1
|
Riffo-Lepe N, González-Sanmiguel J, Armijo-Weingart L, Saavedra-Sieyes P, Hernandez D, Ramos G, San Martín LS, Aguayo LG. Synaptic and synchronic impairments in subcortical brain regions associated with early non-cognitive dysfunction in Alzheimer's disease. Neural Regen Res 2025; 21:01300535-990000000-00688. [PMID: 39885666 PMCID: PMC12094569 DOI: 10.4103/nrr.nrr-d-24-01052] [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: 09/08/2024] [Revised: 11/16/2024] [Accepted: 12/21/2024] [Indexed: 02/01/2025] Open
Abstract
ABSTRACT For many decades, Alzheimer's disease research has primarily focused on impairments within cortical and hippocampal regions, which are thought to be related to cognitive dysfunctions such as memory and language deficits. The exact cause of Alzheimer's disease is still under debate, making it challenging to establish an effective therapy or early diagnosis. It is widely accepted that the accumulation of amyloid-beta peptide in the brain parenchyma leads to synaptic dysfunction, a critical step in Alzheimer's disease development. The traditional amyloid cascade model is initiated by accumulating extracellular amyloid-beta in brain areas essential for memory and language. However, while it is possible to reduce the presence of amyloid-beta plaques in the brain with newer immunotherapies, cognitive symptoms do not necessarily improve. Interestingly, recent studies support the notion that early alterations in subcortical brain regions also contribute to brain damage and precognitive decline in Alzheimer's disease. A body of recent evidence suggests that early Alzheimer's disease is associated with alterations (e.g., motivation, anxiety, and motor impairment) in subcortical areas, such as the striatum and amygdala, in both human and animal models. Also, recent data indicate that intracellular amyloid-beta appears early in subcortical regions such as the nucleus accumbens, locus coeruleus, and raphe nucleus, even without extracellular amyloid plaques. The reported effects are mainly excitatory, increasing glutamatergic transmission and neuronal excitability. In agreement, data in Alzheimer's disease patients and animal models show an increase in neuronal synchronization that leads to electroencephalogram disturbances and epilepsy. The data indicate that early subcortical brain dysfunctions might be associated with non-cognitive symptoms such as anxiety, irritability, and motivation deficits, which precede memory loss and language alterations. Overall, the evidence reviewed suggests that subcortical brain regions could explain early dysfunctions and perhaps be targets for therapies to slow disease progression. Future research should focus on these non-traditional brain regions to reveal early pathological alterations and underlying mechanisms to advance our understanding of Alzheimer's disease beyond the traditionally studied hippocampal and cortical circuits.
Collapse
Affiliation(s)
- Nicolás Riffo-Lepe
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Juliana González-Sanmiguel
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Lorena Armijo-Weingart
- Facultad de Odontología y Ciencias de la Rehabilitación, Universidad San Sebastián, Concepción, Chile
| | - Paulina Saavedra-Sieyes
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - David Hernandez
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Gerson Ramos
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
| | - Loreto S. San Martín
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| | - Luis G. Aguayo
- Laboratorio de Neurofisiología, Departamento de Fisiología, Universidad de Concepción, Concepción, Chile
- Programa de Neurociencia, Psiquiatría y Salud Mental (NEPSAM), Universidad de Concepción, Concepción, Chile
| |
Collapse
|
2
|
Xu YQ, Chen Y, Xing JX, Yao J. Relationship between enriched environment and neurodegeneration: a review from mechanism to therapy. Clin Epigenetics 2025; 17:13. [PMID: 39849536 PMCID: PMC11761206 DOI: 10.1186/s13148-025-01820-4] [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: 12/03/2024] [Accepted: 01/17/2025] [Indexed: 01/25/2025] Open
Abstract
Enriched environment (EE), as a non-pharmacological intervention, has garnered considerable attention for its potential to ameliorate neurodegenerative diseases (NDs). This review delineated the impact of EE on the biological functions associated with NDs, emphasizing its role in enhancing neural plasticity, reducing inflammation, and bolstering cognitive performance. We discussed the molecular underpinnings of the effects of EE, including modulation of key signaling pathways such as extracellular regulated kinase 1/2 (ERK1/2), mitogen-activated protein kinases (MAPK), and AMPK/SIRT1, which were implicated in neuroprotection and synaptic plasticity. Additionally, we scrutinized the influence of EE on epigenetic modifications and autophagy, processes pivotal to ND pathogenesis. Animal models, encompassing both rodents and larger animals, offer insights into the disease-modifying effects of EE, underscoring its potential as a complementary approach to pharmacological interventions. In summary, EE emerges as a promising strategy to augment cognitive function and decelerate the progression of NDs.
Collapse
Affiliation(s)
- Yuan-Qiao Xu
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China
- Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning Province, People's Republic of China
- China Medical University Center of Forensic Investigation, Shenbei New District, No.77, Puhe Road, Shenyang, 110122, People's Republic of China
| | - Yanjiao Chen
- Shanxi Provincial People's Hospital, Taiyuan, People's Republic of China
| | - Jia-Xin Xing
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China.
- Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning Province, People's Republic of China.
- China Medical University Center of Forensic Investigation, Shenbei New District, No.77, Puhe Road, Shenyang, 110122, People's Republic of China.
| | - Jun Yao
- School of Forensic Medicine, China Medical University, Shenyang, People's Republic of China.
- Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang, Liaoning Province, People's Republic of China.
- China Medical University Center of Forensic Investigation, Shenbei New District, No.77, Puhe Road, Shenyang, 110122, People's Republic of China.
| |
Collapse
|
3
|
Rao SP, Imam-Fulani AO, Xie W, Phillip S, Chennavajula K, Lind EB, Zhang Y, Vince R, Lee MK, More SS. Oral prodrug of a novel glutathione surrogate reverses metabolic dysregulation and attenuates neurodegenerative process in APP/PS1 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633247. [PMID: 39868172 PMCID: PMC11761491 DOI: 10.1101/2025.01.15.633247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Glycation-induced oxidative stress underlies the numerous metabolic ravages of Alzheimer's disease (AD). Reduced glutathione levels in AD lead to increased oxidative stress, including glycation-induced pathology. Previously, we showed that the accumulation of reactive 1,2-dicarbonyls such as methylglyoxal, the major precursor of non-enzymatic glycation products, was reduced by the increased function of GSH-dependent glyoxalase-1 enzyme in the brain. In this two-pronged study, we evaluate the therapeutic efficacy of an orally bioavailable prodrug of our lead glyoxalase substrate, pro-ψ-GSH, for the first time in a transgenic Alzheimer's disease mouse model. This prodrug delivers pharmacodynamically relevant brain concentrations of ψ-GSH upon oral delivery. Chronic oral dosing of pro-ψ-GSH effectively reverses the cognitive decline observed in the APP/PS1 mouse model. The prodrug successfully mirrors the robust effects of the parent drug i.e., reducing amyloid pathology, glycation stress, neuroinflammation, and the resultant neurodegeneration in these mice. We also report the first metabolomics study of such a treatment, which yields key biomarkers linked to the reversal of AD-related metabolic dysregulation. Collectively, this study establishes pro-ψ-GSH as a viable, disease-modifying therapy for AD and paves the way for further preclinical advancement of such therapeutics. Metabolomic signatures identified could prove beneficial in the development of treatment-specific clinically translatable biomarkers.
Collapse
Affiliation(s)
- Swetha Pavani Rao
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Aminat O. Imam-Fulani
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Wei Xie
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Samuel Phillip
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Krishna Chennavajula
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Erin B. Lind
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ying Zhang
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN, USA
| | - Robert Vince
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
- Institute for Translational Neuroscience, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Swati S. More
- Center for Drug Design, College of Pharmacy, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
4
|
Chen J, Zou C, Guan H, Zhou X, Hou L, Cui Y, Xu J, Luan P, Zheng D. Caloric restriction leading to attenuation of experimental Alzheimer's disease results from alterations in gut microbiome. CNS Neurosci Ther 2024; 30:e14823. [PMID: 38992870 PMCID: PMC11239325 DOI: 10.1111/cns.14823] [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: 03/13/2024] [Revised: 05/05/2024] [Accepted: 06/17/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Caloric restriction (CR) might be effective for alleviating/preventing Alzheimer's disease (AD), but the biological mechanisms remain unclear. In the current study, we explored whether CR caused an alteration of gut microbiome and resulted in the attenuation of cognitive impairment of AD animal model. METHODS Thirty-week-old male APP/PS1 transgenic mice were used as AD models (AD mouse). CR was achieved by 30% reduction of daily free feeding (ad libitum, AL) amount. The mice were fed with CR protocol or AL protocol for six consecutive weeks. RESULTS We found that with CR treatment, AD mice showed improved ability of learning and spatial memory, and lower levels of Aβ40, Aβ42, IL-1β, TNF-α, and ROS in the brain. By sequencing 16S rDNA, we found that CR treatment resulted in significant diversity in composition and abundance of gut flora. At the phylum level, Deferribacteres (0.04%), Patescibacteria (0.14%), Tenericutes (0.03%), and Verrucomicrobia (0.5%) were significantly decreased in CR-treated AD mice; at the genus level, Dubosiella (10.04%), Faecalibaculum (0.04%), and Coriobacteriaceae UCG-002 (0.01%) were significantly increased in CR-treated AD mice by comparing with AL diet. CONCLUSIONS Our results demonstrate that the attenuation of AD following CR treatment in APP/PS1 mice may result from alterations in the gut microbiome. Thus, gut flora could be a new target for AD prevention and therapy.
Collapse
Affiliation(s)
- Junyu Chen
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Cong Zou
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Hongbing Guan
- Guangdong Yunzhao Medical Technology Co., Ltd.GuangzhouChina
| | - Xiaoming Zhou
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Le Hou
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Yayong Cui
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Junhua Xu
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| | - Ping Luan
- School of Basic Medical SciencesShenzhen UniversityShenzhenChina
| | - Dong Zheng
- Department of Neurology, The Affiliated Brain HospitalGuangzhou Medical UniversityGuangzhouChina
| |
Collapse
|
5
|
The Molecular Effects of Environmental Enrichment on Alzheimer's Disease. Mol Neurobiol 2022; 59:7095-7118. [PMID: 36083518 PMCID: PMC9616781 DOI: 10.1007/s12035-022-03016-w] [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: 04/20/2022] [Accepted: 08/23/2022] [Indexed: 12/02/2022]
Abstract
Environmental enrichment (EE) is an environmental paradigm encompassing sensory, cognitive, and physical stimulation at a heightened level. Previous studies have reported the beneficial effects of EE in the brain, particularly in the hippocampus. EE improves cognitive function as well as ameliorates depressive and anxiety-like behaviors, making it a potentially effective neuroprotective strategy against neurodegenerative diseases such as Alzheimer's disease (AD). Here, we summarize the current evidence for EE as a neuroprotective strategy as well as the potential molecular pathways that can explain the effects of EE from a biochemical perspective using animal models. The effectiveness of EE in enhancing brain activity against neurodegeneration is explored with a view to differences present in early and late life EE exposure, with its potential application in human being discussed. We discuss EE as one of the non pharmacological approaches in preventing or delaying the onset of AD for future research.
Collapse
|
6
|
Mate V, Smolek T, Kazmerova ZV, Jadhav S, Brezovakova V, Jurkanin B, Uhrinova I, Basheer N, Zilka N, Katina S, Novak P. Enriched environment ameliorates propagation of tau pathology and improves cognition in rat model of tauopathy. Front Aging Neurosci 2022; 14:935973. [PMID: 35966785 PMCID: PMC9363241 DOI: 10.3389/fnagi.2022.935973] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 07/06/2022] [Indexed: 11/13/2022] Open
Abstract
Introduction The typical symptoms of Alzheimer's disease (AD) are cognitive impairment, disrupted spatial orientation, behavioral and psychiatric abnormalities, and later motor deficits. Neuropathologically, AD is characterized by deposits of pathological forms of endogenous proteins - amyloid-β, and neurofibrillary tau protein pathology. The latter closely correlates with brain atrophy and clinical impairment. Pharmacological therapies for these pathologies are largely absent, raising the question whether non-pharmacological interventions could be efficacious. Environmental factors can play a role in the manifestation of AD. It is unknown whether enriched environment (EE) can ameliorate the propagation of protein aggregates or their toxic components. Methods We injected insoluble tau extracts from human brains with AD (600 or 900 ng per animal) into hippocampi of SHR72 transgenic rats that express non-mutated truncated human tau 151-391/4R, but usually do not develop hippocampal tangles. The rats had either standard housing, or could access an EE 5×/week for 3 months. Behavioral analysis included the Morris Water Maze (MWM). Histological analysis was used to assess the propagation of tau pathology. Results Animals exposed to EE performed better in the MWM (spatial acquisition duration and total distance, probe test); unexposed animals improved over the course of acquisition trials, but their mean performance remained below that of the EE group. Enriched environment abrogated tau propagation and hippocampal tangle formation in the 600 ng group; in the 900 ng group, tangle formation was ∼10-fold of the 600 ng group, and unaffected by EE. Conclusion Even a small difference in the amount of injected human AD tau can cause a pronounced difference in the number of resulting tangles. EE leads to a noticeably better spatial navigation performance of tau-injected animals. Furthermore, EE seems to be able to slow down tau pathology progression, indicating the possible utility of similar interventions in early stages of AD where tangle loads are still low.
Collapse
Affiliation(s)
- Veronika Mate
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Tomas Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
- Neuroimunology Institute, n.p.o., Bratislava, Slovakia
| | - Zuzana Vince Kazmerova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Santosh Jadhav
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | | | | | - Ivana Uhrinova
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience R&D Services SE, Bratislava, Slovakia
| | - Stanislav Katina
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Institute of Mathematics and Statistics, Masaryk University, Brno, Czechia
- Axon Neuroscience CRM Services SE, Bratislava, Slovakia
| | - Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, Slovakia
- Axon Neuroscience CRM Services SE, Bratislava, Slovakia
| |
Collapse
|
7
|
Antonovaite N, Hulshof LA, Huffels CFM, Hol EM, Wadman WJ, Iannuzzi D. Mechanical alterations of the hippocampus in the APP/PS1 Alzheimer's disease mouse model. J Mech Behav Biomed Mater 2021; 122:104697. [PMID: 34271406 DOI: 10.1016/j.jmbbm.2021.104697] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 06/26/2021] [Accepted: 07/03/2021] [Indexed: 01/22/2023]
Abstract
There is increasing evidence of altered tissue mechanics in neurodegeneration. However, due to difficulties in mechanical testing procedures and the complexity of the brain, there is still little consensus on the role of mechanics in the onset and progression of neurodegenerative diseases. In the case of Alzheimer's disease (AD), magnetic resonance elastography (MRE) studies have indicated viscoelastic differences in the brain tissue of AD patients and healthy controls. However, there is a lack of viscoelastic data from contact mechanical testing at higher spatial resolution. Therefore, we report viscoelastic maps of the hippocampus obtained by a dynamic indentation on brain slices from the APP/PS1 mouse model where individual brain regions are resolved. A comparison of viscoelastic parameters shows that regions in the hippocampus of the APP/PS1 mice are significantly stiffer than wild-type (WT) mice and have increased viscous dissipation. Furthermore, indentation mapping at the cellular scale directly on the plaques and their surroundings did not show local alterations in stiffness although overall mechanical heterogeneity of the tissue was high (SD∼40%).
Collapse
Affiliation(s)
- Nelda Antonovaite
- Department of Physics and Astronomy and LaserLaB, VU Amsterdam, The Netherlands.
| | - Lianne A Hulshof
- Department of Translational Neuroscience, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Christiaan F M Huffels
- Department of Translational Neuroscience, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Elly M Hol
- Department of Translational Neuroscience, University Medical Center Utrecht, Brain Center, Utrecht University, Utrecht, The Netherlands
| | - Wytse J Wadman
- Center for Neuroscience, Swammerdam Institute for Life Sciences, University of Amsterdam, The Netherlands
| | - Davide Iannuzzi
- Department of Physics and Astronomy and LaserLaB, VU Amsterdam, The Netherlands
| |
Collapse
|
8
|
Nakai T, Yamada K, Mizoguchi H. Alzheimer's Disease Animal Models: Elucidation of Biomarkers and Therapeutic Approaches for Cognitive Impairment. Int J Mol Sci 2021; 22:5549. [PMID: 34074018 PMCID: PMC8197360 DOI: 10.3390/ijms22115549] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 05/21/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related and progressive neurodegenerative disorder. It is widely accepted that AD is mainly caused by the accumulation of extracellular amyloid β (Aβ) and intracellular neurofibrillary tau tangles. Aβ begins to accumulate years before the onset of cognitive impairment, suggesting that the benefit of currently available interventions would be greater if they were initiated in the early phases of AD. To understand the mechanisms of AD pathogenesis, various transgenic mouse models with an accelerated accumulation of Aβ and tau tangles have been developed. However, none of these models exhibit all pathologies present in human AD. To overcome these undesirable phenotypes, APP knock-in mice, which were presented with touchscreen-based tasks, were developed to better evaluate the efficacy of candidate therapeutics in mouse models of early-stage AD. This review assesses several AD mouse models from the aspect of biomarkers and cognitive impairment and discusses their potential as tools to provide novel AD therapeutic approaches.
Collapse
Affiliation(s)
- Tsuyoshi Nakai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
| | - Hiroyuki Mizoguchi
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya 466-8560, Japan; (T.N.); (K.Y.)
- Medical Interactive Research and Academia Industry Collaboration Center, Research Institute of Environmental Medicine, Nagoya University, Nagoya 464-8601, Japan
| |
Collapse
|
9
|
Haider S, Shahzad S, Batool Z, Sadir S, Liaquat L, Tabassum S, Perveen T. Spirulina platensis reduces the schizophrenic-like symptoms in rat model by restoring altered APO-E and RTN-4 protein expression in prefrontal cortex. Life Sci 2021; 277:119417. [PMID: 33794248 DOI: 10.1016/j.lfs.2021.119417] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 03/13/2021] [Accepted: 03/13/2021] [Indexed: 01/27/2023]
Abstract
AIMS Schizophrenia (SZ) is recognized as a neuropsychiatric disorder in humans with accelerated mortality and profound morbidity followed with impairments in social as well as vocational functioning. Though various antipsychotics are being considered as approved treatment therapy for the psychotic symptoms of SZ but they also exert adverse effects and also lack efficacy in treating full spectrum of the disorder. Spirulina platensis (blue-green algae), a nutritional supplement, constitutes a variety of multi-nutrients and possesses a large number of neuroprotective activities. Therefore, present experimental work was designed to evaluate the neuroprotective effects of spirulina in ameliorating the psychosis-like symptoms in dizocilpine-induced rat model of SZ. MATERIALS AND METHODS The spirulina was tested as preventive and therapeutic regimen at the dose of 180 mg/kg. After pre- and post-treatment with spirulina, rats were subjected to behavioral assessments followed by biochemical and neurochemical estimations. Biomarkers including APO-E, RTN-4, TNF-α, and IL-6 were also estimated using ELISA. KEY FINDINGS Present results showed that administration of spirulina not only improved behavioral deficits induced by dizocilpine but it also regulates neurotransmission, oligodendrocyte dysfunction and APO-E over expression. Moreover, it also restores the immune response dysfunction by reducing inflammatory cytokines. SIGNIFICANCE Thus, from present findings it may be suggested that spirulina aids in ameliorating the psychosis-like symptoms induced by dizocilpine in animal model possibly via regulation of neurotransmission and other biomarkers that are extensively used to uncover the etiopathology of SZ. Hence, blue-green algae can be used as an effective therapy for preventive or therapeutic measures in SZ.
Collapse
Affiliation(s)
- Saida Haider
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, Pakistan
| | - Sidrah Shahzad
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, Pakistan; Pakistan Navy Medical Training School and College, PNS Shifa, Karachi, Pakistan
| | - Zehra Batool
- Dr. Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Sciences, University of Karachi, Karachi, Pakistan.
| | - Sadia Sadir
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, Pakistan
| | - Laraib Liaquat
- Multidisciplinary Research Lab, Bahria University Medical and Dental College, Bahria University, Karachi, Pakistan
| | - Saiqa Tabassum
- Department of Biosciences, Faculty of Life Science, Shaheed Zulfqar Ali, Bhutto Institute of Science and Technology (Szabist), Karachi, Pakistan
| | - Tahira Perveen
- Neurochemistry and Biochemical Neuropharmacology Research Unit, Department of Biochemistry, University of Karachi, Karachi, Pakistan
| |
Collapse
|
10
|
Zhang Q, Song Q, Gu X, Zheng M, Wang A, Jiang G, Huang M, Chen H, Qiu Y, Bo B, Tong S, Shao R, Li B, Wang G, Wang H, Hu Y, Chen H, Gao X. Multifunctional Nanostructure RAP-RL Rescues Alzheimer's Cognitive Deficits through Remodeling the Neurovascular Unit. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2021; 8:2001918. [PMID: 33511002 PMCID: PMC7816710 DOI: 10.1002/advs.202001918] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 09/02/2020] [Indexed: 05/21/2023]
Abstract
Cerebrovascular dysfunction characterized by the neurovascular unit (NVU) impairment contributes to the pathogenesis of Alzheimer's disease (AD). In this study, a cerebrovascular-targeting multifunctional lipoprotein-biomimetic nanostructure (RAP-RL) constituted with an antagonist peptide (RAP) of receptor for advanced glycation end-products (RAGE), monosialotetrahexosyl ganglioside, and apolipoprotein E3 is developed to recover the functional NVU and normalize the cerebral vasculature. RAP-RL accumulates along the cerebral microvasculature through the specific binding of RAP to RAGE, which is overexpressed on cerebral endothelial cells in AD. It effectively accelerates the clearance of perivascular Aβ, normalizes the morphology and functions of cerebrovasculature, and restores the structural integrity and functions of NVU. RAP-RL markedly rescues the spatial learning and memory in APP/PS1 mice. Collectively, this study demonstrates the potential of the multifunctional nanostructure RAP-RL as a disease-modifying modality for AD treatment and provides the proof of concept that remodeling the functional NVU may represent a promising therapeutic approach toward effective intervention of AD.
Collapse
Affiliation(s)
- Qian Zhang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Qingxiang Song
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Xiao Gu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Mengna Zheng
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Antian Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Gan Jiang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Meng Huang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Huan Chen
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yu Qiu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Bin Bo
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Shanbao Tong
- School of Biomedical Engineering and Med‐X Research InstituteShanghai Jiao Tong University800 Dongchuan RoadShanghai200240China
| | - Rong Shao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Binyin Li
- Department of Neurology & Neuroscience InstituteRuijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine197 Rui Jin Er RoadShanghai200025China
| | - Gang Wang
- Department of Neurology & Neuroscience InstituteRuijin Hospital affiliated to Shanghai Jiao Tong University School of Medicine197 Rui Jin Er RoadShanghai200025China
| | - Hao Wang
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Yongbo Hu
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| | - Hongzhuan Chen
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
- Institute of Interdisciplinary Integrative Biomedical ResearchShuguang HospitalShanghai University of Traditional Chinese Medicine1200 Cailun RoadShanghai201210China
| | - Xiaoling Gao
- Department of Pharmacology and Chemical BiologyState Key Laboratory of Oncogenes and Related GenesShanghai Universities Collaborative Innovation Center for Translational MedicineShanghai Jiao Tong University School of Medicine280 South Chongqing RoadShanghai200025China
| |
Collapse
|
11
|
Kartalou GI, Salgueiro-Pereira AR, Endres T, Lesnikova A, Casarotto P, Pousinha P, Delanoe K, Edelmann E, Castrén E, Gottmann K, Marie H, Lessmann V. Anti-Inflammatory Treatment with FTY720 Starting after Onset of Symptoms Reverses Synaptic Deficits in an AD Mouse Model. Int J Mol Sci 2020; 21:ijms21238957. [PMID: 33255764 PMCID: PMC7734581 DOI: 10.3390/ijms21238957] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 11/19/2020] [Accepted: 11/21/2020] [Indexed: 12/19/2022] Open
Abstract
Therapeutic approaches providing effective medication for Alzheimer's disease (AD) patients after disease onset are urgently needed. Previous studies in AD mouse models suggested that physical exercise or changed lifestyle can delay AD-related synaptic and memory dysfunctions when treatment started in juvenile animals long before onset of disease symptoms, while a pharmacological treatment that can reverse synaptic and memory deficits in AD mice was thus far not identified. Repurposing food and drug administration (FDA)-approved drugs for treatment of AD is a promising way to reduce the time to bring such medication into clinical practice. The sphingosine-1 phosphate analog fingolimod (FTY720) was approved recently for treatment of multiple sclerosis patients. Here, we addressed whether fingolimod rescues AD-related synaptic deficits and memory dysfunction in an amyloid precursor protein/presenilin-1 (APP/PS1) AD mouse model when medication starts after onset of symptoms (at five months). Male mice received intraperitoneal injections of fingolimod for one to two months starting at five to six months. This treatment rescued spine density as well as long-term potentiation in hippocampal cornu ammonis-1 (CA1) pyramidal neurons, that were both impaired in untreated APP/PS1 animals at six to seven months of age. Immunohistochemical analysis with markers of microgliosis (ionized calcium-binding adapter molecule 1; Iba1) and astrogliosis (glial fibrillary acid protein; GFAP) revealed that our fingolimod treatment regime strongly down regulated neuroinflammation in the hippocampus and neocortex of this AD model. These effects were accompanied by a moderate reduction of Aβ accumulation in hippocampus and neocortex. Our results suggest that fingolimod, when applied after onset of disease symptoms in an APP/PS1 mouse model, rescues synaptic pathology that is believed to underlie memory deficits in AD mice, and that this beneficial effect is mediated via anti-neuroinflammatory actions of the drug on microglia and astrocytes.
Collapse
Affiliation(s)
- Georgia-Ioanna Kartalou
- Institute of Physiology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (G.-I.K.); (T.E.); (E.E.)
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
| | - Ana Rita Salgueiro-Pereira
- Université Côte d’Azur, CNRS, IPMC, UMR7275, 06560 Valbonne, France; (A.R.S.-P.); (P.P.); (K.D.); (H.M.)
| | - Thomas Endres
- Institute of Physiology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (G.-I.K.); (T.E.); (E.E.)
| | - Angelina Lesnikova
- Neuroscience Center, HiLIFE, University of Helsinki, 00290 Helsinki, Finland; (A.L.); (P.C.); (E.C.)
| | - Plinio Casarotto
- Neuroscience Center, HiLIFE, University of Helsinki, 00290 Helsinki, Finland; (A.L.); (P.C.); (E.C.)
| | - Paula Pousinha
- Université Côte d’Azur, CNRS, IPMC, UMR7275, 06560 Valbonne, France; (A.R.S.-P.); (P.P.); (K.D.); (H.M.)
| | - Kevin Delanoe
- Université Côte d’Azur, CNRS, IPMC, UMR7275, 06560 Valbonne, France; (A.R.S.-P.); (P.P.); (K.D.); (H.M.)
| | - Elke Edelmann
- Institute of Physiology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (G.-I.K.); (T.E.); (E.E.)
| | - Eero Castrén
- Neuroscience Center, HiLIFE, University of Helsinki, 00290 Helsinki, Finland; (A.L.); (P.C.); (E.C.)
| | - Kurt Gottmann
- Institute of Neuro- and Sensory Physiology, Medical Faculty, Heinrich Heine University, 40225 Duesseldorf, Germany
- Correspondence: (K.G.); (V.L.)
| | - Hélène Marie
- Université Côte d’Azur, CNRS, IPMC, UMR7275, 06560 Valbonne, France; (A.R.S.-P.); (P.P.); (K.D.); (H.M.)
| | - Volkmar Lessmann
- Institute of Physiology, Medical Faculty, Otto-von-Guericke University, 39120 Magdeburg, Germany; (G.-I.K.); (T.E.); (E.E.)
- Center for Behavioral Brain Sciences (CBBS), 39120 Magdeburg, Germany
- Correspondence: (K.G.); (V.L.)
| |
Collapse
|
12
|
Fulopova B, Stuart KE, Bennett W, Bindoff A, King AE, Vickers JC, Canty AJ. Regional differences in beta amyloid plaque deposition and variable response to midlife environmental enrichment in the cortex of APP/PS1 mice. J Comp Neurol 2020; 529:1849-1862. [PMID: 33104234 DOI: 10.1002/cne.25060] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 01/21/2023]
Abstract
Environmentally enriched housing conditions can increase performance on cognitive tasks in APP/PS1 mice; however, the potential effects of environmental enrichment (EE) on disease modification in terms of pathological change are inconclusive. We hypothesized that previous contrasting findings may be attributable to regional differences in susceptibility to amyloid beta (Aβ) plaque deposition in cortical regions that are functionally associated with EE. We characterized fibrillar plaque deposition in 6, 12, and 18-22 months old APP/PS1 mice in the prefrontal (PFC), somatosensory (SS2), and primary motor cortex (M1). We found a significant increase in plaque load between 6 and 12 months in all regions. In animals over 12 months, only the PFC region continued to significantly accumulate plaques. Additionally, 12 months old animals subjected to 6 months of EE showed improved spatial navigation and had significantly fewer plaques in M1 and SS2, but not in the PFC. These findings suggest that the PFC region is selectively susceptible to Aβ deposition and less responsive to the attenuating effects of EE. In contrast, M1 and SS2 regions plateau with respect to Aβ deposition by 12 months of age and are susceptible to amyloid pathology modification by midlife EE.
Collapse
Affiliation(s)
- Barbora Fulopova
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Kimberley E Stuart
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - William Bennett
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Aidan Bindoff
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Anna E King
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - James C Vickers
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| | - Alison J Canty
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, Tasmania, Australia
| |
Collapse
|
13
|
Zheng JJ, Zou R, Huang S, Song TJ, Yu X. Enriched Environment Rearing from Birth Reduced Anxiety, Improved Learning and Memory, and Promoted Social Interactions in Adult Male Mice. Neuroscience 2020; 442:138-150. [PMID: 32652178 DOI: 10.1016/j.neuroscience.2020.07.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 06/11/2020] [Accepted: 07/02/2020] [Indexed: 12/21/2022]
Abstract
Rearing rodents in an enriched environment (EE), with increased sensory stimulations and social interactions, is a well-established model for naturally increasing neural activity. It is well-known that EE-rearing of rodents from adolescence or during adulthood leads to extensive biochemical, morphological, electrophysiological and behavioral changes. Here, we examine the effects of EE-rearing from birth on adult behavior. Through a battery of assays, we found that mice EE-reared from birth had better acquisition and consolidation of memory, in both aversive-based fear conditioning and reward-based contextual association tasks. Moreover, EE-reared mice showed reduced anxiety in novel environments and enhanced social interactions. Together, these results demonstrated that EE-rearing from birth significantly improved motor ability, learning and memory and sociability, while reducing anxiety. A better understanding of how early environmental influences affect behavior is not only important for understanding neural circuit wiring, but also provides insight into developing more effective intervention programs for neurodevelopmental disorders.
Collapse
Affiliation(s)
- Jing-Jing Zheng
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Rong Zou
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Shajin Huang
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China
| | - Tian-Jia Song
- School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China.
| | - Xiang Yu
- Institute of Neuroscience and State Key Laboratory of Neuroscience, CAS Center for Excellence in Brain Science and Intelligence Technology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai 200031, China; School of Life Sciences, Peking-Tsinghua Center for Life Sciences, and Peking University McGovern Institute, Peking University, Beijing 100871, China.
| |
Collapse
|
14
|
TNFAIP3 Deficiency Affects Monocytes, Monocytes-Derived Cells and Microglia in Mice. Int J Mol Sci 2020; 21:ijms21082830. [PMID: 32325694 PMCID: PMC7215837 DOI: 10.3390/ijms21082830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/07/2020] [Accepted: 04/15/2020] [Indexed: 01/23/2023] Open
Abstract
The intracellular-ubiquitin-ending-enzyme tumor necrosis factor alpha-induced protein 3 (TNFAIP3) is a potent inhibitor of the pro-inflammatory nuclear factor kappa-light-chain-enhancer of activated B cell (NF-kB) pathway. Single nucleotide polymorphisms in TNFAIP3 locus have been associated to autoimmune inflammatory disorders, including Multiple Sclerosis (MS). Previously, we reported a TNFAIP3 down-regulated gene expression level in blood and specifically in monocytes obtained from treatment-naïve MS patients compared to healthy controls (HC). Myeloid cells exert a key role in the pathogenesis of MS. Here we evaluated the effect of specific TNFAIP3 deficiency in myeloid cells including monocytes, monocyte-derived cells (M-MDC) and microglia analyzing lymphoid organs and microglia of mice. TNFAIP3 deletion is induced using conditional knock-out mice for myeloid lineage. Flow-cytometry and histological procedures were applied to assess the immune cell populations of spleen, lymph nodes and bone marrow and microglial cell density in the central nervous system (CNS), respectively. We found that TNFAIP3 deletion in myeloid cells induces a reduction in body weight, a decrease in the number of M-MDC and of common monocyte and granulocyte precursor cells (CMGPs). We also reported that the lack of TNFAIP3 in myeloid cells induces an increase in microglial cell density. The results suggest that TNFAIP3 in myeloid cells critically controls the development of M-MDC in lymphoid organ and of microglia in the CNS.
Collapse
|
15
|
NURR1 deficiency is associated to ADHD-like phenotypes in mice. Transl Psychiatry 2019; 9:207. [PMID: 31455763 PMCID: PMC6712038 DOI: 10.1038/s41398-019-0544-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 06/09/2019] [Accepted: 07/17/2019] [Indexed: 01/10/2023] Open
Abstract
The transcription factor NURR1 regulates the dopamine (DA) signaling pathway and exerts a critical role in the development of midbrain dopaminergic neurons (mDA). NURR1 alterations have been linked to DA-associated brain disorders, such as Parkinson's disease and schizophrenia. However, the association between NURR1 defects and the attention-deficit hyperactivity disorder (ADHD), a DA-associated brain disease characterized by hyperactivity, impulsivity and inattention, has never been demonstrated. To date, a comprehensive murine model of ADHD truly reflecting the whole complex human psychiatric disorder still does not exist. NURR1-knockout (NURR1-KO) mice have been reported to exhibit increased spontaneous locomotor activity, but their complete characterization is still lacking. In the present study a wide-ranging test battery was used to perform a comprehensive analysis of the behavioral phenotype of the male NURR1-KO mice. As a result, their hyperactive phenotype was confirmed, while their impulsive behavior was reported for the first time. On the other hand, no anxiety and alterations in motor coordination, sociability and memory were observed. Also, the number of mDA expressing tyrosine hydroxylase, a rate-limiting enzyme of catecholamines biosynthesis, and DA level in brain were not impaired in NURR1-KO mice. Finally, hyperactivity has been shown to be recovered by treatment with methylphenidate, the first line psychostimulant drug used for ADHD. Overall, our study suggests that the NURR1 deficient male mouse may be a satisfactory model to study some ADHD behavioral phenotypes and to test the clinical efficacy of potential therapeutic agents.
Collapse
|
16
|
Bie B, Wu J, Foss JF, Naguib M. Activation of mGluR1 Mediates C1q-Dependent Microglial Phagocytosis of Glutamatergic Synapses in Alzheimer's Rodent Models. Mol Neurobiol 2019; 56:5568-5585. [PMID: 30652266 DOI: 10.1007/s12035-019-1467-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 01/04/2019] [Indexed: 12/29/2022]
Abstract
Microglia and complements appear to be involved in the synaptic and cognitive deficits in Alzheimer's disease (AD), though the mechanisms remain elusive. In this study, utilizing two types of rodent model of AD, we reported increased complement C1q-mediated microglial phagocytosis of hippocampal glutamatergic synapses, which led to synaptic and cognitive deficits. We also found increased activity of the metabotropic glutamate receptor 1 (mGluR1) in hippocampal CA1 in the modeled rodents. Artificial activation of mGluR1 signaling promoted dephosphorylation of fragile X mental retardation protein (FMRP) and facilitated the local translation machinery of synaptic C1q mRNA, thus mimicking the C1q-mediated microglial phagocytosis of hippocampal glutamatergic synapses and synaptic and cognitive deficiency in the modeled rodents. However, suppression of mGluR1 signaling inhibited the dephosphorylation of FMRP and repressed the local translation of synaptic C1q mRNA, which consequently alleviated microglial phagocytosis of synapses and restored the synaptic and cognitive function in the rodent models. These findings illustrate a novel molecular mechanism underlying C1q-mediated microglial phagocytosis of hippocampal glutamatergic synapses in AD.
Collapse
Affiliation(s)
- Bihua Bie
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH, 44195, USA
| | - Jiang Wu
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH, 44195, USA
| | - Joseph F Foss
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH, 44195, USA
| | - Mohamed Naguib
- Anesthesiology Institute, Cleveland Clinic, 9500 Euclid Ave., Cleveland, OH, 44195, USA. .,Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, 9500 Euclid Ave., Mail Code NB3-78, Cleveland, OH, 44195, USA.
| |
Collapse
|
17
|
Shepherd A, Zhang TD, Zeleznikow-Johnston AM, Hannan AJ, Burrows EL. Transgenic Mouse Models as Tools for Understanding How Increased Cognitive and Physical Stimulation Can Improve Cognition in Alzheimer's Disease. Brain Plast 2018; 4:127-150. [PMID: 30564551 PMCID: PMC6296266 DOI: 10.3233/bpl-180076] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Cognitive decline appears as a core feature of dementia, of which the most prevalent form, Alzheimer's disease (AD) affects more than 45 million people worldwide. There is no cure, and therapeutic options remain limited. A number of modifiable lifestyle factors have been identified that contribute to cognitive decline in dementia. Sedentary lifestyle has emerged as a major modifier and accordingly, boosting mental and physical activity may represent a method to prevent decline in dementia. Beneficial effects of increased physical activity on cognition have been reported in healthy adults, showing potential to harness exercise and cognitive stimulation as a therapy in dementia. 'Brain training' (cognitive stimulation) has also been investigated as an intervention protecting against cognitive decline with normal aging. Consequently, the utility of exercise regimes and/or cognitive stimulation to improve cognition in dementia in clinical populations has been a major area of study. However, these therapies are in their infancy and efficacy is unclear. Investigations utilising animal models, where dose and timing of treatment can be tightly controlled, have provided many mechanistic insights. Genetically engineered mouse models are powerful tools to investigate mechanisms underlying cognitive decline, and also how environmental manipulations can alter both cognitive outcomes and pathology. A myriad of effects following physical activity and housing in enriched environments have been reported in transgenic mice expressing Alzheimer's disease-associated mutations. In this review, we comprehensively evaluate all studies applying environmental enrichment and/or increased physical exercise to transgenic mouse models of Alzheimer's disease. It is unclear whether interventions must be applied before first onset of cognitive deficits to be effective. In order to determine the importance of timing of interventions, we specifically scrutinised studies exposing transgenic mice to exercise and environmental enrichment before and after first report of cognitive impairment. We discuss the strengths and weaknesses of these preclinical studies and suggest approaches for enhancing rigor and using mechanistic insights to inform future therapeutic interventions.
Collapse
Affiliation(s)
- Amy Shepherd
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Tracy D Zhang
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Ariel M Zeleznikow-Johnston
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| | - Anthony J Hannan
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia.,Department of Anatomy and Neuroscience, University of Melbourne, Parkville, VIC, Australia
| | - Emma L Burrows
- Florey Institute of Neuroscience and Mental Health, Melbourne Brain Centre, University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
18
|
Cattaud V, Bezzina C, Rey CC, Lejards C, Dahan L, Verret L. Early disruption of parvalbumin expression and perineuronal nets in the hippocampus of the Tg2576 mouse model of Alzheimer's disease can be rescued by enriched environment. Neurobiol Aging 2018; 72:147-158. [DOI: 10.1016/j.neurobiolaging.2018.08.024] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Revised: 08/28/2018] [Accepted: 08/28/2018] [Indexed: 11/30/2022]
|
19
|
Balthazar J, Schöwe NM, Cipolli GC, Buck HS, Viel TA. Enriched Environment Significantly Reduced Senile Plaques in a Transgenic Mice Model of Alzheimer's Disease, Improving Memory. Front Aging Neurosci 2018; 10:288. [PMID: 30319394 PMCID: PMC6168651 DOI: 10.3389/fnagi.2018.00288] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 09/03/2018] [Indexed: 01/27/2023] Open
Abstract
Alzheimer's disease (AD) is associated with a progressive dementia, and there is good evidence that it is more pronounced in individuals that have fewer stimuli during their lives. Environmental stimulation promotes morphological and functional changes in the brain, leading to amplification of cognitive functions, and has been described in humans and animals. In this study, we evaluated the effects of enriched environment (EE) stimulation on spatial memory and senile plaque formation in transgenic mice PDGFB-APPSwInd (TG) that overexpress the human amyloid precursor protein, normally resulting in an increased density of senile plaques. We compared this group of EE stimulated transgenic mice (TG-EE) with an EE stimulated control group of age-matched C57Bl/6 wild type animals (WT-EE). Both groups were exposed to EE stimulation between the ages of 8 and 12 months. As controls of the experiment, there were a group of TG mice not exposed to EE (TG-Ctrl) and a group of WT mice not exposed to EE (WT-Ctrl). The TG-EE group presented improved spatial memory when compared to the TG-Ctrl animals. In addition, the TG-EE group showed a 69.2% reduction in the total density of senile plaques in the hippocampus when compared to the TG-Ctrl group. In this group, the concentration of senile plaques was greater in the dorsal part of the hippocampus, which is linked to spatial localization, and the reduction of this density after the submission to EE was as high as 85.1%. EE stimulation had no effect on the density of amyloid-β (Aβ) oligomers. However, amyloid scavenger receptor class B member 1 (SR-B1) density was significantly decreased in the TG-Ctrl mice, but not in the TG-EE mice, suggesting that cognitive stimulation had an effect on the formation of a cognitive reserve that could prevent the accumulation of senile plaques. It is suggested that the stimulation of old mice by EE for 4 months led to the formation of brain resilience that protected the brain from the deposition of senile plaques, one of the hallmarks of AD, leading to improvement in spatial memory.
Collapse
Affiliation(s)
- Janaina Balthazar
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Natalia Mendes Schöwe
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | | | - Hudson Sousa Buck
- Department of Physiological Sciences, Faculdade de Ciências Médicas, Santa Casa de São Paulo School of Medical Sciences, São Paulo, Brazil
| | - Tania Araujo Viel
- Department of Pharmacology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
- School of Arts, Sciences and Humanities, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
20
|
Intracranial IL-17A overexpression decreases cerebral amyloid angiopathy by upregulation of ABCA1 in an animal model of Alzheimer's disease. Brain Behav Immun 2017; 65:262-273. [PMID: 28526436 PMCID: PMC5537015 DOI: 10.1016/j.bbi.2017.05.012] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 05/15/2017] [Accepted: 05/16/2017] [Indexed: 01/18/2023] Open
Abstract
Neuroinflammation is a pervasive feature of Alzheimer's disease (AD) and characterized by activated microglia, increased proinflammatory cytokines and/or infiltrating immune cells. T helper 17 (Th17) cells are found in AD brain parenchyma and interleukin-17A (IL-17A) is identified around deposits of aggregated amyloid β protein (Aβ). However, the role of IL-17A in AD pathogenesis remains elusive. We overexpressed IL-17A in an AD mouse model via recombinant adeno-associated virus serotype 5 (rAAV5)-mediated intracranial gene delivery. AD model mice subjected to injection of a vehicle (PBS) or rAAV5 carrying the lacZ gene served as controls. IL-17A did not exacerbate neuroinflammation in IL-17A-overexpressing mice. We found that IL-17A overexpression markedly improved glucose metabolism, decreased soluble Aβ levels in the hippocampus and cerebrospinal fluid, drastically reduced cerebral amyloid angiopathy, and modestly but significantly improved anxiety and learning deficits. Moreover, the ATP-binding cassette subfamily A member 1 (ABCA1), which can transport Aβ from the brain into the blood circulation, significantly increased in IL-17A-overexpressing mice. In vitro treatment of brain endothelial bEnd.3 cells with IL-17A induced a dose-dependent increase in protein expression of ABCA1 through ERK activation. Our study suggests that IL-17A may decrease Aβ levels in the brain by upregulating ABCA1 in blood-brain barrier endothelial cells.
Collapse
|
21
|
Ji MH, Tang H, Luo D, Qiu LL, Jia M, Yuan HM, Feng SW, Yang JJ. Environmental conditions differentially affect neurobehavioral outcomes in a mouse model of sepsis-associated encephalopathy. Oncotarget 2017; 8:82376-82389. [PMID: 29137271 PMCID: PMC5669897 DOI: 10.18632/oncotarget.19595] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 06/18/2017] [Indexed: 12/21/2022] Open
Abstract
Brain dysfunction remains a common complication after sepsis development and is an independent risk factor for a poorer prognosis and an increased mortality. Here we tested the hypothesis that the behavioral outcomes after lipopolysaccharides (LPS) administration are exacerbated by an impoverished environment (IE) and alleviated by an enriched environment (EE), respectively. Mice were randomly allocated in a standard environment (SE), an EE, or an IE for 4 weeks after LPS or normal saline (NS) administration. Neurobehavioral alternations were assessed by the open field, novel objective recognition, and fear conditioning tests. The expressions of proinflammatory cytokines (tumor necrosis factor (TNF-α), interleukin-1β (IL-1β), IL-6, IL-10), ionized calcium-binding adaptor molecule-1 (IBA1)-positive cells as well as glial fibrillary acidic protein (GFAP)-positive cells, brain derived neurotrophic factor (BDNF), 5-bromo-2-deoxyuridine-labeled cells in the dentate gyrus of the hippocampus, and the number of dendritic spines in the hippocampal CA1 were determined. Our results showed that the some of the neurocognitive abnormalities induced by LPS administration can be aggravated by stressful conditions such as IE but alleviated by EE. These neurocognitive alternations were accompanied by significant changes in biomarkers of immune response and hippocampal synaptic plasticity. In summary, our study confirmed the negative impact of IE and the positive effects of EE on the cognitive function after LPS administration, with potential implications to the basis of sepsis-related cognitive impairments in the critically ill patients.
Collapse
Affiliation(s)
- Mu-Huo Ji
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Hui Tang
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Dan Luo
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Li-Li Qiu
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| | - Min Jia
- Department of Anesthesiology, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Hong-Mei Yuan
- Department of Anesthesiology, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Shan-Wu Feng
- Department of Anesthesiology, Obstetrics and Gynecology Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Jian-Jun Yang
- Department of Anesthesiology, Zhongda Hospital, Medical School, Southeast University, Nanjing, China
| |
Collapse
|
22
|
Hüttenrauch M, Walter S, Kaufmann M, Weggen S, Wirths O. Limited Effects of Prolonged Environmental Enrichment on the Pathology of 5XFAD Mice. Mol Neurobiol 2016; 54:6542-6555. [DOI: 10.1007/s12035-016-0167-x] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Accepted: 09/27/2016] [Indexed: 10/20/2022]
|
23
|
Li Q, Li X, Wang L, Zhang Y, Chen L. miR-98-5p Acts as a Target for Alzheimer's Disease by Regulating Aβ Production Through Modulating SNX6 Expression. J Mol Neurosci 2016; 60:413-420. [PMID: 27541017 DOI: 10.1007/s12031-016-0815-7] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 08/01/2016] [Indexed: 01/01/2023]
Abstract
Accumulation of amyloid β-peptide (Aβ) in the brain of Alzheimer disease (AD) patients is believed to be the main pathological feature of the disease. Meanwhile, miR-98-5p dysregulation was found in AD. However, whether miR-98-5p is involved in the accumulation of Aβ in AD, the underlying molecule mechanism remains unclear. In the present study, we confirmed that miR-98-5p negatively regulated sorting nexin 6 (SNX6) expression by targeting the 3'-UTR of SNX6 mRNA. Downregulation of miR-98-5p alleviated Aβ-induced viability inhibition and decreased apoptosis in SK-N-SH and SH-SY5Y cells by upregulating SNX6 expression. Furthermore, downregulation of miR-98-5p decreased SNX6-dependent levels of Aβ40, Aβ42, β-site APP-cleaving enzyme 1 (BACE1), soluble amyloid precursor protein β (sAPPβ), and membrane-associated APP β-carboxyl terminal fragment (βCTF) in SK-N-SH and HEK293 cells. Our findings demonstrate that miR-98-5p modulates SNX6 expression and thus plays a critical role in accumulation of Aβ. Therefore, miR-98-5p may be a novel therapeutic target for AD.
Collapse
Affiliation(s)
- Qiushi Li
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People's Republic of China
| | - Xidong Li
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People's Republic of China
| | - Li Wang
- Department of Neurology, Shenyang General Hospital of PLA, Shenyang, Liaoning, 110084, People's Republic of China
| | - Yanhui Zhang
- Department of Neurology, The First Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning, 121000, People's Republic of China
| | - Long Chen
- Department of Anesthesiology, The First Affiliated Hospital of Jinzhou Medical University, 5-2 People Street, Jinzhou, Liaoning, 121000, People's Republic of China.
| |
Collapse
|
24
|
Montgomery KS, Edwards G, Levites Y, Kumar A, Myers CE, Gluck MA, Setlow B, Bizon JL. Deficits in hippocampal-dependent transfer generalization learning accompany synaptic dysfunction in a mouse model of amyloidosis. Hippocampus 2016; 26:455-71. [PMID: 26418152 PMCID: PMC4803574 DOI: 10.1002/hipo.22535] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/11/2015] [Accepted: 09/17/2015] [Indexed: 11/08/2022]
Abstract
Elevated β-amyloid and impaired synaptic function in hippocampus are among the earliest manifestations of Alzheimer's disease (AD). Most cognitive assessments employed in both humans and animal models, however, are insensitive to this early disease pathology. One critical aspect of hippocampal function is its role in episodic memory, which involves the binding of temporally coincident sensory information (e.g., sights, smells, and sounds) to create a representation of a specific learning epoch. Flexible associations can be formed among these distinct sensory stimuli that enable the "transfer" of new learning across a wide variety of contexts. The current studies employed a mouse analog of an associative "transfer learning" task that has previously been used to identify risk for prodromal AD in humans. The rodent version of the task assesses the transfer of learning about stimulus features relevant to a food reward across a series of compound discrimination problems. The relevant feature that predicts the food reward is unchanged across problems, but an irrelevant feature (i.e., the context) is altered. Experiment 1 demonstrated that C57BL6/J mice with bilateral ibotenic acid lesions of hippocampus were able to discriminate between two stimuli on par with control mice; however, lesioned mice were unable to transfer or apply this learning to new problem configurations. Experiment 2 used the APPswe PS1 mouse model of amyloidosis to show that robust impairments in transfer learning are evident in mice with subtle β-amyloid-induced synaptic deficits in the hippocampus. Finally, Experiment 3 confirmed that the same transfer learning impairments observed in APPswePS1 mice were also evident in the Tg-SwDI mouse, a second model of amyloidosis. Together, these data show that the ability to generalize learned associations to new contexts is disrupted even in the presence of subtle hippocampal dysfunction and suggest that, across species, this aspect of hippocampal-dependent learning may be useful for early identification of AD-like pathology.
Collapse
Affiliation(s)
- Karienn S. Montgomery
- Department of Neuroscience and Experimental Therapeutics, Texas A&M Health Science Center, Bryan, TX
| | - George Edwards
- Mitchell Center for Alzheimer’s Disease and Related Brain Disorders, Department of Neurology, University of Texas Health Science Center in Houston, Houston, TX
| | - Yona Levites
- Department of Neuroscience, University of Florida, Gainesville, FL
| | - Ashok Kumar
- Department of Neuroscience, University of Florida, Gainesville, FL
| | - Catherine E. Myers
- VA New Jersey Health Care System, East Orange, NJ 07018
- Department of Pharmacology, Physiology & Neuroscience, New Jersey Medical School, Rutgers University, Newark, NJ
| | - Mark A. Gluck
- Center for Molecular & Behavioral Neuroscience, Rutgers University, Newark, NJ
| | - Barry Setlow
- Department of Psychiatry, University of Florida, Gainesville, FL
| | | |
Collapse
|
25
|
Végh MJ, Heldring CM, Kamphuis W, Hijazi S, Timmerman AJ, Li KW, van Nierop P, Mansvelder HD, Hol EM, Smit AB, van Kesteren RE. Reducing hippocampal extracellular matrix reverses early memory deficits in a mouse model of Alzheimer's disease. Acta Neuropathol Commun 2014; 2:76. [PMID: 24974208 PMCID: PMC4149201 DOI: 10.1186/s40478-014-0076-z] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Accepted: 06/17/2014] [Indexed: 11/13/2022] Open
Abstract
Alzheimer’s disease is caused by increased production or reduced clearance of amyloid-β, which results in the formation amyloid-β plaques and triggers a cascade of downstream events leading to progressive neurodegeneration. The earliest clinical symptoms of Alzheimer’s disease, i.e., memory loss, are however poorly understood from a molecular and cellular perspective. Here we used APPswe/PS1dE9 (APP/PS1) transgenic mice to study the early pre-pathological effects of increased amyloid-β levels on hippocampal synaptic plasticity and memory. Using an unbiased proteomics approach we show that the early increase in amyloid-β levels in APP/PS1 mice at three months of age coincides with a robust and significant upregulation of several protein components of the extracellular matrix in hippocampal synaptosome preparations. This increase in extracellular matrix levels occurred well before the onset of plaque formation and was paralleled by impairments in hippocampal long-term potentiation and contextual memory. Direct injection into the hippocampus of the extracellular matrix inactivating enzyme chondroitinase ABC restored both long-term potentiation and contextual memory performance. These findings indicate an important role for the extracellular matrix in causing early memory loss in Alzheimer’s disease.
Collapse
|
26
|
Cognitive Reserve and Alzheimer’s Disease. Mol Neurobiol 2014; 51:187-208. [DOI: 10.1007/s12035-014-8720-y] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2014] [Accepted: 04/17/2014] [Indexed: 12/13/2022]
|
27
|
Castillo-Carranza DL, Guerrero-Muñoz MJ, Kayed R. Immunotherapy for the treatment of Alzheimer's disease: amyloid-β or tau, which is the right target? Immunotargets Ther 2013; 3:19-28. [PMID: 27471697 PMCID: PMC4918231 DOI: 10.2147/itt.s40131] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the presence of amyloid plaques composed mainly of amyloid-β (Aβ) protein. Overproduction or slow clearance of Aβ initiates a cascade of pathologic events that may lead to formation of neurofibrillary tangles, neuronal cell death, and dementia. Although immunotherapy in animal models has been demonstrated to be successful at removing plaques or prefibrillar forms of Aβ, clinical trials have yielded disappointing results. The lack of substantial cognitive improvement obtained by targeting Aβ raises the question of whether or not this is the correct target. Another important pathologic process in the AD brain is tau aggregation, which seems to become independent once initiated. Recent studies targeting tau in AD mouse models have displayed evidence of cognitive improvement, providing a novel therapeutic approach for the treatment of AD. In this review, we describe new advances in immunotherapy targeting Aβ peptide and tau protein, as well as future directions.
Collapse
Affiliation(s)
- Diana L Castillo-Carranza
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Marcos J Guerrero-Muñoz
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rakez Kayed
- Mitchell Center for Neurodegenerative Diseases, University of Texas Medical Branch, Galveston, TX, USA; Departments of Neurology, Neuroscience, and Cell Biology, University of Texas Medical Branch, Galveston, TX, USA; Sealy Center for Vaccine Development, University of Texas Medical Branch, Galveston, TX, USA
| |
Collapse
|