possible biomarkers. Eur J Neurol 2012; 20:211-3. [DOI: 10.1111/j.1468-1331.2012.03832.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
| | - B. Ray
- Department of Psychiatry, Institute of Psychiatric Research; Indiana University School of Medicine; Indianapolis; IN; USA
| |
Collapse
|
44
|
Choi DY, Lee YJ, Lee SY, Lee YM, Lee HH, Choi IS, Oh KW, Han SB, Nam SY, Hong JT. Attenuation of scopolamine-induced cognitive dysfunction by obovatol. Arch Pharm Res 2012; 35:1279-86. [PMID: 22864751 DOI: 10.1007/s12272-012-0719-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2012] [Revised: 02/16/2012] [Accepted: 03/06/2012] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent cause of dementia in the elderly people. The disease is pathologically characterized by extracellular deposition of beta-amyloid peptide (Aβ), cholinergic neurodegeneration and elevation of acetylcholine esterase (AChE) activity in the affected regions. In this study, we investigated the effects of obovatol on memory dysfunction, which was caused by scopolamine. Obovatol (0.2, 0.5 and 1 mg/kg for 7 day) attenuated scopolamine (1 mg/kg, i.p.)-induced amnesia in a dose-dependent manner, as revealed by the Morris water maze test and step-through passive avoidance test. Mechanism studies exhibited that obovatol dose-dependently alleviated scopolamine-induced increase in Aβ generation and β-secretase activity in the cortex and hippocampus. Obovatol also attenuated scopolamine-induced rise in AChE activity in the cortex and hippocampus. Obovatol might rescue scopolamine-mediated impaired learning and memory function by attenuating Aβ accumulation and stabilizing cholinergic neurotransmission, which suggests that the natural compound could be a useful agent for the prevention of the development or progression of AD neurodegeneration.
Collapse
Affiliation(s)
- Dong-Young Choi
- College of Pharmacy, Chungbuk National University, Cheongju 361-763, Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Wennström M, Nielsen HM. Cell adhesion molecules in Alzheimer's disease. Degener Neurol Neuromuscul Dis 2012; 2:65-77. [PMID: 30890880 DOI: 10.2147/dnnd.s19829] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Cell adhesion molecules (CAMs) mediate interactions between cells and their surroundings that are vital to processes controlling for cell survival, activation, migration, and plasticity. However, increasing evidence suggests that CAMs also mediate mechanisms involved in several neurological diseases. This article reviews the current knowledge on the role of CAMs in amyloid-β (Aβ) metabolism, cell plasticity, neuroinflammation, and vascular changes, all of which are considered central to the pathogenesis and progression of Alzheimer's disease (AD). This paper also outlines the possible roles of CAMs in current and novel AD treatment strategies.
Collapse
Affiliation(s)
- Malin Wennström
- Department of Clinical Sciences Malmö, Lund University, Malmö, Sweden,
| | | |
Collapse
|
46
|
Bailey JA, Lahiri DK. Chromatographic separation of reaction products from the choline acetyltransferase and carnitine acetyltransferase assay: differential ChAT and CrAT activity in brain extracts from Alzheimer's disease versus controls. J Neurochem 2012; 122:672-80. [PMID: 22607230 DOI: 10.1111/j.1471-4159.2012.07793.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Choline acetyltransferase (ChAT) catalyzes the reaction between choline and acetylcoenzyme A (AcCoA) to form acetylcholine (ACh) in nerve terminals. ACh metabolism has implications in numerous aspects of physiology and varied disease states, such as Alzheimer's disease. Therefore a specific, sensitive, and reliable method for detecting ChAT enzyme activity is of great utility in a number of situations. Using an existing radionuclide-based enzyme activity assay, we have observed detectable ChAT signals from non-cholinergic cells, suggesting a contaminant in the assay producing an artifactual signal. Previous reports have suggested that L-acetylcarnitine (LAC) contaminates many assays of ChAT activity, because of difficulties in separating LAC from ACh by organic extraction. To determine the source of this hypothesized artifact and to rectify the problem, we have developed a paper chromatography-based assay for the detection of acetylcholine and other contaminating reaction products of this assay, including LAC. Our first goal was to develop a simple and economical method for resolving and verifying the identities of various reaction products or contaminants that could be performed in most laboratories without specialized equipment. Our second goal was to apply this separation method in postmortem human brain tissue samples. Our assay successfully detected several contaminants, especially in assays using brain tissue, and allowed the separation of the intended ACh product from these contaminants. We further demonstrate that this assay can be used to measure carnitine acetyltransferase (CrAT) activity in the same samples, and assays comparing ChAT and CrAT show that CrAT is highly active in neuronal tissues and in neuronal cell cultures relative to ChAT. Thus, the simple chromatography-based assay we describe allows the measurement of specific reaction products separated from contaminants using commonly available and inexpensive materials. Further, we show that ChAT activity is significantly reduced in brain extracts from Alzheimer's disease compared to controls.
Collapse
Affiliation(s)
- Jason A Bailey
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, IN, USA
| | | |
Collapse
|
47
|
Tweedie D, Ferguson RA, Fishman K, Frankola KA, Van Praag H, Holloway HW, Luo W, Li Y, Caracciolo L, Russo I, Barlati S, Ray B, Lahiri DK, Bosetti F, Greig NH, Rosi S. Tumor necrosis factor-α synthesis inhibitor 3,6'-dithiothalidomide attenuates markers of inflammation, Alzheimer pathology and behavioral deficits in animal models of neuroinflammation and Alzheimer's disease. J Neuroinflammation 2012; 9:106. [PMID: 22642825 PMCID: PMC3405480 DOI: 10.1186/1742-2094-9-106] [Citation(s) in RCA: 154] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Accepted: 05/29/2012] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Neuroinflammation is associated with virtually all major neurodegenerative disorders, including Alzheimer's disease (AD). Although it remains unclear whether neuroinflammation is the driving force behind these disorders, compelling evidence implicates its role in exacerbating disease progression, with a key player being the potent proinflammatory cytokine TNF-α. Elevated TNF-α levels are commonly detected in the clinic and animal models of AD. METHODS The potential benefits of a novel TNF-α-lowering agent, 3,6'-dithiothalidomide, were investigated in cellular and rodent models of neuroinflammation with a specific focus on AD. These included central and systemic inflammation induced by lipopolysaccharide (LPS) and Aβ(1-42) challenge, and biochemical and behavioral assessment of 3xTg-AD mice following chronic 3,6'-dithiothaliodmide. RESULTS 3,6'-Dithiothaliodmide lowered TNF-α, nitrite (an indicator of oxidative damage) and secreted amyloid precursor protein (sAPP) levels in LPS-activated macrophage-like cells (RAW 264.7 cells). This translated into reduced central and systemic TNF-α production in acute LPS-challenged rats, and to a reduction of neuroinflammatory markers and restoration of neuronal plasticity following chronic central challenge of LPS. In mice centrally challenged with A(β1-42) peptide, prior systemic 3,6'-dithiothalidomide suppressed Aβ-induced memory dysfunction, microglial activation and neuronal degeneration. Chronic 3,6'-dithiothalidomide administration to an elderly symptomatic cohort of 3xTg-AD mice reduced multiple hallmark features of AD, including phosphorylated tau protein, APP, Aβ peptide and Aβ-plaque number along with deficits in memory function to levels present in younger adult cognitively unimpaired 3xTg-AD mice. Levels of the synaptic proteins, SNAP25 and synaptophysin, were found to be elevated in older symptomatic drug-treated 3xTg-AD mice compared to vehicle-treated ones, indicative of a preservation of synaptic function during drug treatment. CONCLUSIONS Our data suggest a strong beneficial effect of 3,6'-dithiothalidomide in the setting of neuroinflammation and AD, supporting a role for neuroinflammation and TNF-α in disease progression and their targeting as a means of clinical management.
Collapse
Affiliation(s)
- David Tweedie
- Laboratory of Neurosciences, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Tacrine-6-ferulic acid, a novel multifunctional dimer, inhibits amyloid-β-mediated Alzheimer's disease-associated pathogenesis in vitro and in vivo. PLoS One 2012; 7:e31921. [PMID: 22384101 PMCID: PMC3285653 DOI: 10.1371/journal.pone.0031921] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2011] [Accepted: 01/20/2012] [Indexed: 12/13/2022] Open
Abstract
We have previously synthesized a series of hybrid compounds by linking ferulic acid to tacrine as multifunctional agents based on the hypotheses that Alzheimer's disease (AD) generates cholinergic deficiency and oxidative stress. Interestingly, we found that they may have potential pharmacological activities for treating AD. Here we report for the first time that tacrine-6-ferulic acid (T6FA), one of these compounds, can prevent amyloid-β peptide (Aβ)-induced AD-associated pathological changes in vitro and in vivo. Our results showed that T6FA significantly inhibited auto- and acetylcholinesterase (AChE)-induced aggregation of Aβ1–40in vitro and blocked the cell death induced by Aβ1–40 in PC12 cells. In an AD mouse model by the intracerebroventricular injection of Aβ1–40, T6FA significantly improved the cognitive ability along with increasing choline acetyltransferase and superoxide dismutase activity, decreasing AChE activity and malondialdehyde level. Based on our findings, we conclude that T6FA may be a promising multifunctional drug candidate for AD.
Collapse
|
49
|
Long JM, Lahiri DK. Advances in microRNA experimental approaches to study physiological regulation of gene products implicated in CNS disorders. Exp Neurol 2012; 235:402-18. [PMID: 22245616 DOI: 10.1016/j.expneurol.2011.12.043] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2011] [Revised: 12/13/2011] [Accepted: 12/25/2011] [Indexed: 11/28/2022]
Abstract
The central nervous system (CNS) is a remarkably complex organ system, requiring an equally complex network of molecular pathways controlling the multitude of diverse, cellular activities. Gene expression is a critical node at which regulatory control of molecular networks is implemented. As such, elucidating the various mechanisms employed in the physiological regulation of gene expression in the CNS is important both for establishing a reference for comparison to the diseased state and for expanding the set of validated drug targets available for disease intervention. MicroRNAs (miRNAs) are an abundant class of small RNA that mediates potent inhibitory effects on global gene expression. Recent advances have been made in methods employed to study the contribution of these miRNAs to gene expression. Here we review these latest advances and present a methodological workflow from the perspective of an investigator studying the physiological regulation of a gene of interest. We discuss methods for identifying putative miRNA target sites in a transcript of interest, strategies for validating predicted target sites, assays for detecting miRNA expression, and approaches for disrupting endogenous miRNA function. We consider both advantages and limitations, highlighting certain caveats that inform the suitability of a given method for a specific application. Through careful implementation of the appropriate methodologies discussed herein, we are optimistic that important discoveries related to miRNA participation in CNS physiology and dysfunction are on the horizon.
Collapse
Affiliation(s)
- Justin M Long
- Department of Psychiatry, Institute of Psychiatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | | |
Collapse
|