1
|
Sethi P, Bhaskar R, Singh KK, Gupta S, Han SS, Avinash D, Abomughaid MM, Koul A, Rani B, Ghosh S, Jha NK, Sinha JK. Exploring advancements in early detection of Alzheimer's disease with molecular assays and animal models. Ageing Res Rev 2024; 100:102411. [PMID: 38986845 DOI: 10.1016/j.arr.2024.102411] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/14/2024] [Revised: 07/04/2024] [Accepted: 07/04/2024] [Indexed: 07/12/2024]
Abstract
Alzheimer's Disease (AD) is a challenging neurodegenerative condition, with overwhelming implications for affected individuals and healthcare systems worldwide. Animal models have played a crucial role in studying AD pathogenesis and testing therapeutic interventions. Remarkably, studies on the genetic factors affecting AD risk, such as APOE and TREM2, have provided valuable insights into disease mechanisms. Early diagnosis has emerged as a crucial factor in effective AD management, as demonstrated by clinical studies emphasizing the benefits of initiating treatment at early stages. Novel diagnostic technologies, including RNA sequencing of microglia, offer promising avenues for early detection and monitoring of AD progression. Therapeutic strategies remain to evolve, with a focus on targeting amyloid beta (Aβ) and tau pathology. Advances in animal models, such as APP-KI mice, and the advancement of anti-Aβ drugs signify progress towards more effective treatments. Therapeutically, the focus has shifted towards intricate approaches targeting multiple pathological pathways simultaneously. Strategies aimed at reducing Aβ plaque accumulation, inhibiting tau hyperphosphorylation, and modulating neuroinflammation are actively being explored, both in preclinical models and clinical trials. While challenges continue in developing validated animal models and translating preclinical findings to clinical success, the continuing efforts in understanding AD at molecular, cellular, and clinical levels offer hope for improved management and eventual prevention of this devastating disease.
Collapse
Affiliation(s)
- Paalki Sethi
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India
| | - Rakesh Bhaskar
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - Krishna Kumar Singh
- Symbiosis Centre for Information Technology (SCIT), Rajiv Gandhi InfoTech Park, Hinjawadi, Pune, Maharashtra 411057, India
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Sung Soo Han
- School of Chemical Engineering, Yeungnam University, Gyeonsang 38541, Republic of Korea; Research Institute of Cell Culture, Yeungnam University, Gyeonsang 38541, Republic of Korea
| | - D Avinash
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Mosleh Mohammad Abomughaid
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Bisha, Bisha 61922, Saudi Arabia
| | - Apurva Koul
- Chandigarh Pharmacy College, Chandigarh Group of Colleges, Jhanjeri, Mohali, Punjab 140307, India
| | - Bindu Rani
- Department of Medicine, National Institute of Medical Sciences, NIMS University, Jaipur, Rajsthan, India
| | - Shampa Ghosh
- GloNeuro, Sector 107, Vishwakarma Road, Noida, Uttar Pradesh 201301, India.
| | - Niraj Kumar Jha
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab 140401, India; School of Bioengineering & Biosciences, Lovely Professional University, Phagwara 144411, India; Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun 248007, India; Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India.
| | | |
Collapse
|
2
|
Eryilmaz NC, Arslan M, Kucuk A, Tuna AT, Guney S, Kaplanoglu GT, Kavutcu M. Evaluation of the Effects of Repetitive Anaesthesia Administration on the Brain Tissues and Cognitive Functions of Rats with Experimental Alzheimer's Disease. MEDICINA (KAUNAS, LITHUANIA) 2024; 60:1266. [PMID: 39202547 PMCID: PMC11356554 DOI: 10.3390/medicina60081266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Academic Contribution Register] [Received: 06/05/2024] [Revised: 07/17/2024] [Accepted: 08/02/2024] [Indexed: 09/03/2024]
Abstract
Introduction: We evaluated the effects of repeated ketamine, propofol, and ketamine + propofol administration on cognitive functions and brain tissue of elderly rat models with streptozotocin-induced Alzheimer's disease. Materials and Methods: Thirty elderly male Wistar Albino rats were divided into five groups: control (Group C), Alzheimer's (Group A), Alzheimer's + ketamine (Group AK), Alzheimer's + propofol (Group AP), and Alzheimer's + propofol + ketamine (Group APK). Alzheimer's disease was induced in Groups A, AK, AP, and APK via intracerebroventricular streptozotocin. Four weeks after surgery, ketamine, propofol, and ketamine + propofol were administered intraperitoneally for 3 days to Groups AK, AP, and APK, respectively. The radial arm maze test (RAMT) was performed in the initial, 1st, 2nd, 3rd, and 4th weeks after surgery and daily following anaesthesia. Blood and brain tissue samples were obtained. Results: The RAMT results of Groups A, AK, AP, and APK decreased compared to Group C 2 weeks after Alzheimer's disease onset. Compared to Group A, the RAMT results increased in Groups AK and APK after the first anaesthesia, and in Group AP after the second anaesthesia. Brain tissue paraoxonase-1 (PON-1) and catalase (CAT) activities were low, and the thiobarbituric acid reactive substance (TBARS) level was high in Group A compared to Group C. TBARS levels of Groups AP and APK were lower than Group A, while CAT activity was higher. PON-1 activity was higher in Groups AK, AP, and APK than in Group A. Histopathological changes decreased in Groups AP and AK. A decrease in p53 was found in Group C compared to Group A. Ketamine and propofol were found to be effective at Bcl-2 immunoexpression, but a decrease in Caspase-3 was observed in Group APK. GFAP immunoexpression increased in Group A compared to Group C and in Group AP compared to Group AK. Conclusions: Repetitive anaesthesia application was found to positively affect cognitive functions. This was supported by histopathological and biochemical markers.
Collapse
Affiliation(s)
- Nuray Camgoz Eryilmaz
- Department of Anesthesiology and Reanimation, Gazi University Faculty of Medicine, 06500 Ankara, Türkiye; (N.C.E.); (M.A.)
| | - Mustafa Arslan
- Department of Anesthesiology and Reanimation, Gazi University Faculty of Medicine, 06500 Ankara, Türkiye; (N.C.E.); (M.A.)
| | - Aysegul Kucuk
- Department of Physiology, Kutahya Health Sciences University Faculty of Medicine, 43020 Kutahya, Türkiye
| | - Ayca Tas Tuna
- Department of Anesthesiology and Reanimation, Sakarya University Faculty of Medicine, 54050 Sakarya, Türkiye;
| | - Sevin Guney
- Department of Physiology, Gazi University Faculty of Medicine, 06500 Ankara, Türkiye;
| | - Gulnur Take Kaplanoglu
- Department of Histology and Embryology, Gazi University Faculty of Medicine, 06500 Ankara, Türkiye;
| | - Mustafa Kavutcu
- Department of Medical Biochemistry, Gazi University Faculty of Medicine, 06500 Ankara, Türkiye;
| |
Collapse
|
3
|
Karimi Tari P, Parsons CG, Collingridge GL, Rammes G. Memantine: Updating a rare success story in pro-cognitive therapeutics. Neuropharmacology 2024; 244:109737. [PMID: 37832633 DOI: 10.1016/j.neuropharm.2023.109737] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/16/2023] [Revised: 09/25/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023]
Abstract
The great potential for NMDA receptor modulators as druggable targets in neurodegenerative disorders has been met with limited success. Considered one of the rare exceptions, memantine has consistently demonstrated restorative and prophylactic properties in many AD models. In clinical trials memantine slows the decline in cognitive performance associated with AD. Here, we provide an overview of the basic properties including pharmacological targets, toxicology and cellular effects of memantine. Evidence demonstrating reductions in molecular, physiological and behavioural indices of AD-like impairments associated with memantine treatment are also discussed. This represents both an extension and homage to Dr. Chris Parson's considerable contributions to our fundamental understanding of a success story in the AD treatment landscape.
Collapse
Affiliation(s)
- Parisa Karimi Tari
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada
| | - Chris G Parsons
- Galimedix Therapeutics, Inc., 2704 Calvend Lane, Kensington, 20895, MD, USA
| | - Graham L Collingridge
- Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, ON, M5G 1X5, Canada; Department of Physiology, University of Toronto, Toronto, ON, M5S 1A8, Canada; TANZ Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care Medicine of the Technical University of Munich, School of Medicine, 22, 81675, Munich, Germany.
| |
Collapse
|
4
|
Murakami T, Abe M, Tiksnadi A, Nemoto A, Futamura M, Yamakuni R, Kubo H, Kobayashi N, Ito H, Hanajima R, Hashimoto Y, Ugawa Y. Abnormal motor cortical plasticity as a useful neurophysiological biomarker for Alzheimer's disease pathology. Clin Neurophysiol 2024; 158:170-179. [PMID: 38219406 DOI: 10.1016/j.clinph.2023.12.131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/09/2022] [Revised: 11/27/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024]
Abstract
OBJECTIVE Amyloid-beta (Aβ) and tau accumulations impair long-term potentiation (LTP) induction in animal hippocampi. We investigated relationships between motor-cortical plasticity and biomarkers for Alzheimer's disease (AD) diagnosis in subjects with cognitive decline. METHODS Twenty-six consecutive subjects who complained of memory problems participated in this study. We applied transcranial quadripuse stimulation with an interstimulus interval of 5 ms (QPS5) to induce LTP-like plasticity. Motor-evoked potentials were recorded from the right first-dorsal interosseous muscle before and after QPS5. Cognitive functions, Aβ42 and tau levels in the cerebrospinal fluid (CSF) were measured. Amyloid positron-emission tomography (PET) with11C-Pittsburg compound-B was also conducted. We studied correlations of QPS5-induced plasticity with cognitive functions or AD-related biomarkers. RESULTS QPS5-induced LTP-like plasticity positively correlated with cognitive scores. The degree of LTP-like plasticity negatively correlated with levels of CSF-tau, and the amount of amyloid-PET accumulation at the precuneus, and correlated with the CSF-Aβ42 level positively. In the amyloid-PET positive subjects, non-responder rate of QPS5 was higher than the CSF-tau positive rate. CONCLUSIONS Findings suggest that QPS5-induced LTP-like plasticity is a functional biomarker of AD. QPS5 could detect abnormality at earlier stages than CSF-tau in the amyloid-PET positive subjects. SIGNIFICANCE Assessing motor-cortical plasticity could be a useful neurophysiological biomarker for AD pathology.
Collapse
Affiliation(s)
- Takenobu Murakami
- Department of Neurology, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan; Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishimachi 36-1, Yonago 683-8504, Japan.
| | - Mitsunari Abe
- Center for Neurological Disorders, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Amanda Tiksnadi
- Department of Neurology, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan; Department of Neurology, Cipto Mangunkusumo Hospital, Faculty of Medicine, Universitas Indonesia, Salemba Raya No. 6, Jakarta 10430, Indonesia
| | - Ayaka Nemoto
- Advanced Clinical Research Center, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Miyako Futamura
- Rehabilitation Center, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Ryo Yamakuni
- Department of Radiology, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Hitoshi Kubo
- Advanced Clinical Research Center, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan; Department of Radiological Sciences, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Naoto Kobayashi
- Azuma Street Clinic, Sakaemachi 1-28, Fukushima 960-8031, Japan
| | - Hiroshi Ito
- Advanced Clinical Research Center, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan; Department of Radiology, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Nishimachi 36-1, Yonago 683-8504, Japan
| | - Yasuhiro Hashimoto
- Department of Biochemistry, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| | - Yoshikazu Ugawa
- Department of Neurology, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan; Department of Human Neurophysiology, Faculty of Medicine, Fukushima Medical University, Hikarigaoka 1, Fukushima 960-1295, Japan
| |
Collapse
|
5
|
Colavitta MF, Barrantes FJ. Therapeutic Strategies Aimed at Improving Neuroplasticity in Alzheimer Disease. Pharmaceutics 2023; 15:2052. [PMID: 37631266 PMCID: PMC10459958 DOI: 10.3390/pharmaceutics15082052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/25/2023] [Revised: 07/23/2023] [Accepted: 07/28/2023] [Indexed: 08/27/2023] Open
Abstract
Alzheimer disease (AD) is the most prevalent form of dementia among elderly people. Owing to its varied and multicausal etiopathology, intervention strategies have been highly diverse. Despite ongoing advances in the field, efficient therapies to mitigate AD symptoms or delay their progression are still of limited scope. Neuroplasticity, in broad terms the ability of the brain to modify its structure in response to external stimulation or damage, has received growing attention as a possible therapeutic target, since the disruption of plastic mechanisms in the brain appear to correlate with various forms of cognitive impairment present in AD patients. Several pre-clinical and clinical studies have attempted to enhance neuroplasticity via different mechanisms, for example, regulating glucose or lipid metabolism, targeting the activity of neurotransmitter systems, or addressing neuroinflammation. In this review, we first describe several structural and functional aspects of neuroplasticity. We then focus on the current status of pharmacological approaches to AD stemming from clinical trials targeting neuroplastic mechanisms in AD patients. This is followed by an analysis of analogous pharmacological interventions in animal models, according to their mechanisms of action.
Collapse
Affiliation(s)
- María F. Colavitta
- Laboratory of Molecular Neurobiology, Biomedical Research Institute (BIOMED), Universidad Católica Argentina (UCA)—National Scientific and Technical Research Council (CONICET), Buenos Aires C1107AAZ, Argentina
- Centro de Investigaciones en Psicología y Psicopedagogía (CIPP-UCA), Facultad de Psicología, Av. Alicia Moreau de Justo, Buenos Aires C1107AAZ, Argentina;
| | - Francisco J. Barrantes
- Laboratory of Molecular Neurobiology, Biomedical Research Institute (BIOMED), Universidad Católica Argentina (UCA)—National Scientific and Technical Research Council (CONICET), Buenos Aires C1107AAZ, Argentina
| |
Collapse
|
6
|
Pershina EV, Chernomorets IY, Fedorov DA, Arkhipov VI. Pharmacological Modulation of Excitotoxicity through the Combined Use of NMDA Receptor Inhibition and Group III mGlu Activation Reduces TMT-Induced Neurodegeneration in the Rat Hippocampus. Int J Mol Sci 2023; 24:ijms24098249. [PMID: 37175959 PMCID: PMC10179112 DOI: 10.3390/ijms24098249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/28/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/15/2023] Open
Abstract
We studied the neuroprotective properties of the non-competitive NMDA receptor antagonist memantine, in combination with a positive allosteric modulator of metabotropic glutamate receptors of Group III, VU 0422288. The treatment was started 48 h after the injection of neurotoxic agent trimethyltin (TMT) at 7.5 mg/kg. Three weeks after TMT injection, functional and morphological changes in a rat hippocampus were evaluated, including the expression level of genes characterizing glutamate transmission and neuroinflammation, animal behavior, and hippocampal cell morphology. Significant neuronal cell death occurred in the CA3 and CA4 regions, and to a lesser extent, in the CA1 and CA2 regions. The death of neurons in the CA1 field was significantly reduced in animals with a combined use of memantine and VU 0422288. In the hippocampus of these animals, the level of expression of genes characterizing glutamatergic synaptic transmission (Grin2b, Gria1, EAAT2) did not differ from the level in control animals, as well as the expression of genes characterizing neuroinflammation (IL1b, TGF beta 1, Aif1, and GFAP). However, the expression of genes characterizing neuroinflammation was markedly increased in the hippocampus of animals treated with memantine or VU 0422288 alone after TMT. The results of immunohistochemical studies confirmed a significant activation of microglia in the hippocampus three weeks after TMT injection. In contrast to the hilus, microglia in the CA1 region had an increase in rod-like cells. Moreover, in the CA1 field of the hippocampus of the animals of the MEM + VU group, the amount of such microglia was close to the control. Thus, the short-term modulation of glutamatergic synaptic transmission by memantine and subsequent activation of Group III mGluR significantly affected the dynamics of neurodegeneration in the hippocampus.
Collapse
Affiliation(s)
- Ekaterina V Pershina
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Irina Yu Chernomorets
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Dmitry A Fedorov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| | - Vladimir I Arkhipov
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, 142290 Pushchino, Moscow Region, Russia
| |
Collapse
|
7
|
Bagheri S, Rashno M, Salehi I, Karimi SA, Raoufi S, Komaki A. Geraniol improves passive avoidance memory and hippocampal synaptic plasticity deficits in a rat model of Alzheimer's disease. Eur J Pharmacol 2023; 951:175714. [PMID: 37054939 DOI: 10.1016/j.ejphar.2023.175714] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2023] [Revised: 04/03/2023] [Accepted: 04/11/2023] [Indexed: 04/15/2023]
Abstract
Alzheimer's disease (AD) is the most progressive and irreversible neurodegenerative disease that leads to synaptic loss and cognitive decline. The present study was designed to evaluate the effects of geraniol (GR), a valuable acyclic monoterpene alcohol, with protective and therapeutic effects, on passive avoidance memory, hippocampal synaptic plasticity, and amyloid-beta (Aβ) plaques formation in an AD rat model induced by intracerebroventricular (ICV) microinjection of Aβ1-40. Seventy male Wistar rats were randomly into sham, control, control-GR (100 mg/kg; P.O. (orally), AD, GR-AD (100 mg/kg; P.O.; pretreatment), AD-GR (100 mg/kg; P.O.; treatment), and GR-AD-GR (100 mg/kg; P.O.; pretreatment & treatment). Administration of GR was continued for four consecutive weeks. Training for the passive avoidance test was carried out on the 36th day and a memory retention test was performed 24 h later. On day 38, hippocampal synaptic plasticity (long-term potentiation; LTP) was recorded in perforant path-dentate gyrus (PP-DG) synapses to assess field excitatory postsynaptic potentials (fEPSPs) slope and population spike (PS) amplitude. Subsequently, Aβ plaques were identified in the hippocampus by Congo red staining. The results showed that Aβ microinjection increased passive avoidance memory impairment, suppressed of hippocampal LTP induction, and enhanced of Aβ plaque formation in the hippocampus. Interestingly, oral administration of GR improved passive avoidance memory deficit, ameliorated hippocampal LTP impairment, and reduced Aβ plaque accumulation in the Aβ-infused rats. The results suggest that GR mitigates Aβ-induced passive avoidance memory impairment, possibly through alleviation of hippocampal synaptic dysfunction and inhibition of Aβ plaque formation.
Collapse
Affiliation(s)
- Shokufeh Bagheri
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Masome Rashno
- Asadabad School of Medical Sciences, Asadabad, Iran; Student Research Committee, Asadabad School of Medical Sciences, Asadabad, Iran
| | - Iraj Salehi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Seyed Asaad Karimi
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Safoura Raoufi
- Department of Physiology, School of Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| | - Alireza Komaki
- Department of Neuroscience, School of Science and Advanced Technologies in Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Neurophysiology Research Center, Hamadan University of Medical Sciences, Hamadan, Iran. http://umsha.ac.ir
| |
Collapse
|
8
|
Varshavskaya KB, Mitkevich VA, Makarov AA, Barykin EP. Synthetic, Cell-Derived, Brain-Derived, and Recombinant β-Amyloid: Modelling Alzheimer's Disease for Research and Drug Development. Int J Mol Sci 2022; 23:15036. [PMID: 36499362 PMCID: PMC9738609 DOI: 10.3390/ijms232315036] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/01/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/02/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in the elderly, characterised by the accumulation of senile plaques and tau tangles, neurodegeneration, and neuroinflammation in the brain. The development of AD is a pathological cascade starting according to the amyloid hypothesis with the accumulation and aggregation of the β-amyloid peptide (Aβ), which induces hyperphosphorylation of tau and promotes the pro-inflammatory activation of microglia leading to synaptic loss and, ultimately, neuronal death. Modelling AD-related processes is important for both studying the molecular basis of the disease and the development of novel therapeutics. The replication of these processes is often achieved with the use of a purified Aβ peptide. However, Aβ preparations obtained from different sources can have strikingly different properties. This review aims to compare the structure and biological effects of Aβ oligomers and aggregates of a higher order: synthetic, recombinant, purified from cell culture, or extracted from brain tissue. The authors summarise the applicability of Aβ preparations for modelling Aβ aggregation, neurotoxicity, cytoskeleton damage, receptor toxicity in vitro and cerebral amyloidosis, synaptic plasticity disruption, and cognitive impairment in vivo and ex vivo. Further, the paper discusses the causes of the reported differences in the effect of Aβ obtained from the sources mentioned above. This review points to the importance of the source of Aβ for AD modelling and could help researchers to choose the optimal way to model the Aβ-induced abnormalities.
Collapse
Affiliation(s)
| | | | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Vavilov St. 32, 119991 Moscow, Russia
| | | |
Collapse
|
9
|
Bioactive human Alzheimer brain soluble Aβ: pathophysiology and therapeutic opportunities. Mol Psychiatry 2022; 27:3182-3191. [PMID: 35484241 DOI: 10.1038/s41380-022-01589-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/03/2022] [Revised: 04/11/2022] [Accepted: 04/14/2022] [Indexed: 12/16/2022]
Abstract
The accumulation of amyloid-β protein (Aβ) plays an early role in the pathogenesis of Alzheimer's disease (AD). The precise mechanism of how Aβ accumulation leads to synaptic dysfunction and cognitive impairment remains unclear but is likely due to small soluble oligomers of Aβ (oAβ). Most studies have used chemical synthetic or cell-secreted Aβ oligomers to study their pathogenic mechanisms, but the Aβ derived from human AD brain tissue is less well characterized. Here we review updated knowledge on the extraction and characterization of bioactive human AD brain oAβ and the mechanisms by which they cause hippocampal synaptic dysfunction. Human AD brain-derived oAβ can impair hippocampal long-term potentiation (LTP) and enhance long-term depression (LTD). Many studies suggest that oAβ may directly disrupt neuronal NMDA receptors, AMPA receptors and metabotropic glutamate receptors (mGluRs). oAβ also impairs astrocytic synaptic functions, including glutamate uptake, D-serine release, and NMDA receptor function. We also discuss oAβ-induced neuronal hyperexcitation. These results may suggest a multi-target approach for the treatment of AD, including both oAβ neutralization and reversal of glutamate-mediated excitotoxicity.
Collapse
|
10
|
Taniguchi K, Yamamoto F, Amamo A, Tamaoka A, Sanjo N, Yokota T, Kametani F, Araki W. Amyloid-β oligomers interact with NMDA receptors containing GluN2B subunits and metabotropic glutamate receptor 1 in primary cortical neurons: relevance to the synapse pathology of Alzheimer’s disease. Neurosci Res 2022; 180:90-98. [DOI: 10.1016/j.neures.2022.03.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/14/2021] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 01/22/2023]
|
11
|
Sagar NA, Tarafdar S, Agarwal S, Tarafdar A, Sharma S. Polyamines: Functions, Metabolism, and Role in Human Disease Management. Med Sci (Basel) 2021; 9:44. [PMID: 34207607 PMCID: PMC8293435 DOI: 10.3390/medsci9020044] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/03/2021] [Revised: 06/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
Putrescine, spermine, and spermidine are the important polyamines (PAs), found in all living organisms. PAs are formed by the decarboxylation of amino acids, and they facilitate cell growth and development via different cellular responses. PAs are the integrated part of the cellular and genetic metabolism and help in transcription, translation, signaling, and post-translational modifications. At the cellular level, PA concentration may influence the condition of various diseases in the body. For instance, a high PA level is detrimental to patients suffering from aging, cognitive impairment, and cancer. The levels of PAs decline with age in humans, which is associated with different health disorders. On the other hand, PAs reduce the risk of many cardiovascular diseases and increase longevity, when taken in an optimum quantity. Therefore, a controlled diet is an easy way to maintain the level of PAs in the body. Based on the nutritional intake of PAs, healthy cell functioning can be maintained. Moreover, several diseases can also be controlled to a higher extend via maintaining the metabolism of PAs. The present review discusses the types, important functions, and metabolism of PAs in humans. It also highlights the nutritional role of PAs in the prevention of various diseases.
Collapse
Affiliation(s)
- Narashans Alok Sagar
- Department of Agriculture and Environmental Sciences, National Institute of Food Technology Entrepreneurship and Management, Kundli, Sonepat 131028, Haryana, India
- Food Microbiology Lab, Division of Livestock Products Technology, ICAR-Indian Veterinary Research Institute, Izatnagar 243122, Uttar Pradesh, India
| | - Swarnava Tarafdar
- Department of Radiodiagnosis and Imaging, All India Institute of Medical Science, Rishikesh 249203, Uttarakhand, India;
| | - Surbhi Agarwal
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh 160012, India;
| | - Ayon Tarafdar
- Livestock Production and Management Section, ICAR-Indian Veterinary Research Institute, Izatnagar 243122, Uttar Pradesh, India;
| | - Sunil Sharma
- Department of Agriculture and Environmental Sciences, National Institute of Food Technology Entrepreneurship and Management, Kundli, Sonepat 131028, Haryana, India
| |
Collapse
|
12
|
Toniolo S, Sen A, Husain M. Modulation of Brain Hyperexcitability: Potential New Therapeutic Approaches in Alzheimer's Disease. Int J Mol Sci 2020; 21:E9318. [PMID: 33297460 PMCID: PMC7730926 DOI: 10.3390/ijms21239318] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/29/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/12/2022] Open
Abstract
People with Alzheimer's disease (AD) have significantly higher rates of subclinical and overt epileptiform activity. In animal models, oligomeric Aβ amyloid is able to induce neuronal hyperexcitability even in the early phases of the disease. Such aberrant activity subsequently leads to downstream accumulation of toxic proteins, and ultimately to further neurodegeneration and neuronal silencing mediated by concomitant tau accumulation. Several neurotransmitters participate in the initial hyperexcitable state, with increased synaptic glutamatergic tone and decreased GABAergic inhibition. These changes appear to activate excitotoxic pathways and, ultimately, cause reduced long-term potentiation, increased long-term depression, and increased GABAergic inhibitory remodelling at the network level. Brain hyperexcitability has therefore been identified as a potential target for therapeutic interventions aimed at enhancing cognition, and, possibly, disease modification in the longer term. Clinical trials are ongoing to evaluate the potential efficacy in targeting hyperexcitability in AD, with levetiracetam showing some encouraging effects. Newer compounds and techniques, such as gene editing via viral vectors or brain stimulation, also show promise. Diagnostic challenges include identifying best biomarkers for measuring sub-clinical epileptiform discharges. Determining the timing of any intervention is critical and future trials will need to carefully stratify participants with respect to the phase of disease pathology.
Collapse
Affiliation(s)
- Sofia Toniolo
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| | - Arjune Sen
- Oxford Epilepsy Research Group, Nuffield Department Clinical Neurosciences, John Radcliffe Hospital, Oxford OX3 9DU, UK;
| | - Masud Husain
- Cognitive Neurology Group, Nuffield Department of Clinical Neurosciences, John Radcliffe Hospital, University of Oxford, Oxford OX3 9DU, UK;
- Wellcome Trust Centre for Integrative Neuroimaging, Department of Experimental Psychology, University of Oxford, Oxford OX2 6AE, UK
| |
Collapse
|
13
|
Ahnaou A, White E, Biermans R, Manyakov NV, Drinkenburg WHIM. In Vivo Plasticity at Hippocampal Schaffer Collateral-CA1 Synapses: Replicability of the LTP Response and Pharmacology in the Long-Evans Rat. Neural Plast 2020; 2020:6249375. [PMID: 33273904 PMCID: PMC7676971 DOI: 10.1155/2020/6249375] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/13/2020] [Revised: 09/17/2020] [Accepted: 10/06/2020] [Indexed: 01/15/2023] Open
Abstract
Broad issues associated with non-replicability have been described in experimental pharmacological and behavioral cognitive studies. Efforts to prevent biases that contribute to non-replicable scientific protocols and to improve experimental rigor for reproducibility are increasingly seen as a basic requirement for the integrity of scientific research. Synaptic plasticity, encompassing long-term potentiation (LTP), is believed to underlie mechanisms of learning and memory. The present study was undertaken in Long-Evans (LE) rats, a strain of rat commonly used in cognitive behavioral tests, to (1) compare three LTP tetanisation protocols, namely, the high-frequency stimulation (HFS), the theta-burst stimulation (TBS), and the paired-pulse facilitation (PPF) at the Schaffer collateral-CA1 stratum radiatum synapse and to (2) assess sensitivity to acute pharmacology. Results: (1) When compared to Sprague-Dawley (SD) rats, the HFS using a stimulus intensity of 50% of the maximum slope obtained from input/output (I/O) curves elicited lower and higher thresholds of synaptic plasticity responses in SD and LE rats, respectively. The 2-train TBS protocol significantly enhanced the LTP response in LE rats over the 5- and 10-train TBS protocols in both strains, and the 5 × TBS protocol inducing a subthreshold LTP response was used in subsequent pharmacological studies in LE rats. The PPF protocol, investigating the locus of the LTP response, showed no difference for the selected interstimulus intervals. (2) Scopolamine, a nonspecific muscarinic antagonist, had a subtle effect, whereas donepezil, an acetylcholinesterase inhibitor, significantly enhanced the LTP response, demonstrating the sensitivity of the TBS protocol to enhanced cholinergic tone. MK-801, a noncompetitive N-methyl-D-aspartate (NMDA) antagonist, significantly reduced LTP response, while memantine, another NMDA antagonist, had no effect on LTP response, likely associated with a weaker TBS protocol. PQ10, a phosphodiesterase-10 inhibitor, significantly enhanced the TBS-induced LTP response, but did not change the PPF response. Overall, the results confirm the strain-dependent differences in the form of synaptic plasticity, replicate earlier plasticity results, and report effective protocols that generate a relatively subthreshold margin of LTP induction and maintenance, which are suitable for pharmacology in the LE rat strain mainly used in cognitive studies.
Collapse
Affiliation(s)
- A. Ahnaou
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - E. White
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - R. Biermans
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - N. V. Manyakov
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV., Turnhoutseweg 30, B-2340 Beerse, Belgium
| | - W. H. I. M. Drinkenburg
- Department of Neuroscience, Janssen Research & Development, Janssen Pharmaceutica NV., Turnhoutseweg 30, B-2340 Beerse, Belgium
| |
Collapse
|
14
|
Bürge M, Kratzer S, Mattusch C, Hofmann C, Kreuzer M, Parsons CG, Rammes G. The anaesthetic xenon partially restores an amyloid beta-induced impairment in murine hippocampal synaptic plasticity. Neuropharmacology 2019; 151:21-32. [PMID: 30940537 DOI: 10.1016/j.neuropharm.2019.03.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/14/2018] [Revised: 03/15/2019] [Accepted: 03/26/2019] [Indexed: 02/08/2023]
Abstract
BACKGROUND It is controversially discussed whether general anaesthesia increases the risk of Alzheimer's disease (AD) or accelerates its progression. One important factor in AD pathogenesis is the accumulation of soluble amyloid beta (Aβ) oligomers which affect N-methyl-d-aspartate (NMDA) receptor function and abolish hippocampal long-term potentiation (LTP). NMDA receptor antagonists, at concentrations allowing physiological activation, can prevent Aβ-induced deficits in LTP. The anaesthetics xenon and S-ketamine both act as NMDA receptor antagonists and have been reported to be neuroprotective. In this study, we investigated the effects of subanaesthetic concentrations of these drugs on LTP deficits induced by different Aβ oligomers and compared them to the effects of radiprodil, a NMDA subunit 2B (GluN2B)-selective antagonist. METHODS We applied different Aβ oligomers to murine brain slices and recorded excitatory postsynaptic field potentials before and after high-frequency stimulation in the CA1 region of hippocampus. Radiprodil, xenon and S-ketamine were added and recordings evoked from a second input were measured. RESULTS Xenon and radiprodil, applied at low concentrations, partially restored the LTP deficit induced by pre-incubated Aβ1-42. S-ketamine showed no effect. None of the drugs tested were able to ameliorate Aβ1-40-induced LTP-deficits. CONCLUSIONS Xenon administered at subanaesthetic concentrations partially restored Aβ1-42-induced impairment of LTP, presumably via its weak NMDA receptor antagonism. The effects were in a similar range than those obtained with the NMDA-GluN2B antagonist radiprodil. Our results point to protective properties of xenon in the context of pathological distorted synaptic physiology which might be a meaningful alternative for anaesthesia in AD patients.
Collapse
Affiliation(s)
- Martina Bürge
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany; Department of Perioperative Medicine, Barts Heart Centre, St Bartholomew's Hospital, West Smithfield, London EC1A 7BE, United Kingdom.
| | - Stephan Kratzer
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Corinna Mattusch
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany; Hexal AG, Industriestr. 25, 83607 Holzkirchen, Germany
| | - Carolin Hofmann
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | - Matthias Kreuzer
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| | | | - Gerhard Rammes
- Department of Anaesthesiology and Intensive Care Medicine, Klinikum rechts der Isar, Technische Universität München, Ismaninger Str. 22, 81675 Munich, Germany
| |
Collapse
|
15
|
Rammes G, Seeser F, Mattusch K, Zhu K, Haas L, Kummer M, Heneka M, Herms J, Parsons CG. The NMDA receptor antagonist Radiprodil reverses the synaptotoxic effects of different amyloid-beta (Aβ) species on long-term potentiation (LTP). Neuropharmacology 2018; 140:184-192. [PMID: 30016667 DOI: 10.1016/j.neuropharm.2018.07.021] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/22/2018] [Revised: 06/22/2018] [Accepted: 07/14/2018] [Indexed: 12/29/2022]
Abstract
Aβ1-42 is well accepted to be a primary early pathogenic agent in Alzheimer's disease (AD). However, other amyloid peptides are now gaining considerable attention as potential key participants in AD due to their proposed higher neuronal toxicity. Impairment of the glutamatergic system is also widely accepted to be associated with pathomechanisms underlying AD. There is ample evidence that Aβ1-42 affects GLUN2B subunit containing N-methyl-D-aspartate receptor function and abolishes the induction of long term potentiation (LTP). In this study we show that different β-amyloid species, 1-42 Aβ1-42 and 1-40 (Aβ1-40) as well as post-translationally modified forms such as pyroglutamate-modified amyloid-(AβpE3) and nitrated Aβ (3NTyr10-Aβ), when applied for 90 min to murine hippocampal slices, concentration-dependently prevented the development of CA1-LTP after tetanic stimulation of the Schaffer collaterals with IC50s of 2, 9, 2 and 35 nM, respectively whilst having no effect on baseline AMPA receptor mediated fEPSPs. Aβ1-43 had no effect. Interestingly, the combination of all Aβ species did not result in any synergistic or additive inhibitory effect on LTP - the calculated pooled Aβ species IC50 was 20 nM. A low concentration (10 nM) of the GLUN2B receptor antagonist Radiprodil restored LTP in the presence of Aβ1-42, 3NTyr10-Aβ, Aβ1-40, but not AβpE3. In contrast to AMPA receptor mediated fEPSPs, all different β-amyloid species tested at 50 nM supressed baseline NMDA-EPSC amplitudes. Similarly, all different Aβ species tested decreased spine density. As with LTP, Radiprodil (10 nM) reversed the synaptic toxicity of Aβ species but not that of AβpE3. These data do not support the enhanced toxic actions reported for some Aβ species such as AβpE3, nor synergistic toxicity of the combination of different Aβ species. However, whilst in our hands AβpE3-42 was actually less toxic than Aβ1-42, its effects were not reversed by Radiprodil indicating that the target receptors/subunits mediating such synaptotoxicity may differ between the different Aβ species tested.
Collapse
Affiliation(s)
- Gerhard Rammes
- Department of Anaesthesiology, Technische Universität München, Munich, Germany.
| | - Franziska Seeser
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Korinna Mattusch
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Kaichuan Zhu
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Laura Haas
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Markus Kummer
- Clinical Neuroscience Unit, Dept. of Neurology, University of Bonn, Germany
| | - Michael Heneka
- Clinical Neuroscience Unit, Dept. of Neurology, University of Bonn, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Chris G Parsons
- Non-Clinical Science, Merz Pharmaceuticals GmbH, Frankfurt am Main, Germany
| |
Collapse
|
16
|
Kodam A, Ourdev D, Maulik M, Hariharakrishnan J, Banerjee M, Wang Y, Kar S. A role for astrocyte-derived amyloid β peptides in the degeneration of neurons in an animal model of temporal lobe epilepsy. Brain Pathol 2018; 29:28-44. [PMID: 29665128 DOI: 10.1111/bpa.12617] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/13/2017] [Accepted: 04/11/2018] [Indexed: 12/13/2022] Open
Abstract
Kainic acid, an analogue of the excitatory neurotransmitter glutamate, can trigger seizures and neurotoxicity in the hippocampus and other limbic structures in a manner that mirrors the neuropathology of human temporal lobe epilepsy (TLE). However, the underlying mechanisms associated with the neurotoxicity remain unclear. Since amyloid-β (Aβ) peptides, which are critical in the development of Alzheimer's disease, can mediate toxicity by activating glutamatergic NMDA receptors, it is likely that the enhanced glutamatergic transmission that renders hippocampal neurons vulnerable to kainic acid treatment may involve Aβ peptides. Thus, we seek to establish what role Aβ plays in kainic acid-induced toxicity using in vivo and in vitro paradigms. Our results show that systemic injection of kainic acid to adult rats triggers seizures, gliosis and loss of hippocampal neurons, along with increased levels/processing of amyloid precursor protein (APP), resulting in the enhanced production of Aβ-related peptides. The changes in APP levels/processing were evident primarily in activated astrocytes, implying a role for astrocytic Aβ in kainic acid-induced toxicity. Accordingly, we showed that treating rat primary cultured astrocytes with kainic acid can lead to increased Aβ production/secretion without any compromise in cell viability. Additionally, we revealed that kainic acid reduces neuronal viability more in neuronal/astrocyte co-cultures than in pure neuronal culture, and this is attenuated by precluding Aβ production. Collectively, these results indicate that increased production/secretion of Aβ-related peptides from activated astrocytes can contribute to neurotoxicity in kainic acid-treated rats. Since kainic acid administration can lead to neuropathological changes resembling TLE, it is likely that APP/Aβ peptides derived from astrocytes may have a role in TLE pathogenesis.
Collapse
Affiliation(s)
- A Kodam
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - D Ourdev
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - M Maulik
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - J Hariharakrishnan
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - M Banerjee
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - Y Wang
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| | - S Kar
- Department of Psychiatry, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8.,Department of Medicine, University of Alberta, Edmonton, Alberta, Canada, T6G 2M8
| |
Collapse
|
17
|
Zhang D, Mably AJ, Walsh DM, Rowan MJ. Peripheral Interventions Enhancing Brain Glutamate Homeostasis Relieve Amyloid β- and TNFα- Mediated Synaptic Plasticity Disruption in the Rat Hippocampus. Cereb Cortex 2018; 27:3724-3735. [PMID: 27390019 DOI: 10.1093/cercor/bhw193] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/18/2022] Open
Abstract
Dysregulation of glutamate homeostasis in the interstitial fluid of the brain is strongly implicated in causing synaptic dysfunction in many neurological and psychiatric illnesses. In the case of Alzheimer's disease (AD), amyloid β (Aβ)-mediated disruption of synaptic plasticity and memory can be alleviated by interventions that directly remove glutamate or block certain glutamate receptors. An alternative strategy is to facilitate the removal of excess glutamate from the nervous system by activating peripheral glutamate clearance systems. One such blood-based system, glutamate oxaloacetate transaminase (GOT), is activated by oxaloacetate, which acts as a co-substrate. We report here that synthetic and AD brain-derived Aβ-mediated inhibition of synaptic long-term potentiation in the hippocampus is alleviated by oxaloacetate. Moreover the effect of oxaloacetate was GOT-dependent. The disruptive effects of a general inhibitor of excitatory amino acid transport or TNFα, a pro-inflammatory mediator of Aβ action, were also reversed by oxaloacetate. Furthermore, another intervention that increases peripheral glutamate clearance, peritoneal dialysis, mimicked the beneficial effect of oxaloacetate. These findings lend support to the promotion of the peripheral clearance of glutamate as a means to alleviate synaptic dysfunction that is caused by impaired glutamate homeostasis in the brain.
Collapse
Affiliation(s)
- Dainan Zhang
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Alexandra J Mably
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Dominic M Walsh
- Laboratory for Neurodegenerative Research, Ann Romney Center for Neurologic Diseases, Brigham & Women's Hospital, Harvard Institute of Medicine, 77 Avenue Louis Pasteur, Boston, MA 02115, USA
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| |
Collapse
|
18
|
Salgado-Puga K, Rodríguez-Colorado J, Prado-Alcalá RA, Peña-Ortega F. Subclinical Doses of ATP-Sensitive Potassium Channel Modulators Prevent Alterations in Memory and Synaptic Plasticity Induced by Amyloid-β. J Alzheimers Dis 2018; 57:205-226. [PMID: 28222502 DOI: 10.3233/jad-160543] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/31/2022]
Abstract
In addition to coupling cell metabolism and excitability, ATP-sensitive potassium channels (KATP) are involved in neural function and plasticity. Moreover, alterations in KATP activity and expression have been observed in Alzheimer's disease (AD) and during amyloid-β (Aβ)-induced pathology. Thus, we tested whether KATP modulators can influence Aβ-induced deleterious effects on memory, hippocampal network function, and plasticity. We found that treating animals with subclinical doses (those that did not change glycemia) of a KATP blocker (Tolbutamide) or a KATP opener (Diazoxide) differentially restrained Aβ-induced memory deficit, hippocampal network activity inhibition, and long-term synaptic plasticity unbalance (i.e., inhibition of LTP and promotion of LTD). We found that the protective effect of Tolbutamide against Aβ-induced memory deficit was strong and correlated with the reestablishment of synaptic plasticity balance, whereas Diazoxide treatment produced a mild protection against Aβ-induced memory deficit, which was not related to a complete reestablishment of synaptic plasticity balance. Interestingly, treatment with both KATP modulators renders the hippocampus resistant to Aβ-induced inhibition of hippocampal network activity. These findings indicate that KATP are involved in Aβ-induced pathology and they heighten the potential role of KATP modulation as a plausible therapeutic strategy against AD.
Collapse
Affiliation(s)
- Karla Salgado-Puga
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Javier Rodríguez-Colorado
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Roberto A Prado-Alcalá
- Departamento de Neurobiología Conductual y Cognitiva, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| | - Fernando Peña-Ortega
- Departamento de Neurobiología del Desarrollo y Neurofisiología, Instituto de Neurobiología, Universidad Nacional Autónoma de México, Querétaro, QRO, México
| |
Collapse
|
19
|
Handa AK, Fatima T, Mattoo AK. Polyamines: Bio-Molecules with Diverse Functions in Plant and Human Health and Disease. Front Chem 2018; 6:10. [PMID: 29468148 PMCID: PMC5807879 DOI: 10.3389/fchem.2018.00010] [Citation(s) in RCA: 159] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/13/2017] [Accepted: 01/15/2018] [Indexed: 12/13/2022] Open
Abstract
Biogenic amines-polyamines (PAs), particularly putrescine, spermidine and spermine are ubiquitous in all living cells. Their indispensable roles in many biochemical and physiological processes are becoming commonly known, including promoters of plant life and differential roles in human health and disease. PAs positively impact cellular functions in plants-exemplified by increasing longevity, reviving physiological memory, enhancing carbon and nitrogen resource allocation/signaling, as well as in plant development and responses to extreme environments. Thus, one or more PAs are commonly found in genomic and metabolomics studies using plants, particulary during different abiotic stresses. In humans, a general decline in PA levels with aging occurs parallel with some human health disorders. Also, high PA dose is detrimental to patients suffering from cancer, aging, innate immunity and cognitive impairment during Alzheimer and Parkinson diseases. A dichotomy exists in that while PAs may increase longevity and reduce some age-associated cardiovascular diseases, in disease conditions involving higher cellular proliferation, their intake has negative consequences. Thus, it is essential that PA levels be rigorously quantified in edible plant sources as well as in dietary meats. Such a database can be a guide for medical experts in order to recommend which foods/meats a patient may consume and which ones to avoid. Accordingly, designing both high and low polyamine diets for human consumption are in vogue, particularly in medical conditions where PA intake may be detrimental, for instance, cancer patients. In this review, literature data has been collated for the levels of the three main PAs, putrescine, spermidine and spermine, in different edible sources-vegetables, fruits, cereals, nuts, meat, sea food, cheese, milk, and eggs. Based on our analysis of vast literature, the effects of PAs in human/animal health fall into two broad, Yang and Yin, categories: beneficial for the physiological processes in healthy cells and detrimental under pathological conditions.
Collapse
Affiliation(s)
- Avtar K. Handa
- Department of Horticulture and Landscape Architecture, Purdue University, West Lafayette, IN, United States
| | - Tahira Fatima
- Department of Horticulture and Landscape Architecture, Purdue University, West Lafayette, IN, United States
| | - Autar K. Mattoo
- Sustainable Agricultural Systems Laboratory, Henry A. Wallace Beltsville Agricultural Research Center, Agricultural Research Service (ARS-USDA), Beltsville, MD, United States
| |
Collapse
|
20
|
Rammes G, Mattusch C, Wulff M, Seeser F, Kreuzer M, Zhu K, Deussing JM, Herms J, Parsons CG. Involvement of GluN2B subunit containing N-methyl-d-aspartate (NMDA) receptors in mediating the acute and chronic synaptotoxic effects of oligomeric amyloid-beta (Aβ) in murine models of Alzheimer's disease (AD). Neuropharmacology 2017; 123:100-115. [PMID: 28174113 DOI: 10.1016/j.neuropharm.2017.02.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/02/2016] [Revised: 01/17/2017] [Accepted: 02/03/2017] [Indexed: 01/05/2023]
Abstract
To elucidate whether a permanent reduction of the GluN2B subunit affects the pathology of Alzheimer's disease (AD), we cross-bred mice heterozygous for GluN2B receptors in the forebrain (hetGluN2B) with a mouse model for AD carrying a mutated amyloid precursor protein with the Swedish and Arctic mutation (mAPP) resulting in a hetGluN2B/mAPP transgenic. By means of voltage-sensitive dye imaging (VSDI) in the di-synaptic hippocampal pathway and the recording of field excitatory postsynaptic potentials (fEPSPs), hippocampal slices of all genotypes (WT, hetGluN2B, mAPP and hetGluN2B/mAPP, age 9-18 months) were tested for spatiotemporal activity propagation and long-term potentiation (LTP) induction. CA1-LTP induced by high frequency stimulation (HFS; 100 Hz/1s) was not different in all genotypes. Aβ1-42 (50 nM)-application reduced potentiation of fEPSP in WT and hetGluN2B/mAPP mice, LTP in mAPP and hetGluN2B mice was not affected. For VSDI a fast depolarization signal was evoked in the granule cell layer and propagation was analysed in hippocampal CA3 and CA1 region before and after theta stimulation (100pulses/5 Hz). LTP was not significantly different between all genotypes. In mAPP mice θ-stim produced an epileptiform activity reflected in a pronounced prolongation of the FDS compared to the other genotypes. In slices of hetGluN2B/mAPP and GluN2B mice, however, these parameters were similar to WT mice indicating a reversal effect of the attenuated GluN2B expression. The induction of a hetGluN2B mutation in the mAPP reversed some pathophysiological changes on hippocampal LTP and provide further evidence for the involvement of the glutamatergic system in AD and emphasize the GluN2B subunit as a potential target for AD treatment.
Collapse
Affiliation(s)
- Gerhard Rammes
- Department of Anaesthesiology, Technische Universität München, Munich, Germany; Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany.
| | - Corinna Mattusch
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Matthias Wulff
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Franziska Seeser
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Matthias Kreuzer
- Department of Anaesthesiology, Technische Universität München, Munich, Germany
| | - Kaichuan Zhu
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Jan M Deussing
- Department of Stress Neurobiology and Neurogenetics, Max Planck Institute of Psychiatry, Munich, Germany
| | - Jochen Herms
- German Center for Neurodegenerative Diseases (DZNE), Munich, Germany
| | - Chris G Parsons
- Non-Clinical Science, Merz Pharmaceuticals GmbH, Frankfurt am Main, Germany
| |
Collapse
|
21
|
The Rapid Effect of Bisphenol-A on Long-Term Potentiation in Hippocampus Involves Estrogen Receptors and ERK Activation. Neural Plast 2017; 2017:5196958. [PMID: 28255459 PMCID: PMC5307006 DOI: 10.1155/2017/5196958] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 09/12/2016] [Revised: 12/11/2016] [Accepted: 12/25/2016] [Indexed: 12/29/2022] Open
Abstract
Bisphenol-A (BPA), a widely used synthetic compound in plastics, disrupts endocrine function and interferes with physiological actions of endogenous gonadal hormones. Chronic effects of BPA on reproductive function, learning and memory, brain structure, and social behavior have been intensively investigated. However, less is known about the influence of BPA on long-term potentiation (LTP), one of the major cellular mechanisms that underlie learning and memory. In the present study, for the first time we investigated the effect of different doses of BPA on hippocampal LTP in rat brain slices. We found a biphasic effect of BPA on LTP in the dentate gyrus: exposure to BPA at a low dose (100 nM) enhanced LTP and exposure to BPA at a high dose (1000 nM) inhibited LTP compared with vehicle controls. The rapid facilitatory effect of low-dose BPA on hippocampal LTP required membrane-associated estrogen receptor (ER) and involved activation of the extracellular signal-regulated kinase (ERK) signaling pathway. Coadministration of 17β-estradiol (E2, the primary estrogen hormone) and BPA (100 nM) abolished both the BPA-induced enhancement of LTP and the E2-induced enhancement of baseline fEPSP, suggesting a complex interaction between BPA- and E2-mediated signaling pathways. Our investigation implies that even nanomolar levels of endocrine disrupters (e.g., BPA) can induce significant effects on hippocampal LTP.
Collapse
|
22
|
Zhang D, Jin B, Ondrejcak T, Rowan MJ. Opposite in vivo effects of agents that stimulate or inhibit the glutamate/cysteine exchanger system xc- on the inhibition of hippocampal LTP by Aß. Hippocampus 2016; 26:1655-1665. [PMID: 27701797 DOI: 10.1002/hipo.22667] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 09/21/2016] [Indexed: 01/17/2023]
Abstract
Aggregated amyloid ß-protein (Aß) is pathognomonic of Alzheimer's disease and certain assemblies of Aß are synaptotoxic. Excess glutamate or diminished glutathione reserve are both implicated in mediating or modulating Aß-induced disruption of synaptic plasticity. The system xc- antiporter promotes Na+ -independent exchange of cystine with glutamate thereby providing a major source of extracellular glutamate and intracellular glutathione concentrations. Here we probed the ability of two drugs with opposite effects on system xc-, the inhibitor sulfasalazine and facilitator N-acetylcysteine, to modulate the ability of Aß1-42 to inhibit long-term potentiation (LTP) in the CA1 area of the anaesthetized rat. Whereas acute systemic treatment with sulfasalazine lowered the threshold for Aß to interfere with synaptic plasticity, N-acetylcysteine prevented the inhibition of LTP by Aß alone or in combination with sulfasalazine. Moreover acute N-acetylcysteine also prevented the inhibition of LTP by TNFα, a putative mediator of Aß actions, and repeated systemic N-acetylcysteine treatment for 7 days reversed the delayed deleterious effect of Aß on LTP. Since both of these drugs are widely used clinically, further evaluation of their potential beneficial and deleterious actions in early Alzheimer's disease seems warranted. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Dainan Zhang
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland.,Department of Neurosurgery, the First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Baozhe Jin
- Department of Neurosurgery, the First Affiliated Hospital of Xinxiang Medical University, Weihui, 453100, China
| | - Tomas Ondrejcak
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics, and Trinity College Institute of Neuroscience, Trinity College, Dublin 2, Ireland
| |
Collapse
|
23
|
Guerra GP, Rubin MA, Mello CF. Modulation of learning and memory by natural polyamines. Pharmacol Res 2016; 112:99-118. [PMID: 27015893 DOI: 10.1016/j.phrs.2016.03.023] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 01/03/2016] [Revised: 03/09/2016] [Accepted: 03/11/2016] [Indexed: 01/08/2023]
Abstract
Spermine and spermidine are natural polyamines that are produced mainly via decarboxylation of l-ornithine and the sequential transfer of aminopropyl groups from S-adenosylmethionine to putrescine by spermidine synthase and spermine synthase. Spermine and spermidine interact with intracellular and extracellular acidic residues of different nature, including nucleic acids, phospholipids, acidic proteins, carboxyl- and sulfate-containing polysaccharides. Therefore, multiple actions have been suggested for these polycations, including modulation of the activity of ionic channels, protein synthesis, protein kinases, and cell proliferation/death, within others. In this review we summarize these neurochemical/neurophysiological/morphological findings, particularly those that have been implicated in the improving and deleterious effects of spermine and spermidine on learning and memory of naïve animals in shock-motivated and nonshock-motivated tasks, from a historical perspective. The interaction with the opioid system, the facilitation and disruption of morphine-induced reward and the effect of polyamines and putative polyamine antagonists on animal models of cognitive diseases, such as Alzheimer's, Huntington, acute neuroinflammation and brain trauma are also reviewed and discussed. The increased production of polyamines in Alzheimer's disease and the biphasic nature of the effects of polyamines on memory and on the NMDA receptor are also considered. In light of the current literature on polyamines, which include the description of an inborn error of the metabolism characterized by mild-to moderate mental retardation and polyamine metabolism alterations in suicide completers, we can anticipate that polyamine targets may be important for the development of novel strategies and approaches for understanding the etiopathogenesis of important central disorders and their pharmacological treatment.
Collapse
Affiliation(s)
- Gustavo Petri Guerra
- Department of Food Technology, Federal Technological University of Paraná, Campus Medianeira, Medianeira, PR 85884-000, Brazil
| | - Maribel Antonello Rubin
- Department of Biochemistry, Center of Exact and Natural Sciences, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil.
| | - Carlos Fernando Mello
- Department of Physiology and Pharmacology, Center of Health Sciences, Federal University of Santa Maria (UFSM), Santa Maria, RS 97105-900, Brazil.
| |
Collapse
|
24
|
Jia XT, Ye-Tian, Yuan-Li, Zhang GJ, Liu ZQ, Di ZL, Ying XP, Fang Y, Song EF, Qi JS, Pan YF. Exendin-4, a glucagon-like peptide 1 receptor agonist, protects against amyloid-β peptide-induced impairment of spatial learning and memory in rats. Physiol Behav 2016; 159:72-9. [PMID: 26992957 DOI: 10.1016/j.physbeh.2016.03.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/23/2015] [Revised: 03/10/2016] [Accepted: 03/14/2016] [Indexed: 02/08/2023]
Abstract
Type 2 diabetes mellitus (T2DM) and Alzheimer's disease (AD) share specific molecular mechanisms, and agents with proven efficacy in one may be useful against the other. The glucagon-like peptide-1 (GLP-1) receptor agonist exendin-4 has similar properties to GLP-1 and is currently in clinical use for T2DM treatment. Thus, this study was designed to characterize the effects of exendin-4 on the impairment of learning and memory induced by amyloid protein (Aβ) and its probable molecular underlying mechanisms. The results showed that (1) intracerebroventricular (i.c.v.) injection of Aβ1-42 resulted in a significant decline of spatial learning and memory of rats in water maze tests; (2) pretreatment with exendin-4 effectively and dose-dependently protected against the Aβ1-42-induced impairment of spatial learning and memory; (3) exendin-4 treatment significantly decreased the expression of Bax and cleaved caspase-3 and increased the expression of Bcl2 in Aβ1-42-induced Alzheimer's rats. The vision and swimming speed of the rats among all groups in the visible platform tests did not show any difference. These findings indicate that systemic pretreatment with exendin-4 can effectively prevent the behavioral impairment induced by neurotoxic Aβ1-42, and the underlying protective mechanism of exendin-4 may be involved in the Bcl2, Bax and caspase-3 pathways. Thus, the application of exendin-4 or the activation of its signaling pathways may be a promising strategy to ameliorate the degenerative processes observed in AD.
Collapse
Affiliation(s)
- Xiao-Tao Jia
- Department of Neurology, The Affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, PR China
| | - Ye-Tian
- Department of Neurology, The Affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, PR China
| | - Yuan-Li
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, PR China
| | - Ge-Juan Zhang
- Department of Neurology, The Affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, PR China
| | - Zhi-Qin Liu
- Department of Neurology, The Affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, PR China
| | - Zheng-Li Di
- Department of Neurology, The Affiliated Xi'an Central Hospital of Xi'an Jiaotong University College of Medicine, Xi'an, Shaanxi 710003, PR China
| | - Xiao-Ping Ying
- Department of Pathology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, PR China
| | - Yan Fang
- Department of Pathology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, PR China
| | - Er-Fei Song
- Department of Biology, York University, Toronto, ON M3J 1P3, Canada
| | - Jin-Shun Qi
- Department of Physiology, Key Laboratory of Cellular Physiology, Ministry of Education, Shanxi Medical University, Taiyuan 030001, PR China.
| | - Yan-Fang Pan
- Department of Pathology, Shaanxi University of Chinese Medicine, Xianyang, Shaanxi 712046, PR China.
| |
Collapse
|
25
|
Memantine alters striatal plasticity inducing a shift of synaptic responses toward long-term depression. Neuropharmacology 2016; 101:341-50. [DOI: 10.1016/j.neuropharm.2015.10.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/17/2015] [Revised: 10/07/2015] [Accepted: 10/08/2015] [Indexed: 01/11/2023]
|
26
|
Kaleka KS, Gerges NZ. Neurogranin restores amyloid β-mediated synaptic transmission and long-term potentiation deficits. Exp Neurol 2015; 277:115-123. [PMID: 26721336 DOI: 10.1016/j.expneurol.2015.12.013] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/29/2015] [Revised: 12/08/2015] [Accepted: 12/18/2015] [Indexed: 11/16/2022]
Abstract
Amyloid β (Aβ) is widely considered one of the early causes of cognitive deficits observed in Alzheimer's disease. Many of the deficits caused by Aβ are attributed to its disruption of synaptic function represented by its blockade of long-term potentiation (LTP) and its induction of synaptic depression. Identifying pathways that reverse these synaptic deficits may open the door to new therapeutic targets. In this study, we explored the possibility that Neurogranin (Ng)-a postsynaptic calmodulin (CaM) targeting protein that enhances synaptic function-may rescue Aβ-mediated deficits in synaptic function. Our results show that Ng is able to reverse synaptic depression and LTP deficits induced by Aβ. Furthermore, Ng's restoration of synaptic transmission is through the insertion of GluA1-containing α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors (AMPARs). These restorative effects of Ng are dependent on the interaction of Ng and CaM and CaM-dependent activation of CaMKII. Overall, this study identifies a novel mechanism to rescue synaptic deficits induced by Aβ oligomers. It also suggests Ng and CaM signaling as potential therapeutic targets for Alzheimer's disease as well as important tools to further explore the pathophysiology underlying the disease.
Collapse
Affiliation(s)
- Kanwardeep Singh Kaleka
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States
| | - Nashaat Z Gerges
- Department of Cell Biology, Neurobiology and Anatomy, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States; Neuroscience Research Center, Medical College of Wisconsin, 8701 West Watertown Plank Rd., Milwaukee, WI 53132, United States.
| |
Collapse
|
27
|
Effects of memantine on hippocampal long-term potentiation, gamma activity, and sensorimotor gating in freely moving rats. Neurobiol Aging 2015; 36:2544-54. [DOI: 10.1016/j.neurobiolaging.2015.05.017] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/12/2015] [Revised: 05/25/2015] [Accepted: 05/29/2015] [Indexed: 12/20/2022]
|
28
|
Harte NP, Klyubin I, McCarthy EK, Min S, Garrahy SA, Xie Y, Davey GP, Boland JJ, Rowan MJ, Mok KH. Amyloid Oligomers and Mature Fibrils Prepared from an Innocuous Protein Cause Diverging Cellular Death Mechanisms. J Biol Chem 2015. [PMID: 26221033 DOI: 10.1074/jbc.m115.676072] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/22/2022] Open
Abstract
Despite significant advances, the molecular identity of the cytotoxic species populated during in vivo amyloid formation crucial for the understanding of neurodegenerative disorders is yet to be revealed. In this study lysozyme prefibrillar oligomers and fibrils in both mature and sonicated states have been isolated through an optimized ultrafiltration/ultracentrifugation method and characterized with various optical spectroscopic techniques, atomic force microscopy, and transmission electron microscopy. We examined their level and mode of toxicity on rat pheochromocytoma (PC12) cells in both differentiated and undifferentiated states. We find that oligomers and fibrils display cytotoxic capabilities toward cultured cells in vitro, with oligomers producing elevated levels of cellular injury toward undifferentiated PC12 cells (PC12(undiff)). Furthermore, dual flow cytometry staining experiments demonstrate that the oligomers and mature fibrils induce divergent cellular death pathways (apoptosis and secondary necrosis, respectively) in these PC12 cells. We have also shown that oligomers but not sonicated mature fibrils inhibit hippocampal long term potentiation, a form of synaptic plasticity implicated in learning and memory, in vivo. We conclude that our in vitro and in vivo findings confer a level of resistance toward amyloid fibrils, and that the PC 12-based comparative cytotoxicity assay can provide insights into toxicity differences between differently aggregated protein species.
Collapse
Affiliation(s)
- Níal P Harte
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Igor Klyubin
- Department of Pharmacology and Therapeutics, Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Eoin K McCarthy
- School of Chemistry, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Soyoung Min
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Sarah Ann Garrahy
- TCIN, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Yongjing Xie
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Gavin P Davey
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland; TCIN, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - John J Boland
- School of Chemistry, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - Michael J Rowan
- Department of Pharmacology and Therapeutics, Trinity College Institute of Neuroscience (TCIN), Trinity College Dublin, the University of Dublin, Dublin 2, Ireland
| | - K Hun Mok
- Trinity Biomedical Sciences Institute (TBSI), School of Biochemistry and Immunology, Trinity College Dublin, the University of Dublin, Dublin 2, Ireland; Centre for Research on Adaptive Nanostructures and Nanodevices (CRANN), Trinity College Dublin, the University of Dublin, Dublin 2, Ireland.
| |
Collapse
|
29
|
Protection against β-amyloid-induced synaptic and memory impairments via altering β-amyloid assembly by bis(heptyl)-cognitin. Sci Rep 2015; 5:10256. [PMID: 26194093 PMCID: PMC4508546 DOI: 10.1038/srep10256] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/14/2015] [Accepted: 04/07/2015] [Indexed: 12/15/2022] Open
Abstract
β-amyloid (Aβ) oligomers have been closely implicated in the pathogenesis of Alzheimer’s disease (AD). We found, for the first time, that bis(heptyl)-cognitin, a novel dimeric acetylcholinesterase (AChE) inhibitor derived from tacrine, prevented Aβ oligomers-induced inhibition of long-term potentiation (LTP) at concentrations that did not interfere with normal LTP. Bis(heptyl)-cognitin also prevented Aβ oligomers-induced synaptotoxicity in primary hippocampal neurons. In contrast, tacrine and donepezil, typical AChE inhibitors, could not prevent synaptic impairments in these models, indicating that the modification of Aβ oligomers toxicity by bis(heptyl)-cognitin might be attributed to a mechanism other than AChE inhibition. Studies by using dot blotting, immunoblotting, circular dichroism spectroscopy, and transmission electron microscopy have shown that bis(heptyl)-cognitin altered Aβ assembly via directly inhibiting Aβ oligomers formation and reducing the amount of preformed Aβ oligomers. Molecular docking analysis further suggested that bis(heptyl)-cognitin presumably interacted with the hydrophobic pockets of Aβ, which confers stabilizing powers and assembly alteration effects on Aβ. Most importantly, bis(heptyl)-cognitin significantly reduced cognitive impairments induced by intra-hippocampal infusion of Aβ oligomers in mice. These results clearly demonstrated how dimeric agents prevent Aβ oligomers-induced synaptic and memory impairments, and offered a strong support for the beneficial therapeutic effects of bis(heptyl)-cognitin in the treatment of AD.
Collapse
|
30
|
Jiang L, Huang M, Xu S, Wang Y, An P, Feng C, Chen X, Wei X, Han Y, Wang Q. Bis(propyl)-cognitin Prevents β-amyloid-induced Memory Deficits as Well as Synaptic Formation and Plasticity Impairments via the Activation of PI3-K Pathway. Mol Neurobiol 2015; 53:3832-3841. [DOI: 10.1007/s12035-015-9317-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 04/26/2015] [Accepted: 06/23/2015] [Indexed: 01/09/2023]
|
31
|
Portbury SD, Adlard PA. Traumatic Brain Injury, Chronic Traumatic Encephalopathy, and Alzheimer’s Disease: Common Pathologies Potentiated by Altered Zinc Homeostasis. J Alzheimers Dis 2015; 46:297-311. [DOI: 10.3233/jad-143048] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/14/2022]
|
32
|
Trotman M, Vermehren P, Gibson CL, Fern R. The dichotomy of memantine treatment for ischemic stroke: dose-dependent protective and detrimental effects. J Cereb Blood Flow Metab 2015; 35:230-9. [PMID: 25407270 PMCID: PMC4426739 DOI: 10.1038/jcbfm.2014.188] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 08/14/2014] [Revised: 10/02/2014] [Accepted: 10/03/2014] [Indexed: 12/23/2022]
Abstract
Excitotoxicity is a major contributor to cell death during the acute phase of ischemic stroke but aggressive pharmacological targeting of excitotoxicity has failed clinically. Here we investigated whether pretreatment with low doses of memantine, within the range currently used and well tolerated for the treatment of Alzheimer's disease, produce a protective effect in stroke. A coculture preparation exposed to modeled ischemia showed cell death associated with rapid glutamate rises and cytotoxic Ca(2+) influx. Cell death was significantly enhanced in the presence of high memantine concentrations. However, low memantine concentrations significantly protected neurons and glia via excitotoxic cascade interruption. Mice were systemically administered a range of memantine doses (0.02, 0.2, 2, 10, and 20 mg/kg/day) starting 24 hours before 60 minutes reversible focal cerebral ischemia and continuing for a 48-hour recovery period. Low dose (0.2 mg/kg/day) memantine treatment significantly reduced lesion volume (by 30% to 50%) and improved behavioral outcomes in stroke lesions that had been separated into either small/striatal or large/striatocortical infarcts. However, higher doses of memantine (20 mg/kg/day) significantly increased injury. These results show that clinically established low doses of memantine should be considered for patients 'at risk' of stroke, while higher doses are contraindicated.
Collapse
Affiliation(s)
- Melissa Trotman
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | - Philipp Vermehren
- Department of Cell Physiology and Pharmacology, University of Leicester, Leicester, UK
| | - Claire L Gibson
- School of Psychology, University of Leicester, Leicester, UK
| | - Robert Fern
- Peninsula School of Medicine and Dentistry, University of Plymouth, Plymouth, UK
| |
Collapse
|
33
|
Izzo NJ, Staniszewski A, To L, Fa M, Teich AF, Saeed F, Wostein H, Walko T, Vaswani A, Wardius M, Syed Z, Ravenscroft J, Mozzoni K, Silky C, Rehak C, Yurko R, Finn P, Look G, Rishton G, Safferstein H, Miller M, Johanson C, Stopa E, Windisch M, Hutter-Paier B, Shamloo M, Arancio O, LeVine H, Catalano SM. Alzheimer's therapeutics targeting amyloid beta 1-42 oligomers I: Abeta 42 oligomer binding to specific neuronal receptors is displaced by drug candidates that improve cognitive deficits. PLoS One 2014; 9:e111898. [PMID: 25390368 PMCID: PMC4229098 DOI: 10.1371/journal.pone.0111898] [Citation(s) in RCA: 105] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/19/2014] [Accepted: 10/02/2014] [Indexed: 01/09/2023] Open
Abstract
Synaptic dysfunction and loss caused by age-dependent accumulation of synaptotoxic beta amyloid (Abeta) 1-42 oligomers is proposed to underlie cognitive decline in Alzheimer's disease (AD). Alterations in membrane trafficking induced by Abeta oligomers mediates reduction in neuronal surface receptor expression that is the basis for inhibition of electrophysiological measures of synaptic plasticity and thus learning and memory. We have utilized phenotypic screens in mature, in vitro cultures of rat brain cells to identify small molecules which block or prevent the binding and effects of Abeta oligomers. Synthetic Abeta oligomers bind saturably to a single site on neuronal synapses and induce deficits in membrane trafficking in neuronal cultures with an EC50 that corresponds to its binding affinity. The therapeutic lead compounds we have found are pharmacological antagonists of Abeta oligomers, reducing the binding of Abeta oligomers to neurons in vitro, preventing spine loss in neurons and preventing and treating oligomer-induced deficits in membrane trafficking. These molecules are highly brain penetrant and prevent and restore cognitive deficits in mouse models of Alzheimer's disease. Counter-screening these compounds against a broad panel of potential CNS targets revealed they are highly potent and specific ligands of the sigma-2/PGRMC1 receptor. Brain concentrations of the compounds corresponding to greater than 80% receptor occupancy at the sigma-2/PGRMC1 receptor restore cognitive function in transgenic hAPP Swe/Ldn mice. These studies demonstrate that synthetic and human-derived Abeta oligomers act as pharmacologically-behaved ligands at neuronal receptors--i.e. they exhibit saturable binding to a target, they exert a functional effect related to their binding and their displacement by small molecule antagonists blocks their functional effect. The first-in-class small molecule receptor antagonists described here restore memory to normal in multiple AD models and sustain improvement long-term, representing a novel mechanism of action for disease-modifying Alzheimer's therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Izzo
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| | - Agnes Staniszewski
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Lillian To
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Mauro Fa
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Andrew F. Teich
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Faisal Saeed
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harrison Wostein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Thomas Walko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Anisha Vaswani
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Meghan Wardius
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Zanobia Syed
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Jessica Ravenscroft
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Kelsie Mozzoni
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Colleen Silky
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Patricia Finn
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Rishton
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Hank Safferstein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Miles Miller
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Conrad Johanson
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | - Edward Stopa
- Department of Pathology and Neurosurgery, The Warren Alpert Medical School of Brown University, Providence, Rhode Island, United States of America
| | | | | | - Mehrdad Shamloo
- Stanford University Medical School Behavioral and Functional Neuroscience Laboratory, Palo Alto, California, United States of America
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, New York, United States of America
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Susan M. Catalano
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
34
|
Izzo NJ, Xu J, Zeng C, Kirk MJ, Mozzoni K, Silky C, Rehak C, Yurko R, Look G, Rishton G, Safferstein H, Cruchaga C, Goate A, Cahill MA, Arancio O, Mach RH, Craven R, Head E, LeVine H, Spires-Jones TL, Catalano SM. Alzheimer's therapeutics targeting amyloid beta 1-42 oligomers II: Sigma-2/PGRMC1 receptors mediate Abeta 42 oligomer binding and synaptotoxicity. PLoS One 2014; 9:e111899. [PMID: 25390692 PMCID: PMC4229119 DOI: 10.1371/journal.pone.0111899] [Citation(s) in RCA: 135] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/14/2014] [Accepted: 10/02/2014] [Indexed: 12/18/2022] Open
Abstract
Amyloid beta (Abeta) 1-42 oligomers accumulate in brains of patients with Mild Cognitive Impairment (MCI) and disrupt synaptic plasticity processes that underlie memory formation. Synaptic binding of Abeta oligomers to several putative receptor proteins is reported to inhibit long-term potentiation, affect membrane trafficking and induce reversible spine loss in neurons, leading to impaired cognitive performance and ultimately to anterograde amnesia in the early stages of Alzheimer's disease (AD). We have identified a receptor not previously associated with AD that mediates the binding of Abeta oligomers to neurons, and describe novel therapeutic antagonists of this receptor capable of blocking Abeta toxic effects on synapses in vitro and cognitive deficits in vivo. Knockdown of sigma-2/PGRMC1 (progesterone receptor membrane component 1) protein expression in vitro using siRNA results in a highly correlated reduction in binding of exogenous Abeta oligomers to neurons of more than 90%. Expression of sigma-2/PGRMC1 is upregulated in vitro by treatment with Abeta oligomers, and is dysregulated in Alzheimer's disease patients' brain compared to age-matched, normal individuals. Specific, high affinity small molecule receptor antagonists and antibodies raised against specific regions on this receptor can displace synthetic Abeta oligomer binding to synaptic puncta in vitro and displace endogenous human AD patient oligomers from brain tissue sections in a dose-dependent manner. These receptor antagonists prevent and reverse the effects of Abeta oligomers on membrane trafficking and synapse loss in vitro and cognitive deficits in AD mouse models. These findings suggest sigma-2/PGRMC1 receptors mediate saturable oligomer binding to synaptic puncta on neurons and that brain penetrant, small molecules can displace endogenous and synthetic oligomers and improve cognitive deficits in AD models. We propose that sigma-2/PGRMC1 is a key mediator of the pathological effects of Abeta oligomers in AD and is a tractable target for small molecule disease-modifying therapeutics.
Collapse
Affiliation(s)
- Nicholas J. Izzo
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Jinbin Xu
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Chenbo Zeng
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Molly J. Kirk
- Departments of Neurology and Neuroscience, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Department of Neurology, Northeastern University, Boston, Massachusetts, United States of America
| | - Kelsie Mozzoni
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Colleen Silky
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Courtney Rehak
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Raymond Yurko
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gary Look
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Gilbert Rishton
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Hank Safferstein
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| | - Carlos Cruchaga
- Department of Psychiatry, Washington University, St. Louis, Missouri, United States of America
| | - Alison Goate
- Department of Psychiatry, Washington University, St. Louis, Missouri, United States of America
| | - Michael A. Cahill
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga New South Wales, Australia
| | - Ottavio Arancio
- Department of Pathology and Cell Biology and Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University New York, New York, United States of America
| | - Robert H. Mach
- Mallinckrodt Institute of Radiology, Washington University, St. Louis, Missouri, United States of America
| | - Rolf Craven
- Department of Molecular and Biological Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Elizabeth Head
- Department of Molecular and Biological Pharmacology, University of Kentucky, Lexington, Kentucky, United States of America
| | - Harry LeVine
- Sanders-Brown Center on Aging, University of Kentucky, Lexington, Kentucky, United States of America
| | - Tara L. Spires-Jones
- Departments of Neurology and Neuroscience, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- The University of Edinburgh, Center for Cognitive and Neural Systems and Euan MacDonald Centre for Motorneurone Disease, Edinburgh, Scotland
| | - Susan M. Catalano
- Cognition Therapeutics Inc., Pittsburgh, Pennsylvania, United States of America
| |
Collapse
|
35
|
Li F, Chen X, Wang F, Xu S, Chang L, Anwyl R, Wang Q. Chronic pre-treatment with memantine prevents amyloid-beta protein-mediated long-term potentiation disruption. Neural Regen Res 2014; 8:49-55. [PMID: 25206371 PMCID: PMC4107499 DOI: 10.3969/j.issn.1673-5374.2013.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/01/2012] [Accepted: 10/10/2012] [Indexed: 01/13/2023] Open
Abstract
Previous studies indicate that memantine, a low-affinity N-methyl-D-aspartate receptor antagonist, exerted acute protective effects against amyloid-β protein-induced neurotoxicity. In the present study, the chronic effects and mechanisms of memantine were investigated further using electrophysiological methods. The results showed that 7-day intraperitoneal application of memantine, at doses of 5 mg/kg or 20 mg/kg, did not alter hippocampal long-term potentiation induction in rats, while 40 mg/kg memantine presented potent long-term potentiation inhibition. Then further in vitro studys were carried out in 5 mg/kg and 20 mg/kg memantine treated rats. We found that 20 mg/kg memantine attenuated the potent long-term potentiation inhibition caused by exposure to amyloid-β protein in the dentate gyrus in vitro. These findings are the first to demonstrate the antagonizing effect of long-term systematic treatment of memantine against amyloid-β protein triggered long-term potentiation inhibition to improve synaptic plasticity.
Collapse
Affiliation(s)
- Fushun Li
- Department of Physiology, Medical School, Research Center of Behavioral Science, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Xiaowei Chen
- Department of Physiology, Medical School, Research Center of Behavioral Science, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Feiming Wang
- Zhenhai High School, Ningbo 315211, Zhejiang Province, China
| | - Shujun Xu
- Department of Physiology, Medical School, Research Center of Behavioral Science, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Lan Chang
- Department of Physiology, Medical School, Research Center of Behavioral Science, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Roger Anwyl
- Trinity College Institute of Neuroscience, Department of Physiology, Trinity College, Dublin, Ireland
| | - Qinwen Wang
- Department of Physiology, Medical School, Research Center of Behavioral Science, Ningbo University, Ningbo 315211, Zhejiang Province, China
| |
Collapse
|
36
|
Köktürk S, Ceylan S, Etus V, Yasa N, Ceylan S. Morinda citrifolia L. (noni) and memantine attenuate periventricular tissue injury of the fourth ventricle in hydrocephalic rabbits. Neural Regen Res 2014; 8:773-82. [PMID: 25206724 PMCID: PMC4146082 DOI: 10.3969/j.issn.1673-5374.2013.09.001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/23/2012] [Accepted: 02/24/2013] [Indexed: 11/22/2022] Open
Abstract
This study was designed to evaluate the neuroprotective effects of Morinda citrifolia L. (Rubiaceae), commonly known as noni, and memantine (a N-methy-D-aspartate receptor inhibitor) on hydrocephalus-induced neurodegenerative disorders. Kaolin was injected into the cistern magna of male adult New Zealand rabbits to establish a hydrocephalus animal model. Memantine (20 mg/kg, intraperitoneally; memantine-treated group) or noni (5 mL/kg, intragastrically; noni-treated group) was administered daily for 2 weeks. Microtubule-associated protein-2 and caspase-3 immunohistochemistry were performed to detect neuronal degeneration and apoptosis in the periventricular tissue of the fourth ventricle of rabbits. Microtubule-associated protein-2 staining density was significantly decreased in the hydrocephalic group, while the staining density was significantly increased in the memantine- and noni-treated groups, especially in the noni-treated group. Noni treatment decreased the number of caspase-3-positive cells in rabbits with hydrocephalus, while memantine had no effect. These findings suggest that noni exhibits more obvious inhibitory effects on hydrocephalus-induced neurodegenerative disorders than memantine in periventricular tissue of the fourth ventricle.
Collapse
Affiliation(s)
- Sibel Köktürk
- Department of Histology and Embriyology, Faculty of Medicine, Ordu University, Ordu, Turkey
| | - Süreyya Ceylan
- Department of Histology and Embriyology, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Volkan Etus
- Department of Neurosurgery, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Nezih Yasa
- Department of Neurosurgery, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| | - Savaş Ceylan
- Department of Neurosurgery, Faculty of Medicine, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
37
|
Monaco J, Casellato C, Koch G, D'Angelo E. Cerebellar theta burst stimulation dissociates memory components in eyeblink classical conditioning. Eur J Neurosci 2014; 40:3363-70. [DOI: 10.1111/ejn.12700] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/03/2014] [Revised: 07/24/2014] [Accepted: 07/25/2014] [Indexed: 11/27/2022]
Affiliation(s)
- J. Monaco
- Brain Connectivity Center; C. Mondino National Neurological Institute; Via Mondino 2 Pavia I-27100 Italy
| | - C. Casellato
- NeuroEngineering and Medical Robotics Laboratory; Department of Electronics; Information and Bioengineering; Politecnico di Milano; Milano Italy
| | - G. Koch
- Non-invasive Brain Stimulation Unit; Santa Lucia Foundation IRCCS; Via Ardeatina 306 00179 Rome Italy
| | - E. D'Angelo
- Brain Connectivity Center; C. Mondino National Neurological Institute; Via Mondino 2 Pavia I-27100 Italy
- Department of Brain and Behavioral Sciences; University of Pavia; Via Forlanini 6 Pavia I-27100 Italy
| |
Collapse
|
38
|
Tozzi A, Sclip A, Tantucci M, de Iure A, Ghiglieri V, Costa C, Di Filippo M, Borsello T, Calabresi P. Region- and age-dependent reductions of hippocampal long-term potentiation and NMDA to AMPA ratio in a genetic model of Alzheimer's disease. Neurobiol Aging 2014; 36:123-33. [PMID: 25104560 DOI: 10.1016/j.neurobiolaging.2014.07.002] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/25/2014] [Revised: 05/15/2014] [Accepted: 07/08/2014] [Indexed: 12/14/2022]
Abstract
To characterize the mechanisms underlying region- and age-dependent hippocampal synaptic dysfunction in Alzheimer's disease, we used transgenic CRND8 mice, expressing the Swedish-Indiana APP mutation. In 2-month-old mice, no β-amyloid plaques deposition, but the presence of soluble oligomers, were found in CA1 area but not in dentate gyrus (DG). At this age, long-term potentiation (LTP) was reduced selectively in CA1. In 6-month-old mice, the presence of soluble oligomers was accompanied by accumulation of β-amyloid plaques and decreased LTP in CA1 and DG regions. In both regions, the loss of LTP was linked to reduced N-methyl-D-aspartate (NMDA) to alpha-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) current ratio. The acetylcholine-esterase inhibitor, neostigmine rescued LTP in CA1 area at early stage of the disease but not after plaques deposition. Conversely, the NMDA receptor antagonist memantine restored LTP selectively in DG at later stages of the disease. Both these effects were associated with a normalization of the NMDA to AMPA ratio. The association between the recovery of LTP and the normalization of the NMDA to AMPA ratio provides information on new possible therapeutic strategies in Alzheimer's disease.
Collapse
Affiliation(s)
- Alessandro Tozzi
- Clinica Neurologica, Università di Perugia, Department of Medicine, Ospedale S. Maria della Misericordia, Perugia, Italy; Laboratory of Neurophysiology, IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Alessandra Sclip
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Michela Tantucci
- Clinica Neurologica, Università di Perugia, Department of Medicine, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Antonio de Iure
- Clinica Neurologica, Università di Perugia, Department of Medicine, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Veronica Ghiglieri
- Laboratory of Neurophysiology, IRCCS, Fondazione Santa Lucia, Rome, Italy
| | - Cinzia Costa
- Clinica Neurologica, Università di Perugia, Department of Medicine, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Massimiliano Di Filippo
- Clinica Neurologica, Università di Perugia, Department of Medicine, Ospedale S. Maria della Misericordia, Perugia, Italy
| | - Tiziana Borsello
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche "Mario Negri", Milan, Italy
| | - Paolo Calabresi
- Clinica Neurologica, Università di Perugia, Department of Medicine, Ospedale S. Maria della Misericordia, Perugia, Italy; Laboratory of Neurophysiology, IRCCS, Fondazione Santa Lucia, Rome, Italy.
| |
Collapse
|
39
|
Potassium 2-(1-hydroxypentyl)-benzoate promotes long-term potentiation in Aβ1-42-injected rats and APP/PS1 transgenic mice. Acta Pharmacol Sin 2014; 35:869-78. [PMID: 24858312 DOI: 10.1038/aps.2014.29] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/02/2013] [Accepted: 03/12/2014] [Indexed: 01/13/2023] Open
Abstract
AIM Potassium 2-(1-hydroxypentyl)-benzoate (dl-PHPB) is a new drug candidate for ischemic stroke. The aim of this study was to investigate the effects of dl-PHPB on memory deficits and long-term potentiation (LTP) impairment in animal models of Alzheimer's disease. METHODS The expression of NMDA receptor subunits GluN1 and GluN2B in the hippocampus and cortex of APP/PS1 transgenic mice were detected using Western blot analysis. Memory deficits of the mice were evaluated with the passive avoidance test. LTP impairment was studied in the dentate region of Aβ1-42-injected rats and APP/PS1 transgenic mice. RESULTS APP/PS1 transgenic mice showed significantly lower levels of GluN1 and p-GluN2B in hippocampus, and chronic administration of dl-PHPB (100 mg · kg(-1) · d(-1), po) reversed the downregulation of p-GluN2B, but did not change GluN1 level in the hippocampus. Furthermore, chronic administration of dl-PHPB reversed the memory deficits in APP/PS1 transgenic mice. In the dentate region of normal rats, injection of dl-PHPB (100 μmol/L, icv) did not change the basal synaptic transmission, but significantly enhanced the high-frequency stimulation (HFS)-induced LTP, which was completely prevented by pre-injection of APV (150 μmol/L, icv). Chronic administration of dl-PHPB (100 mg · kg(-1) · d(-1), po) reversed LTP impairment in Aβ1-42-injected normal rats and APP/PS1 transgenic mice. CONCLUSION Chronic administration of dl-PHPB improves learning and memory and promotes LTP in the animal models of Alzheimer's disease, possibly via increasing p-GluN2B expression in the hippocampus.
Collapse
|
40
|
Zhang J, Wang C, Deng T, Xue Z, Chen X, Chang L, Wang Q. The preventive effect of NR2B and NR2D-containing NMDAR antagonists on Aβ-induced LTP disruption in the dentate gyrus of rats. Metab Brain Dis 2013; 28:697-704. [PMID: 23975536 DOI: 10.1007/s11011-013-9424-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Received: 04/01/2013] [Accepted: 07/17/2013] [Indexed: 01/10/2023]
Abstract
Amyloid β-protein (Aβ) in the brain of Alzheimer's disease (AD) potently inhibits the synaptic plasticity subsequently causing the cognitive deficits. Long-term potentiation (LTP) of synaptic transmission is thought to be an important cellular mechanism underlying memory formation. Different NR2 subunits are involved in NMDA receptor-dependent LTP. In the present study, we investigated the roles of NR2B and NR2D-containing NMDAR on Aβ(1-42)-induced LTP deficits in the hippocampal slices of rats by using selective NMDAR antagonists. First, we found that Aβ(1-42) significantly inhibited the LTP in the dentate gyrus of slices as reported before. Following that the Aβ(1-42)-induced LTP inhibition was prevented by the pre-perfusion of the specific NR2B-containing NMDAR antagonists ifenprodil (approximately >200-fold selectivity for NR2B) and Ro25-6981 (>3,000-fold selectivity for NR2B), as well as PPDA, a specific NR2D receptor antagonist. Meanwhile, the antagonists on their own had no or only partial effects on the normal LTP in the same dose condition. These findings not only support the effects of NR2B and NR2D subunits on Aβ(1-42)-induced LTP deficits, but also imply that preferentially targeting NR2B- and NR2D-containing NMDARs may provide an effective means to prevent cognitive deficits in the early AD.
Collapse
Affiliation(s)
- Junfang Zhang
- Zhejiang Provincial Key Laboratory of Pathophysiology, Department of Physiology and Pharmacology, School of Medicine, Ningbo University, No.818 Fenghua Road, Ningbo, Zhejiang, China, 315211
| | | | | | | | | | | | | |
Collapse
|
41
|
Jensen LE, Bultynck G, Luyten T, Amijee H, Bootman MD, Roderick HL. Alzheimer's disease-associated peptide Aβ42 mobilizes ER Ca(2+) via InsP3R-dependent and -independent mechanisms. Front Mol Neurosci 2013; 6:36. [PMID: 24204331 PMCID: PMC3817845 DOI: 10.3389/fnmol.2013.00036] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/17/2013] [Accepted: 10/14/2013] [Indexed: 11/13/2022] Open
Abstract
Dysregulation of Ca2+ homeostasis is considered to contribute to the toxic action of the Alzheimer's disease (AD)-associated amyloid-β-peptide (Aβ). Ca2+ fluxes across the plasma membrane and release from intracellular stores have both been reported to underlie the Ca2+ fluxes induced by Aβ42. Here, we investigated the contribution of Ca2+ release from the endoplasmic reticulum (ER) to the effects of Aβ42 upon Ca2+ homeostasis and the mechanism by which Aβ42 elicited these effects. Consistent with previous reports, application of soluble oligomeric forms of Aβ42 induced an elevation in intracellular Ca2+. The Aβ42-stimulated Ca2+ signals persisted in the absence of extracellular Ca2+ indicating a significant contribution of Ca2+ release from the ER Ca2+ store to the generation of these signals. Moreover, inositol 1,4,5-trisphosphate (InsP3) signaling contributed to Aβ42-stimulated Ca2+ release. The Ca2+ mobilizing effect of Aβ42 was also observed when applied to permeabilized cells deficient in InsP3 receptors, revealing an additional direct effect of Aβ42 upon the ER, and a mechanism for induction of toxicity by intracellular Aβ42.
Collapse
Affiliation(s)
- Laura E Jensen
- Babraham Institute, Babraham Research Campus Babraham, Cambridge, UK
| | | | | | | | | | | |
Collapse
|
42
|
Wang ZC, Zhao J, Li S. Dysregulation of synaptic and extrasynaptic N-methyl-D-aspartate receptors induced by amyloid-β. Neurosci Bull 2013; 29:752-60. [PMID: 24136243 DOI: 10.1007/s12264-013-1383-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 12/09/2012] [Accepted: 02/25/2013] [Indexed: 02/07/2023] Open
Abstract
The toxicity of amyloid-beta (Aβ) is strongly associated with Alzheimer's disease (AD), which has a high incidence in the elderly worldwide. Recent evidence showed that alteration in the activity of N-methyl-D-aspartate receptors (NMDARs) plays a key role in Aβ-induced neurotoxicity. However, the activation of synaptic and extrasynaptic NMDARs has distinct consequences for plasticity, gene regulation, neuronal death, and Aβ production. This review focuses on the dysregulation of synaptic and extrasynaptic NMDARs induced by Aβ. On one hand, Aβ downregulates the synaptic NMDAR response by promoting NMDAR endocytosis, leading to either neurotoxicity or neuroprotection. On the other hand, Aβ enhances the activation of extrasynaptic NMDARs by decreasing neuronal glutamate uptake and inducing glutamate spillover, subsequently causing neurotoxicity. In addition, selective enhancement of synaptic activity by low doses of NMDA, or reduction of extrasynaptic activity by memantine, a non-competitive NMDAR antagonist, halts Aβ-induced neurotoxicity. Therefore, future neuroprotective drugs for AD should aim at both the enhancement of synaptic activity and the disruption of extrasynaptic NMDAR-dependent death signaling.
Collapse
Affiliation(s)
- Zhi-Cong Wang
- Department of Physiology, Dalian Medical University, Dalian, 116044, China
| | | | | |
Collapse
|
43
|
Calabresi P, Castrioto A, Di Filippo M, Picconi B. New experimental and clinical links between the hippocampus and the dopaminergic system in Parkinson's disease. Lancet Neurol 2013; 12:811-21. [DOI: 10.1016/s1474-4422(13)70118-2] [Citation(s) in RCA: 135] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/19/2023]
|
44
|
Parsons CG, Danysz W, Dekundy A, Pulte I. Memantine and cholinesterase inhibitors: complementary mechanisms in the treatment of Alzheimer's disease. Neurotox Res 2013; 24:358-69. [PMID: 23657927 PMCID: PMC3753463 DOI: 10.1007/s12640-013-9398-z] [Citation(s) in RCA: 218] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/28/2013] [Revised: 04/25/2013] [Accepted: 04/27/2013] [Indexed: 12/11/2022]
Abstract
This review describes the preclinical mechanisms that may underlie the increased therapeutic benefit of combination therapy-with the N-methyl-D-aspartate receptor antagonist, memantine, and an acetylcholinesterase inhibitor (AChEI)-for the treatment of Alzheimer's disease (AD). Memantine, and the AChEIs target two different aspects of AD pathology. Both drug types have shown significant efficacy as monotherapies for the treatment of AD. Furthermore, clinical observations indicate that their complementary mechanisms offer superior benefit as combination therapy. Based on the available literature, the authors have considered the preclinical mechanisms that could underlie such a combined approach. Memantine addresses dysfunction in glutamatergic transmission, while the AChEIs serve to increase pathologically lowered levels of the neurotransmitter acetylcholine. In addition, preclinical studies have shown that memantine has neuroprotective effects, acting to prevent glutamatergic over-stimulation and the resulting neurotoxicity. Interrelations between the glutamatergic and cholinergic pathways in regions of the brain that control learning and memory mean that combination treatment has the potential for a complex influence on disease pathology. Moreover, studies in animal models have shown that the combined use of memantine and the AChEIs can produce greater improvements in measures of memory than either treatment alone. As an effective approach in the clinical setting, combination therapy with memantine and an AChEI has been a welcome advance for the treatment of patients with AD. Preclinical data have shown how these drugs act via two different, but interconnected, pathological pathways, and that their complementary activity may produce greater effects than either drug individually.
Collapse
Affiliation(s)
- Chris G Parsons
- In Vitro Pharmacology, Merz Pharmaceuticals GmbH, Eckenheimer Landstrasse 100, 60318, Frankfurt, Germany.
| | | | | | | |
Collapse
|
45
|
Li S, Jin M, Zhang D, Yang T, Koeglsperger T, Fu H, Selkoe DJ. Environmental novelty activates β2-adrenergic signaling to prevent the impairment of hippocampal LTP by Aβ oligomers. Neuron 2013; 77:929-41. [PMID: 23473322 DOI: 10.1016/j.neuron.2012.12.040] [Citation(s) in RCA: 139] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Accepted: 12/27/2012] [Indexed: 01/11/2023]
Abstract
A central question about human brain aging is whether cognitive enrichment slows the development of Alzheimer changes. Here, we show that prolonged exposure to an enriched environment (EE) facilitated signaling in the hippocampus of wild-type mice that promoted long-term potentiation. A key feature of the EE effect was activation of β2-adrenergic receptors and downstream cAMP/PKA signaling. This EE pathway prevented LTP inhibition by soluble oligomers of amyloid β-protein (Aβ) isolated from AD cortex. Protection by EE occurred in both young and middle-aged wild-type mice. Exposure to novelty afforded greater protection than did aerobic exercise. Mice chronically fed a β-adrenergic agonist without EE were protected from hippocampal impairment by Aβ oligomers. Thus, EE enhances hippocampal synaptic plasticity by activating β-adrenoceptor signaling and mitigating synaptotoxicity of human Aβ oligomers. These mechanistic insights support using prolonged exposure to cognitive novelty and/or oral β-adrenergic agonists to lessen the effects of Aβ accumulation during aging.
Collapse
Affiliation(s)
- Shaomin Li
- Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Esposito Z, Belli L, Toniolo S, Sancesario G, Bianconi C, Martorana A. Amyloid β, glutamate, excitotoxicity in Alzheimer's disease: are we on the right track? CNS Neurosci Ther 2013; 19:549-55. [PMID: 23593992 DOI: 10.1111/cns.12095] [Citation(s) in RCA: 145] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/14/2012] [Revised: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) has a devastating impact on aged people worldwide. Although sophisticated and advanced molecular methods have been developed for its diagnosis since early phases, pharmacological treatment still represents an unresolved topic. The more the disease progresses, the more the uneffectiveness of antidementia drugs emerges. New and encouraging results from experimental works indicate that glutamate pathway may play a substantial role in the pathogenesis since early stages of the disease. Several experimental data together with the clinical use of the uncompetitive N-methyl-d-aspartate (NMDA) antagonist memantine strengthen this idea. Unfortunately, definitive data on the glutamatergic transmission involvement in AD are still incomplete. Moreover, clinical results indicate only temporarily limited effects of memantine. Currently, memantine is indicated for moderate-to-severe cases of AD, an indication that may limit its efficacy and impact on Alzheimer's dementia. The association of memantine with the acetylcholinesterase inhibitor drugs used to treat dementia symptoms appears to be beneficial, in both experimental and clinical studies. Because cholinergic and glutamatergic dysfunction occurs early in AD, the coadministration of appropriate treatment in early stages of the disease might represent a valid option from the beginning of cognitive decline. Moreover, to better evaluate drug efficacy, the association of the recently introduced biomarkers with a clinical AD profile should be considered an aim to pursue.
Collapse
Affiliation(s)
- Zaira Esposito
- Centro per il Decadimento Cognitivo, Sacro Cuore Don Calabria, Negrar-Verona, Italy
| | | | | | | | | | | |
Collapse
|
47
|
Bartlett TE, Wang YT. The intersections of NMDAR-dependent synaptic plasticity and cell survival. Neuropharmacology 2013; 74:59-68. [PMID: 23357336 DOI: 10.1016/j.neuropharm.2013.01.012] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 11/14/2012] [Revised: 01/09/2013] [Accepted: 01/14/2013] [Indexed: 10/27/2022]
Abstract
The discovery of a requirement for N-methyl d-aspartate receptor (NMDAR) activation in long-term potentiation (LTP) set off an explosion of interest in the mechanisms of NMDAR-dependent synaptic plasticity. Meanwhile other research has advanced our understanding of how NMDAR activation regulates neuronal death and survival. Surprisingly, there have been few attempts to correlate these important areas of research. Here we review current knowledge of the various mechanisms of NMDAR-dependent synaptic plasticity that are shared with neuronal survival and death, while drawing comparisons with the proneurotrophin/neurotrophin receptor and intracellular signaling systems. Our conclusion is that NMDAR-dependent LTP and long-term depression (LTD) share many common mechanisms with cell survival and cell death, respectively. The intersections of plasticity and cell survival may represent novel avenues for neuroprotection. This article is part of the Special Issue entitled 'Glutamate Receptor-Dependent Synaptic Plasticity'.
Collapse
Affiliation(s)
- Thomas E Bartlett
- Brain Research Centre, Room F270, University of British Columbia, 2211 Wesbrook Mall, Vancouver, BC V6T 2B5, Canada
| | | |
Collapse
|
48
|
Danysz W, Parsons CG. Alzheimer's disease, β-amyloid, glutamate, NMDA receptors and memantine--searching for the connections. Br J Pharmacol 2013; 167:324-52. [PMID: 22646481 DOI: 10.1111/j.1476-5381.2012.02057.x] [Citation(s) in RCA: 358] [Impact Index Per Article: 29.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022] Open
Abstract
β-amyloid (Aβ) is widely accepted to be one of the major pathomechanisms underlying Alzheimer's disease (AD), although there is presently lively debate regarding the relative roles of particular species/forms of this peptide. Most recent evidence indicates that soluble oligomers rather than plaques are the major cause of synaptic dysfunction and ultimately neurodegeneration. Soluble oligomeric Aβ has been shown to interact with several proteins, for example glutamatergic receptors of the NMDA type and proteins responsible for maintaining glutamate homeostasis such as uptake and release. As NMDA receptors are critically involved in neuronal plasticity including learning and memory, we felt that it would be valuable to provide an up to date review of the evidence connecting Aβ to these receptors and related neuronal plasticity. Strong support for the clinical relevance of such interactions is provided by the NMDA receptor antagonist memantine. This substance is the only NMDA receptor antagonist used clinically in the treatment of AD and therefore offers an excellent tool to facilitate translational extrapolations from in vitro studies through in vivo animal experiments to its ultimate clinical utility.
Collapse
Affiliation(s)
- Wojciech Danysz
- Merz Pharmaceuticals GmbH, Eckenheimer Landstraße, Frankfurt am Main, Germany
| | | |
Collapse
|
49
|
Revett TJ, Baker GB, Jhamandas J, Kar S. Glutamate system, amyloid ß peptides and tau protein: functional interrelationships and relevance to Alzheimer disease pathology. J Psychiatry Neurosci 2013; 38:6-23. [PMID: 22894822 PMCID: PMC3529221 DOI: 10.1503/jpn.110190] [Citation(s) in RCA: 211] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022] Open
Abstract
Alzheimer disease is the most prevalent form of dementia globally and is characterized premortem by a gradual memory loss and deterioration of higher cognitive functions and postmortem by neuritic plaques containing amyloid ß peptide and neurofibrillary tangles containing phospho-tau protein. Glutamate is the most abundant neurotransmitter in the brain and is essential to memory formation through processes such as long-term potentiation and so might be pivotal to Alzheimer disease progression. This review discusses how the glutamatergic system is impaired in Alzheimer disease and how interactions of amyloid ß and glutamate influence synaptic function, tau phosphorylation and neurodegeneration. Interestingly, glutamate not only influences amyloid ß production, but also amyloid ß can alter the levels of glutamate at the synapse, indicating that small changes in the concentrations of both molecules could influence Alzheimer disease progression. Finally, we describe how the glutamate receptor antagonist, memantine, has been used in the treatment of individuals with Alzheimer disease and discuss its effectiveness.
Collapse
Affiliation(s)
| | | | | | - Satyabrata Kar
- Correspondence to: S. Kar, Centre for Prions and Protein Folding Diseases, Departments of Medicine (Neurology) and Psychiatry, University of Alberta, Edmonton AB T6G 2M8;
| |
Collapse
|
50
|
Yang H, Wu D, Zhang X, Wang X, Peng Y, Hu Z. Telencephalin protects PAJU cells from amyloid beta protein-induced apoptosis by activating the ezrin/radixin/moesin protein family/phosphatidylinositol-3-kinase/protein kinase B pathway. Neural Regen Res 2012; 7:2189-98. [PMID: 25538739 PMCID: PMC4268718 DOI: 10.3969/j.issn.1673-5374.2012.028.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 03/18/2012] [Accepted: 06/13/2012] [Indexed: 11/20/2022] Open
Abstract
Telencephalin is a neural glycoprotein that reduces apoptosis induced by amyloid beta protein in the human neural tumor cell line PAJU. In this study, we examined the role of the ezrin/radixin/moesin protein family/phosphatidylinositol-3-kinase/protein kinase B pathway in this process. Western blot analysis demonstrated that telencephalin, phosphorylated ezrin/radixin/moesin and phosphatidylinositol-3-kinase/protein kinase B were not expressed in PAJU cells transfected with empty plasmid, while they were expressed in PAJU cells transfected with a telencephalin expression plasmid. After treatment with 1.0 nM amyloid beta protein 42, expression of telencephalin and phosphorylated phosphatidylinositol-3-kinase/protein kinase B in the transfected cells gradually diminished, while levels of phosphorylated ezrin/radixin/moesin increased. In addition, the high levels of telencephalin, phosphorylated ezrin/radixin/moesin and phosphatidylinositol-3-kinase/protein kinase B expression in PAJU cells transfected with a telencephalin expression plasmid could be suppressed by the phosphatidylinositol-3-kinase inhibitor LY294002. These findings indicate that telencephalin activates the ezrin/radixin/moesin family/phosphatidylinositol-3-kinase/protein kinase B pathway and protects PAJU cells from amyloid beta protein-induced apoptosis.
Collapse
Affiliation(s)
- Heping Yang
- Department of Neurology, Shangrao No. 5 People's Hospital, the Second Affiliated Hospital of Nanchang University, Shangrao 334000, Jiangxi Province, China
- Department of Neurology, Ruikang Hospital Affiliated to Guangxi Traditional Chinese Medical University, Nanning 530011, Guangxi Zhuang Autonomous Region, China
- Department of Neurology, Xiangya Second Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Dapeng Wu
- Department of Neurology, Xiangya Second Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiaojie Zhang
- Department of Psychiatry, Xiangya Second Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Xiang Wang
- Department of Neurology, Xiangya Second Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Yi Peng
- Department of Neurology, Xiangya Second Hospital, Central South University, Changsha 410011, Hunan Province, China
| | - Zhiping Hu
- Department of Neurology, Xiangya Second Hospital, Central South University, Changsha 410011, Hunan Province, China
| |
Collapse
|