1
|
Sun C, Gao X, Sha S, Wang S, Shan Y, Li L, Xing C, Guan H, Du H. Berberine alleviates Alzheimer's disease by activating autophagy and inhibiting ferroptosis through the JNK-p38MAPK signaling pathway. Int Immunopharmacol 2025; 155:114550. [PMID: 40215776 DOI: 10.1016/j.intimp.2025.114550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 03/19/2025] [Accepted: 03/22/2025] [Indexed: 04/29/2025]
Abstract
INTRODUCTION Alzheimer's disease (AD) is a neurodegenerative disease characterized by amyloid beta (Aβ) deposition, phosphorylated Tau protein aggregation, inflammation, and neuronal damage. Neuronal autophagy plays an important role in ameliorating central nervous system diseases such as AD. As an emerging form of iron-dependent cell death, ferroptosis has attracted great attention in the field of neurodegenerative diseases. Berberine (BBR), a natural alkaloid, has demonstrated excellent in inflammation reduction, inhibition of Aβ production, and neuroprotection, making it a potential candidate for AD treatment. However, the mechanisms of autophagy and ferroptosis in BBR treatment of AD have not been elucidated. OBJECTIVES This study aimed to investigate the potential of BBR in alleviating AD and evaluate its molecular mechanism through a combination of network pharmacology and biological experiments. METHODS We assessed alterations in Aβ plaques, neurons, neuroinflammation, and autophagy-related markers in the mice brain using immunofluorescence staining. Network pharmacology and molecular docking were used to analyze the potential targets and signaling pathways of BBR in the treatment of AD. Morris Water Maze (MWM) and new object recognition (NOR) experiments were used to test the spatial memory ability of mice. In addition, we validated the relationship between JNK-P38MAPK, autophagy, ferroptosis, and BBR treatment in 5xFAD mice and A β-induced SH-SY5Y cell models. RESULTS The results of immunofluorescence staining showed that BBR effectively mitigated Aβ plaque deposition, ameliorated neuronal damage and neuroinflammation. The autophagy-related markers Beclin1 and LC3B were upregulated and P62 was downregulated after BBR treatment. The expression levels of ROS and lipid peroxide MDA decreased significantly after BBR treatment. qPCR results showed that the expression levels of ferroptosis-related genes TFR1, ASCL4, DMT1, and IREB2 were decreased, while the expression levels of FTH1 and SLC7A11 increased after BBR treatment. Behavioral experiments showed that BBR treatment enhanced spatial memory impairment in 5xFAD mice. Network pharmacological and in vitro analyses demonstrated that BBR activated autophagy and inhibited ferroptosis by inhibiting the JNK-P38MAPK signaling pathway. Following treatment with an autophagy inhibitor on SH-SY5Y cells, autophagy was markedly suppressed, and ferroptosis was induced. CONCLUSION In summary, we found that BBR alleviates AD by inhibiting the JNK-P38MAPK pathway to enhance autophagy and inhibit ferroptosis, further reducing Aβ plaque deposition, inhibiting inflammatory response, and improving neuronal damage.
Collapse
Affiliation(s)
- Chunbin Sun
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Xiaoyu Gao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Shuang Sha
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Si Wang
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming 650500, China
| | - Yubang Shan
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Luping Li
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Cencan Xing
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083,China.
| | - Hongyan Guan
- China Testing & Certification International Group Co., Ltd., Beijing 100024, China.
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083,China.
| |
Collapse
|
2
|
Chen JY, Li CY, Mong MC, Yin MC. Preventive effects of coixol, an active compound of adlay seed, in NGF-differentiated PC12 cells against beta-amyloid 25-35-induced neurotoxicity. ASIAN BIOMED 2024; 18:224-235. [PMID: 39483714 PMCID: PMC11524674 DOI: 10.2478/abm-2024-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Background The health benefits of coixol, an active compound of adlay seed, have attracted certain attention. Adlay seed is often adopted in traditional Chinese medicine for the treatment of various inflammatory disorders. Thus, it is hypothesized that coixol could protect neuronal cells. Objectives The preventive effects of coixol against Abeta25-35-induced damage in nerve growth factor-differentiated PC12 cells were explored. Methods Differentiated PC12 cells were treated with coixol at 0.125 μM, 0.25 μM, 0.5 μM, 1 μM, and 2 μM for 48 h. Then, cells were further exposed to Abeta25-35 at 20 μM for 24 h. Results Coixol treatments at 0.25-2 μM exhibited antiapoptotic effect via increasing Bcl-2 mRNA expression, mitochondrial membrane potential, and Na+-K+ ATPase activity as well as decreasing Bax mRNA expression, caspase-3 activity, and intracellular Ca2+ release. In addition, coixol treatments at 0.25-2 μM alleviated oxidative and inflammatory responses via lowering reactive oxygen species level, increasing glutathione content, promoting the activity of glutathione peroxidase, glutathione reductase, and catalase, decreasing the generation of tumor necrosis factor-α, interleukin (IL)-1β, IL-6, and prostaglandin E2. Furthermore, coixol treatments at 0.25-2 μM diminished intracellular Ca2+ release, and restricted nuclear factor kappa B-binding activity and phosphorylation of p65 and p38. Coixol treatments at 0.5-2 μM increased protein generation of nuclear factor E2-related factor 2, and limited protein production of inducible nitric oxide synthase and receptor of advanced glycation end product. Conclusion Our novel findings suggested that coixol was a compelling agent against beta-amyloid peptide-induced neurotoxicity.
Collapse
Affiliation(s)
- Jan-Yow Chen
- Department of Internal Medicine, China Medical University Hospital, Taichung404328, Taiwan
| | - Chien-Yu Li
- Department of Neurosurgery, Asia University Hospital, Taichung413305, Taiwan
| | - Mei-Chin Mong
- Department of Food Nutrition and Health Biotechnology, Asia University, Taichung413305, Taiwan
| | - Mei-Chin Yin
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung404328, Taiwan
- Office of Research and Development, Asia University, Taichung413305, Taiwan
| |
Collapse
|
3
|
Tang X, Deng P, Jiang Y, Zhang L, He Y, Yang H. An Overview of Recent Advances in the Neuroprotective Potentials of Fisetin against Diverse Insults in Neurological Diseases and the Underlying Signaling Pathways. Biomedicines 2023; 11:2878. [PMID: 38001882 PMCID: PMC10669030 DOI: 10.3390/biomedicines11112878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/18/2023] [Accepted: 10/22/2023] [Indexed: 11/26/2023] Open
Abstract
The nervous system plays a leading role in the regulation of physiological functions and activities in the body. However, a variety of diseases related to the nervous system have a serious impact on human health. It is increasingly clear that neurological diseases are multifactorial pathological processes involving multiple cellular systems, and the onset of these diseases usually involves a diverse array of molecular mechanisms. Unfortunately, no effective therapy exists to slow down the progression or prevent the development of diseases only through the regulation of a single factor. To this end, it is pivotal to seek an ideal therapeutic approach for challenging the complicated pathological process to achieve effective treatment. In recent years, fisetin, a kind of flavonoid widely existing in fruits, vegetables and other plants, has shown numerous interesting biological activities with clinical potentials including anti-inflammatory, antioxidant and neurotrophic effects. In addition, fisetin has been reported to have diverse pharmacological properties and neuroprotective potentials against various neurological diseases. The neuroprotective effects were ascribed to its unique biological properties and multiple clinical pharmacological activities associated with the treatment of different neurological disorders. In this review, we summarize recent research progress regarding the neuroprotective potential of fisetin and the underlying signaling pathways of the treatment of several neurological diseases.
Collapse
Affiliation(s)
- Xiangwen Tang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an 710054, China; (X.T.); (L.Z.)
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China; (P.D.); (Y.J.)
| | - Peng Deng
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China; (P.D.); (Y.J.)
| | - Yizhen Jiang
- Basic Medical School Academy, Shaanxi University of Traditional Chinese Medicine, Xianyang 712046, China; (P.D.); (Y.J.)
| | - Lingling Zhang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an 710054, China; (X.T.); (L.Z.)
| | - Yuqing He
- School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, China;
| | - Hao Yang
- Translational Medicine Center, Hong Hui Hospital, Xi’an Jiaotong University, Xi’an 710054, China; (X.T.); (L.Z.)
| |
Collapse
|
4
|
Sharma H, Chang KA, Hulme J, An SSA. Mammalian Models in Alzheimer's Research: An Update. Cells 2023; 12:2459. [PMID: 37887303 PMCID: PMC10605533 DOI: 10.3390/cells12202459] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/28/2023] Open
Abstract
A form of dementia distinct from healthy cognitive aging, Alzheimer's disease (AD) is a complex multi-stage disease that currently afflicts over 50 million people worldwide. Unfortunately, previous therapeutic strategies developed from murine models emulating different aspects of AD pathogenesis were limited. Consequently, researchers are now developing models that express several aspects of pathogenesis that better reflect the clinical situation in humans. As such, this review seeks to provide insight regarding current applications of mammalian models in AD research by addressing recent developments and characterizations of prominent transgenic models and their contributions to pathogenesis as well as discuss the advantages, limitations, and application of emerging models that better capture genetic heterogeneity and mixed pathologies observed in the clinical situation.
Collapse
Affiliation(s)
- Himadri Sharma
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| | - Keun-A Chang
- Neuroscience Research Institute, Gachon University, Incheon 21565, Republic of Korea
| | - John Hulme
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| | - Seong Soo A. An
- Department of Bionano Technology, Gachon Bionano Research Institute, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si 461-701, Gyeonggi-do, Republic of Korea
| |
Collapse
|
5
|
Fila M, Pawlowska E, Szczepanska J, Blasiak J. Autophagy may protect the brain against prolonged consequences of headache attacks: A narrative/hypothesis review. Headache 2023; 63:1154-1166. [PMID: 37638395 DOI: 10.1111/head.14625] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 06/25/2023] [Accepted: 07/14/2023] [Indexed: 08/29/2023]
Abstract
OBJECTIVE To assess the potential of autophagy in migraine pathogenesis. BACKGROUND The interplay between neurons and microglial cells is important in migraine pathogenesis. Migraine-related effects, such as cortical spreading depolarization and release of calcitonin gene-related peptide, may initiate adenosine triphosphate (ATP)-mediating pro-nociceptive signaling in the meninges causing headaches. Such signaling may be induced by the interaction of ATP with purinergic receptor P2X 7 (P2X7R) on microglial cells leading to a Ca2+ -mediated pH increase in lysosomes and release of autolysosome-like vehicles from microglial cells indicating autophagy impairment. METHODS A search in PubMed was conducted with the use of the terms "migraine," "autophagy," "microglia," and "degradation" in different combinations. RESULTS Impaired autophagy in microglia may activate secretory autophagy and release of specific proteins, including brain-derived neurotrophic factor (BDNF), which can be also released through the pores induced by P2X7R activation in microglial cells. BDNF may be likewise released from microglial cells upon ATP- and Ca2+ -mediated activation of another purinergic receptor, P2X4R. BDNF released from microglia might induce autophagy in neurons to clear cellular debris produced by oxidative stress, which is induced in the brain as the response to migraine-related energy deficit. Therefore, migraine-related signaling may impair degradative autophagy, stimulate secretory autophagy in microglia, and degradative autophagy in neurons. These effects are mediated by purinergic receptors P2X4R and P2X7R, BDNF, ATP, and Ca2+ . CONCLUSION Different effects of migraine-related events on degradative autophagy in microglia and neurons may prevent prolonged changes in the brain related to headache attacks.
Collapse
Affiliation(s)
- Michal Fila
- Department of Developmental Neurology and Epileptology, Polish Mother's Memorial Hospital Research Institute, Lodz, Poland
| | - Elzbieta Pawlowska
- Department of Pediatric Dentistry, Medical University of Lodz, Lodz, Poland
| | - Joanna Szczepanska
- Department of Pediatric Dentistry, Medical University of Lodz, Lodz, Poland
| | - Janusz Blasiak
- Department of Molecular Genetics, University of Lodz, Lodz, Poland
| |
Collapse
|
6
|
Liu S, Chen L, Li J, Sun Y, Xu Y, Li Z, Zhu Z, Li X. Asiaticoside Mitigates Alzheimer's Disease Pathology by Attenuating Inflammation and Enhancing Synaptic Function. Int J Mol Sci 2023; 24:11976. [PMID: 37569347 PMCID: PMC10418370 DOI: 10.3390/ijms241511976] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 07/21/2023] [Accepted: 07/24/2023] [Indexed: 08/13/2023] Open
Abstract
Alzheimer's disease (AD) is a prevalent neurodegenerative disorder, hallmarked by the accumulation of amyloid-β (Aβ) plaques and neurofibrillary tangles. Due to the uncertainty of the pathogenesis of AD, strategies aimed at suppressing neuroinflammation and fostering synaptic repair are eagerly sought. Asiaticoside (AS), a natural triterpenoid derivative derived from Centella asiatica, is known for its anti-inflammatory, antioxidant, and wound-healing properties; however, its neuroprotective function in AD remains unclear. Our current study reveals that AS, when administered (40 mg/kg) in vivo, can mitigate cognitive dysfunction and attenuate neuroinflammation by inhibiting the activation of microglia and proinflammatory factors in Aβ1-42-induced AD mice. Further mechanistic investigation suggests that AS may ameliorate cognitive impairment by inhibiting the activation of the p38 MAPK pathway and promoting synaptic repair. Our findings propose that AS could be a promising candidate for AD treatment, offering neuroinflammation inhibition and enhancement of synaptic function.
Collapse
Affiliation(s)
- Sai Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Long Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Jinran Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yuan Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| | - Yue Xu
- Division of Pharmaceutics and Pharmacology, College of Pharmacy, The Ohio State University, Columbus, OH 43210, USA
| | - Zhaoxing Li
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
| | - Zheying Zhu
- School of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, UK
| | - Xinuo Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, China
- Jiangsu Provincial Key Laboratory of Drug Metabolism and Pharmacokinetics, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
7
|
Basheer N, Smolek T, Hassan I, Liu F, Iqbal K, Zilka N, Novak P. Does modulation of tau hyperphosphorylation represent a reasonable therapeutic strategy for Alzheimer's disease? From preclinical studies to the clinical trials. Mol Psychiatry 2023; 28:2197-2214. [PMID: 37264120 PMCID: PMC10611587 DOI: 10.1038/s41380-023-02113-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 04/28/2023] [Accepted: 05/05/2023] [Indexed: 06/03/2023]
Abstract
Protein kinases (PKs) have emerged as one of the most intensively investigated drug targets in current pharmacological research, with indications ranging from oncology to neurodegeneration. Tau protein hyperphosphorylation was the first pathological post-translational modification of tau protein described in Alzheimer's disease (AD), highlighting the role of PKs in neurodegeneration. The therapeutic potential of protein kinase inhibitors (PKIs)) and protein phosphatase 2 A (PP2A) activators in AD has recently been explored in several preclinical and clinical studies with variable outcomes. Where a number of preclinical studies demonstrate a visible reduction in the levels of phospho-tau in transgenic tauopathy models, no reduction in neurofibrillary lesions is observed. Amongst the few PKIs and PP2A activators that progressed to clinical trials, most failed on the efficacy front, with only a few still unconfirmed and potential positive trends. This suggests that robust preclinical and clinical data is needed to unequivocally evaluate their efficacy. To this end, we take a systematic look at the results of preclinical and clinical studies of PKIs and PP2A activators, and the evidence they provide regarding the utility of this approach to evaluate the potential of targeting tau hyperphosphorylation as a disease modifying therapy.
Collapse
Affiliation(s)
- Neha Basheer
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia
| | - Tomáš Smolek
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia
| | - Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Fei Liu
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY, 10314, USA
| | - Khalid Iqbal
- Department of Neurochemistry, Inge Grundke-Iqbal Research Floor, New York State Institute for Basic Research in Developmental Disabilities, 1050 Forest Hill Road, Staten Island, NY, 10314, USA
| | - Norbert Zilka
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia.
- AXON Neuroscience R&D Services SE, Bratislava, 811 02, Slovakia.
| | - Petr Novak
- Institute of Neuroimmunology, Slovak Academy of Sciences, Bratislava, 845 10, Slovakia.
- AXON Neuroscience CRM Services SE, Bratislava, 811 02, Slovakia.
| |
Collapse
|
8
|
Petrushanko IY, Mitkevich VA, Makarov AA. Effect of β-amyloid on blood-brain barrier properties and function. Biophys Rev 2023; 15:183-197. [PMID: 37124923 PMCID: PMC10133432 DOI: 10.1007/s12551-023-01052-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Accepted: 03/13/2023] [Indexed: 05/02/2023] Open
Abstract
The deposition of beta-amyloid (Aβ) aggregates in the brain, accompanied by impaired cognitive function, is a characteristic feature of Alzheimer's disease (AD). An important role in this process is played by vascular disorders, in particular, a disturbance of the blood-brain barrier (BBB). The BBB controls the entry of Aβ from plasma to the brain via the receptor for advanced glycation end products (RAGE) and the removal of brain-derived Aβ via the low-density lipoprotein receptor-related protein (LRP1). The balance between the input of Aβ to the brain from the periphery and its output is disturbed during AD. Aβ changes the redox-status of BBB cells, which in turn changes the functioning of mitochondria and disrupts the barrier function of endothelial cells by affecting tight junction proteins. Aβ oligomers have the greatest toxic effect on BBB cells, and oligomers are most rapidly transferred by transcytosis from the brain side of the BBB to the blood side. Both the cytotoxic effect of Aβ and the impairment of barrier function are partly due to the interaction of Aβ monomers and oligomers with membrane-bound RAGE. AD therapies based on the disruption of this interaction or the creation of decoys for Aβ are being developed. The question of the transfer of various Aβ isoforms through the BBB is important, since it can influence the development of AD. It is shown that the rate of input of Aβ40 and Aβ42 from the blood into the brain is different. The actual question of the transfer of pathogenic Aβ isoforms with post-translational modifications or mutations through the BBB still remains open.
Collapse
Affiliation(s)
- Irina Yu. Petrushanko
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Vladimir A. Mitkevich
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander A. Makarov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
9
|
Jin R, Ning X, Liu X, Zhao Y, Ye G. Porphyromonas gingivalis-induced periodontitis could contribute to cognitive impairment in Sprague–Dawley rats via the P38 MAPK signaling pathway. Front Cell Neurosci 2023; 17:1141339. [PMID: 37056710 PMCID: PMC10086325 DOI: 10.3389/fncel.2023.1141339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 03/13/2023] [Indexed: 03/30/2023] Open
Abstract
BackgroundPeriodontitis is one of the most common oral diseases and has been shown to be a risk factor for systemic diseases. Our aim was to investigate the relationship between periodontitis and cognitive impairment and to explore the role of the P38 MAPK signaling pathway in this process.MethodsWe established a periodontitis model by ligating the first molars of SD rats with silk thread and injecting Porphyromonas gingivalis (P. gingivalis) or P. gingivalis plus the P38 MAPK inhibitor SB203580 at the same time for ten weeks. We assessed alveolar bone resorption and spatial learning and memory using microcomputed tomography and the Morris water maze test, respectively. We used transcriptome sequencing to explore the genetic differences between the groups. The gingival tissue, peripheral blood and hippocampal tissue were assessed for the cytokines TNF-α, IL-1β, IL-6, IL-8 and C reactive protein (CRP) with enzyme-linked immunosorbent assay (ELISA) and reverse transcription polymerase chain reaction (RT–PCR). We observed the presence of P. gingivalis in the hippocampus of rats by paraffin-fluorescence in situ hybridization (FISH). We determined the activation of microglia by immunofluorescence. Finally, Western blot analysis was employed to determine the expression of amyloid precursor protein (APP), β-site APP-cleaving enzyme 1 (BACE1) and P38MAPK pathway activation.ResultsWe demonstrated that silk ligature-induced periodontitis plus injection of P. gingivalis into subgingival tissue could lead to memory and cognitive impairment. Transcriptome sequencing results suggested that there were neurodegenerative diseases in the P. gingivalis group, and the MWM test showed that periodontitis reduced the spatial learning and memory ability of mild cognitive impairment (MCI) model rats. We found high levels of inflammatory factors (TNF-α, IL-1β, IL-6, and IL-8) and CRP in the gingiva, peripheral blood and hippocampus, and the expression of APP and BACE1 was upregulated, as was the P38 MAPK pathway activation. Activated microglia and the presence of P. gingivalis were also found in the hippocampus. P38 MAPK inhibitors mitigated all of these changes.ConclusionOur findings strongly suggest that topical application of P. gingivalis increases the inflammatory burden in the peripheral and central nervous systems (CNS) and that neuroinflammation induced by activation of P38 MAPK leads to impaired learning and memory in SD rats. It can also modulate APP processing. Therefore, P38 MAPK may serve as a linking pathway between periodontitis and cognitive impairment.
Collapse
Affiliation(s)
- Ru Jin
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital, Chongqing Medical University, Chongqing, China
| | - Xiaoqiao Ning
- The First People’s Hospital of Wanzhou, Chongqing, China
| | - Xiang Liu
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital, Chongqing Medical University, Chongqing, China
| | - Yueyang Zhao
- Department of Anatomy, Chongqing Medical University, Chongqing, China
| | - Guo Ye
- Chongqing Key Laboratory of Oral Disease and Biomedical Sciences and Chongqing Municipal Key Laboratory of Oral Biomedical Engineering of Higher Education, Stomatological Hospital, Chongqing Medical University, Chongqing, China
- *Correspondence: Guo Ye,
| |
Collapse
|
10
|
MicroRNAs and MAPKs: Evidence of These Molecular Interactions in Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24054736. [PMID: 36902178 PMCID: PMC10003111 DOI: 10.3390/ijms24054736] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 02/24/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder known to be the leading cause of dementia worldwide. Many microRNAs (miRNAs) were found deregulated in the brain or blood of AD patients, suggesting a possible key role in different stages of neurodegeneration. In particular, mitogen-activated protein kinases (MAPK) signaling can be impaired by miRNA dysregulation during AD. Indeed, the aberrant MAPK pathway may facilitate the development of amyloid-beta (Aβ) and Tau pathology, oxidative stress, neuroinflammation, and brain cell death. The aim of this review was to describe the molecular interactions between miRNAs and MAPKs during AD pathogenesis by selecting evidence from experimental AD models. Publications ranging from 2010 to 2023 were considered, based on PubMed and Web of Science databases. According to obtained data, several miRNA deregulations may regulate MAPK signaling in different stages of AD and conversely. Moreover, overexpressing or silencing miRNAs involved in MAPK regulation was seen to improve cognitive deficits in AD animal models. In particular, miR-132 is of particular interest due to its neuroprotective functions by inhibiting Aβ and Tau depositions, as well as oxidative stress, through ERK/MAPK1 signaling modulation. However, further investigations are required to confirm and implement these promising results.
Collapse
|
11
|
Wang D, Chen F, Han Z, Yin Z, Ge X, Lei P. Relationship Between Amyloid-β Deposition and Blood-Brain Barrier Dysfunction in Alzheimer's Disease. Front Cell Neurosci 2021; 15:695479. [PMID: 34349624 PMCID: PMC8326917 DOI: 10.3389/fncel.2021.695479] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 06/23/2021] [Indexed: 12/14/2022] Open
Abstract
Amyloid-β (Aβ) is the predominant pathologic protein in Alzheimer's disease (AD). The production and deposition of Aβ are important factors affecting AD progression and prognosis. The deposition of neurotoxic Aβ contributes to damage of the blood-brain barrier. However, the BBB is also crucial in maintaining the normal metabolism of Aβ, and dysfunction of the BBB aggravates Aβ deposition. This review characterizes Aβ deposition and BBB damage in AD, summarizes their interactions, and details their respective mechanisms.
Collapse
Affiliation(s)
- Dong Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | | | - Zhaoli Han
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Zhenyu Yin
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| | - Xintong Ge
- Tianjin Neurological Institute, Tianjin, China
- Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Geriatrics Institute, Tianjin, China
| |
Collapse
|
12
|
Wu L, Zheng Q, Guo YY, Zhang KN, Luo J, Xiao S, Li WJ, Yang M. Effect of Zhenxin Xingshui Yizhi Fang on Aβ 25-35 induced expression of related transporters in HBMEC cell model. JOURNAL OF ETHNOPHARMACOLOGY 2020; 260:112783. [PMID: 32240783 DOI: 10.1016/j.jep.2020.112783] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 06/11/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Aβ (β-amyloid) deposition and abnormal transport were suggested to be risk factors for Alzheimer's disease (AD). Zhenxin Xingshui Yizhi Fang (XSF), an ancient prescription in traditional Chinese medicine, was first recorded in Qianjin Yifang for treating palpitation, hypnosia, amnesia. It is reported that XSF could improve mice learning memory ability, reduce the deposition of senile plaques in hippocampus of rat brain. In this study, the neuroprotective effect of XSF against Aβ25-35-induced apoptosis in cultured human brain microvascular endothelial cells (HBMEC) and its potential mechanism were investigated. MATERIALS AND METHODS HBMEC cells were treated with Aβ25-35 to established neurotoxic cell model. After that, the cells were treated with 125, 250, 500 μg/mL XSF to observe the protective effect. The viability of HBMEC cells were evaluated by MTT assay, the Aβ25-35-induced apoptosis was characterized by Hoechst-33258 and the activity of cysteinyl aspartate specific proteinase-3. The expression level of Aβ1-42 in cells induced by Aβ25-35 was measured by human Aβ1-42 kit. Protein and mRNA expression levels of advanced glycation end products (RAGE), low density lipoprotein receptor-related protein 1 (LRP1), glucose transporter 1 and 3 (GLUT1 and GLUT3) were assayed by capillary electrophoresis immunoassay and quantitative real-time polymerase chain reaction analyses. RESULTS In Aβ25-35 induced neurotoxic cells, the percentage of apoptotic cells, the concentration of Aβ1-42 and CASPASE-3 activity, protein and mRNA expression levels of RAGE increased significantly, but that of LRP1, GLUT1 and GLUT3 significantly decreased. XSF could inhibit the apoptotic of cells, reduced the concentration of Aβ1-42 and CASPASE-3 expression, downregulate RAGE and upregulate LRP1, GLUT1 and GLUT3 expression. CONCLUSION The results suggest that XSF can reduce the cytotoxicity of HBMEC induced by Aβ25-35, inhibit apoptosis, and regulate the transport of Aβ on BBB and energy metabolism disorder in HBMEC.
Collapse
Affiliation(s)
- Ling Wu
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Qin Zheng
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China.
| | - Yuan-Yuan Guo
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Ke-Nan Zhang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Jun Luo
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Shuai Xiao
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Wen-Jing Li
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| | - Ming Yang
- Key Laboratory of Modern Preparation of Traditional Chinese Medicine, Ministry of Education, State Key Lab of Innovation Drug and Efficient Energy-Saving Pharmaceutical Equipment, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi, 330004, China
| |
Collapse
|
13
|
Falcicchia C, Tozzi F, Arancio O, Watterson DM, Origlia N. Involvement of p38 MAPK in Synaptic Function and Dysfunction. Int J Mol Sci 2020; 21:ijms21165624. [PMID: 32781522 PMCID: PMC7460549 DOI: 10.3390/ijms21165624] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Many studies have revealed a central role of p38 MAPK in neuronal plasticity and the regulation of long-term changes in synaptic efficacy, such as long-term potentiation (LTP) and long-term depression (LTD). However, p38 MAPK is classically known as a responsive element to stress stimuli, including neuroinflammation. Specific to the pathophysiology of Alzheimer’s disease (AD), several studies have shown that the p38 MAPK cascade is activated either in response to the Aβ peptide or in the presence of tauopathies. Here, we describe the role of p38 MAPK in the regulation of synaptic plasticity and its implication in an animal model of neurodegeneration. In particular, recent evidence suggests the p38 MAPK α isoform as a potential neurotherapeutic target, and specific inhibitors have been developed and have proven to be effective in ameliorating synaptic and memory deficits in AD mouse models.
Collapse
Affiliation(s)
- Chiara Falcicchia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
| | - Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy;
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA;
| | | | - Nicola Origlia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-3153193
| |
Collapse
|
14
|
Accorroni A, Rutigliano G, Sabatini M, Frascarelli S, Borsò M, Novelli E, Bandini L, Ghelardoni S, Saba A, Zucchi R, Origlia N. Exogenous 3-Iodothyronamine Rescues the Entorhinal Cortex from β-Amyloid Toxicity. Thyroid 2020; 30:147-160. [PMID: 31709926 DOI: 10.1089/thy.2019.0255] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Background: A novel form of thyroid hormone (TH) signaling is represented by 3-iodothyronamine (T1AM), an endogenous TH derivative that interacts with specific molecular targets, including trace amine-associated receptor 1 (TAAR1), and induces pro-learning and anti-amnestic effects in mice. Dysregulation of TH signaling has long been hypothesized to play a role in Alzheimer's disease (AD). In the present investigation, we explored the neuroprotective role of T1AM in beta amyloid (Aβ)-induced synaptic and behavioral impairment, focusing on the entorhinal cortex (EC), an area that is affected early by AD pathology. Methods: Field potentials were evoked in EC layer II, and long-term potentiation (LTP) was elicited by high frequency stimulation (HFS). T1AM (5 μM) and/or Aβ(1-42) (200 nM), were administered for 10 minutes, starting 5 minutes before HFS. Selective TAAR1 agonist RO5166017 (250 nM) and TAAR1 antagonist EPPTB (5 nM) were also used. The electrophysiological experiments were repeated in EC-slices taken from a mouse model of AD (mutant human amyloid precursor protein [mhAPP], J20 line). We also assessed the in vivo effects of T1AM on EC-dependent associative memory deficits, which were detected in mhAPP mice by behavioral evaluations based on the novel-object recognition paradigm. TAAR1 expression was determined by Western blot, whereas T1AM and its metabolite 3-iodothyroacetic acid (TA1) were assayed by high-performance liquid chromatography coupled to mass spectrometry. Results: We demonstrate the presence of endogenous T1AM and TAAR1 in the EC of wild-type and mhAPP mice. Exposure to Aβ(1-42) inhibited LTP, and T1AM perfusion (at a concentration of 5 μM, leading to an actual concentration in the perfusion buffer ranging from 44 to 298 nM) restored it, whereas equimolar amounts of 3,5,3'-triiodo-L-thyronine (T3) and TA1 were ineffective. The response to T1AM was abolished by the TAAR1 antagonist EPPTB, whereas it was mimicked by the TAAR1 agonist RO5166017. In the EC of APPJ20 mice, LTP could not be elicited, but it was rescued by T1AM. The intra-cerebro-ventricular administration of T1AM (0.89 μg/kg) also restored recognition memory that was impaired in mhAPP mice. Conclusions: Our results suggest that T1AM and TAAR1 are part of an endogenous system that can be modulated to prevent synaptic and behavioral deficits associated with Aβ-related toxicity.
Collapse
Affiliation(s)
- Alice Accorroni
- Scuola Superiore di Studi Universitari e di Perfezionamento Sant'Anna, Pisa, Italy
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| | - Grazia Rutigliano
- Scuola Superiore di Studi Universitari e di Perfezionamento Sant'Anna, Pisa, Italy
| | | | | | - Marco Borsò
- Department of Pathology, University of Pisa, Pisa, Italy
| | - Elena Novelli
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| | | | | | | | | | - Nicola Origlia
- Institute of Neuroscience of the Italian National Research Council (CNR), Pisa, Italy
| |
Collapse
|
15
|
Liu PP, Xie Y, Meng XY, Kang JS. History and progress of hypotheses and clinical trials for Alzheimer's disease. Signal Transduct Target Ther 2019; 4:29. [PMID: 31637009 PMCID: PMC6799833 DOI: 10.1038/s41392-019-0063-8] [Citation(s) in RCA: 391] [Impact Index Per Article: 65.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 07/07/2019] [Accepted: 07/17/2019] [Indexed: 12/20/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disease characterized by progressive memory loss along with neuropsychiatric symptoms and a decline in activities of daily life. Its main pathological features are cerebral atrophy, amyloid plaques, and neurofibrillary tangles in the brains of patients. There are various descriptive hypotheses regarding the causes of AD, including the cholinergic hypothesis, amyloid hypothesis, tau propagation hypothesis, mitochondrial cascade hypothesis, calcium homeostasis hypothesis, neurovascular hypothesis, inflammatory hypothesis, metal ion hypothesis, and lymphatic system hypothesis. However, the ultimate etiology of AD remains obscure. In this review, we discuss the main hypotheses of AD and related clinical trials. Wealthy puzzles and lessons have made it possible to develop explanatory theories and identify potential strategies for therapeutic interventions for AD. The combination of hypometabolism and autophagy deficiency is likely to be a causative factor for AD. We further propose that fluoxetine, a selective serotonin reuptake inhibitor, has the potential to treat AD.
Collapse
Affiliation(s)
- Pei-Pei Liu
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Yi Xie
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Xiao-Yan Meng
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| | - Jian-Sheng Kang
- Clinical Systems Biology Laboratories, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052 Henan China
| |
Collapse
|
16
|
Nolte ED, Nolte KA, Yan SS. Anxiety and task performance changes in an aging mouse model. Biochem Biophys Res Commun 2019; 514:246-251. [PMID: 31029428 DOI: 10.1016/j.bbrc.2019.04.049] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Accepted: 04/07/2019] [Indexed: 12/13/2022]
Abstract
Due to the increasing focus on aging as an important risk factor for many serious diseases and an emphasis on animal models that have translational value, an increasing number of animal models are being aged. Animal behavior tests can be used to assess effects of aging in mouse models. Female mice begin exhibiting anxiety-like behaviors at 12 months of age which become more serious at 24 months, while males exhibit no age-induced anxiety-like behaviors. Males and females equally demonstrate a failure of daily task performance at 24 months. Despite these cognitive changes, the mice do not show changes in gross motor function. These results suggest cognitive impairment in non-genetically modified aging mice.
Collapse
Affiliation(s)
- Erika D Nolte
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66047, USA
| | - Keith A Nolte
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66047, USA
| | - Shirley ShiDu Yan
- Department of Pharmacology and Toxicology, Higuchi Bioscience Center, University of Kansas, Lawrence, KS 66047, USA.
| |
Collapse
|
17
|
Activation of α7 nicotinic acetylcholine receptor alleviates Aβ1-42-induced neurotoxicity via downregulation of p38 and JNK MAPK signaling pathways. Neurochem Int 2018; 120:238-250. [DOI: 10.1016/j.neuint.2018.09.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Revised: 08/23/2018] [Accepted: 09/09/2018] [Indexed: 01/08/2023]
|
18
|
Singhrang N, Tocharus C, Thummayot S, Sutheerawattananonda M, Tocharus J. Protective effects of silk lutein extract from Bombyx mori cocoons on β-Amyloid peptide-induced apoptosis in PC12 cells. Biomed Pharmacother 2018; 103:582-587. [DOI: 10.1016/j.biopha.2018.04.045] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2018] [Revised: 04/06/2018] [Accepted: 04/06/2018] [Indexed: 10/17/2022] Open
|
19
|
Yu Q, Du F, Douglas JT, Yu H, Yan SS, Yan SF. Mitochondrial Dysfunction Triggers Synaptic Deficits via Activation of p38 MAP Kinase Signaling in Differentiated Alzheimer's Disease Trans-Mitochondrial Cybrid Cells. J Alzheimers Dis 2018; 59:223-239. [PMID: 28598851 DOI: 10.3233/jad-170283] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Loss of synapse and synaptic dysfunction contribute importantly to cognitive impairment in Alzheimer's disease (AD). Mitochondrial dysfunction and oxidative stress are early pathological features in AD-affected brain. However, the effect of AD mitochondria on synaptogenesis remains to be determined. Using human trans-mitochondrial "cybrid" (cytoplasmic hybrid) neuronal cells whose mitochondria were transferred from platelets of patients with sporadic AD or age-matched non-AD subjects with relatively normal cognition, we provide the first evidence of mitochondrial dysfunction compromises synaptic development and formation of synapse in AD cybrid cells in response to chemical-induced neuronal differentiation. Compared to non-AD control cybrids, AD cybrid cells showed synaptic loss which was evidenced by a significant reduction in expression of two synaptic marker proteins: synaptophysin (presynaptic marker) and postsynaptic density protein-95, and neuronal proteins (MAP-2 and NeuN) upon neuronal differentiation. In parallel, AD-mediated synaptic deficits correlate to mitochondrial dysfunction and oxidative stress as well as activation of p38 MAP kinase. Notably, inhibition of p38 MAP kinase by pharmacological specific p38 inhibitor significantly increased synaptic density, improved mitochondrial function, and reduced oxidative stress. These results suggest that activation of p38 MAP kinase signaling pathway contributes to AD-mediated impairment in neurogenesis, possibly by inhibiting the neuronal differentiation. Our results provide new insight into the crosstalk of dysfunctional AD mitochondria to synaptic formation and maturation via activation of p38 MAP kinase. Therefore, blockade of p38 MAP kinase signal transduction could be a potential therapeutic strategy for AD by alleviating loss of synapses.
Collapse
Affiliation(s)
- Qing Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Cheng Du, China.,Departments of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Fang Du
- Departments of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Justin T Douglas
- Nuclear Magnetic Resonance Laboratory, Molecular Structures group, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Cheng Du, China
| | - Shirley ShiDu Yan
- Departments of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| | - Shi Fang Yan
- Departments of Pharmacology and Toxicology, and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS, USA
| |
Collapse
|
20
|
Criscuolo C, Cerri E, Fabiani C, Capsoni S, Cattaneo A, Domenici L. The retina as a window to early dysfunctions of Alzheimer's disease following studies with a 5xFAD mouse model. Neurobiol Aging 2018; 67:181-188. [PMID: 29735432 DOI: 10.1016/j.neurobiolaging.2018.03.017] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 03/10/2018] [Accepted: 03/16/2018] [Indexed: 10/17/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease leading to neuronal dysfunctions with cognitive impairment. AD can affect visual pathways and visual cortex and result in various visual changes and problems. However, how early the visual dysfunctions occur in AD is still a matter of discussion. Here, we used electrophysiological techniques to show the presence of early anomalies in AD visual system. To this aim, we used a familial AD (FAD) model, the 5xFAD transgenic mouse, characterized by severe progressive amyloid pathology and cognitive deficits. We investigated the retina and primary visual cortex responsivity together with behavioral assessment of the visual acuity. Visual tests and recordings were conducted at different ages in 5xFAD mice, corresponding to different phases of neurodegeneration and beta amyloid accumulation. We showed that the visual system is impaired in 5xFAD mice. In particular, we found that the inner retina impairment precedes neuronal disorders in other brain areas and cognitive deficits. Thus, noninvasive retinal electrophysiology can provide a support for assessing early visual dysfunctions in AD.
Collapse
Affiliation(s)
- Chiara Criscuolo
- Neuroscience Institute of the National Council of Research (CNR), Pisa, Italy
| | - Elisa Cerri
- Neuroscience Institute of the National Council of Research (CNR), Pisa, Italy
| | - Carlotta Fabiani
- Neuroscience Institute of the National Council of Research (CNR), Pisa, Italy
| | - Simona Capsoni
- Neuroscience Institute of the National Council of Research (CNR), Pisa, Italy; Bio@SNS Laboratory, Scuola Normale Superiore, Pisa, Italy; Department of Biomedical and Surgical Specialty Sciences, University of Ferrara, Ferrara, Italy
| | | | - Luciano Domenici
- Neuroscience Institute of the National Council of Research (CNR), Pisa, Italy; Department of Biotechnological and Applied Clinical Sciences (DISCAB), University of L'Aquila, L'Aquila, Italy.
| |
Collapse
|
21
|
Gu Y, Ma LJ, Bai XX, Jie J, Zhang XF, Chen D, Li XP. Mitogen-activated protein kinase phosphatase 1 protects PC12 cells from amyloid beta-induced neurotoxicity. Neural Regen Res 2018; 13:1842-1850. [PMID: 30136701 PMCID: PMC6128043 DOI: 10.4103/1673-5374.238621] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The mitogen-activated protein kinase (MAPK) signaling pathway plays an important role in the regulation of cell growth, proliferation, differentiation, transformation and death. Mitogen-activated protein kinase phosphatase 1 (MKP1) has an inhibitory effect on the p38MAPK and JNK pathways, but it is unknown whether it plays a role in Aβ-induced oxidative stress and neuronal inflammation. In this study, PC12 cells were infected with MKP1 shRNA, MKP1 lentivirus or control lentivirus for 12 hours, and then treated with 0.1, 1, 10 or 100 μM amyloid beta 42 (Aβ42). The cell survival rate was measured using the cell counting kit-8 assay. MKP1, tumor necrosis factor-alpha (TNF-α) and interleukin-1β (IL-1β) mRNA expression levels were analyzed using quantitative real time-polymerase chain reaction. MKP1 and phospho-c-Jun N-terminal kinase (JNK) expression levels were assessed using western blot assay. Reactive oxygen species (ROS) levels were detected using 2′,7′-dichlorofluorescein diacetate. Mitochondrial membrane potential was measured using flow cytometry. Superoxide dismutase activity and malondialdehyde levels were evaluated using the colorimetric method. Lactate dehydrogenase activity was measured using a microplate reader. Caspase-3 expression levels were assessed by enzyme-linked immunosorbent assay. Apoptosis was evaluated using the terminal deoxynucleotidyl transferase dUTP nick end labeling method. MKP1 overexpression inhibited Aβ-induced JNK phosphorylation and the increase in ROS levels. It also suppressed the Aβ-induced increase in TNF-α and IL-1β levels as well as apoptosis in PC12 cells. In contrast, MKP1 knockdown by RNA interference aggravated Aβ-induced oxidative stress, inflammation and cell damage in PC12 cells. Furthermore, the JNK-specific inhibitor SP600125 abolished this effect of MKP1 knockdown on Aβ-induced neurotoxicity. Collectively, these results show that MKP1 mitigates Aβ-induced apoptosis, oxidative stress and neuroinflammation by inhibiting the JNK signaling pathway, thereby playing a neuroprotective role.
Collapse
Affiliation(s)
- Yue Gu
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Lian-Jun Ma
- Endoscopy Center, the China-Japan Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Xue Bai
- Cadre's Wards, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Jing Jie
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiu-Fang Zhang
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Dong Chen
- Department of Respiratory Medicine, the First Hospital of Jilin University, Changchun, Jilin Province, China
| | - Xiao-Ping Li
- Department of Pediatrics, the First Hospital of Jilin University, Changchun, Jilin Province, China
| |
Collapse
|
22
|
Lachén-Montes M, González-Morales A, Zelaya MV, Pérez-Valderrama E, Ausín K, Ferrer I, Fernández-Irigoyen J, Santamaría E. Olfactory bulb neuroproteomics reveals a chronological perturbation of survival routes and a disruption of prohibitin complex during Alzheimer's disease progression. Sci Rep 2017; 7:9115. [PMID: 28831118 PMCID: PMC5567385 DOI: 10.1038/s41598-017-09481-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 07/26/2017] [Indexed: 12/31/2022] Open
Abstract
Olfactory dysfunction is among the earliest features of Alzheimer’s disease (AD). Although neuropathological abnormalities have been detected in the olfactory bulb (OB), little is known about its dynamic biology. Here, OB- proteome analysis showed a stage-dependent synaptic proteostasis impairment during AD evolution. In addition to progressive modulation of tau and amyloid precursor protein (APP) interactomes, network-driven proteomics revealed an early disruption of upstream and downstream p38 MAPK pathway and a subsequent impairment of Phosphoinositide-dependent protein kinase 1 (PDK1)/Protein kinase C (PKC) signaling axis in the OB from AD subjects. Moreover, a mitochondrial imbalance was evidenced by a depletion of Prohibitin-2 (Phb2) levels and a specific decrease in the phosphorylated isoforms of Phb1 in intermediate and advanced AD stages. Interestingly, olfactory Phb subunits were also deregulated across different types of dementia. Phb2 showed a specific up-regulation in mixed dementia, while Phb1 isoforms were down-regulated in frontotemporal lobar degeneration (FTLD). However, no differences were observed in the olfactory expression of Phb subunits in progressive supranuclear palsy (PSP). To sum up, our data reflect, in part, the missing links in the biochemical understanding of olfactory dysfunction in AD, unveiling Phb complex as a differential driver of neurodegeneration at olfactory level.
Collapse
Affiliation(s)
- Mercedes Lachén-Montes
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Andrea González-Morales
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - María Victoria Zelaya
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Pathological Anatomy Department, Navarra Hospital Complex, Pamplona, Spain
| | - Estela Pérez-Valderrama
- Proteored-ISCIII. Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Karina Ausín
- Proteored-ISCIII. Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Isidro Ferrer
- Institut de Neuropatologia, IDIBELL-Hospital Universitari de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, CIBERNED (Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas), Barcelona, Spain
| | - Joaquín Fernández-Irigoyen
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.,IDISNA, Navarra Institute for Health Research, Pamplona, Spain.,Proteored-ISCIII. Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain
| | - Enrique Santamaría
- Clinical Neuroproteomics Group, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain. .,IDISNA, Navarra Institute for Health Research, Pamplona, Spain. .,Proteored-ISCIII. Proteomics Unit, Navarrabiomed, Departamento de Salud, Universidad Pública de Navarra, Pamplona, Spain.
| |
Collapse
|
23
|
Qi XM, Ma JF. The role of amyloid beta clearance in cerebral amyloid angiopathy: more potential therapeutic targets. Transl Neurodegener 2017; 6:22. [PMID: 28824801 PMCID: PMC5559841 DOI: 10.1186/s40035-017-0091-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2017] [Accepted: 08/01/2017] [Indexed: 01/09/2023] Open
Abstract
Cerebral amyloid angiopathy (CAA) is characterized by the deposition of amyloid β-protein (Aβ) in the leptomeningeal and cortical blood vessels, which is an age-dependent risk factor for intracerebral hemorrhage (ICH), ischemic stroke and contributes to cerebrovascular dysfunction leading to cognitive impairment. However clinical prevention and treatment of the disease is very difficult because of its occult onset and severity of the symptoms. In recent years, many anti-amyloid β immunotherapies have not demonstrated clinical efficacy in subjects with Alzheimer’s disease (AD), and the failure may be due to the deposition of Aβ in the cerebrovascular export pathway resulting in further damage to blood vessels and aggravating CAA. So decreased clearance of Aβ in blood vessels plays a crucial role in the development of CAA and AD, and identification of the molecular pathways involved will provide new targets for treatment. In this review, we mainly describe the mechanisms of Aβ clearance through vessels, especially in terms of some proteins and receptors involved in this process.
Collapse
Affiliation(s)
- Xue-Mei Qi
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| | - Jian-Fang Ma
- Department of Neurology & Institute of Neurology, Ruijin Hospital Affiliated to Shanghai Jiaotong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
24
|
Antineuroinflammatory Effects of Modified Wu-Zi-Yan-Zong Prescription in β-Amyloid-Stimulated BV2 Microglia via the NF- κB and ERK/p38 MAPK Signaling Pathways. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2017; 2017:8470381. [PMID: 28798805 PMCID: PMC5535734 DOI: 10.1155/2017/8470381] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 04/08/2017] [Accepted: 05/09/2017] [Indexed: 11/17/2022]
Abstract
Modified Wu-Zi-Yan-Zong prescription (MWP), a traditional Chinese medicinal decoction, has possessed the neuroprotective and anti-inflammatory properties. The mechanisms associated with these properties, however, are not completely understood. We designed the experiments to elucidate the antineuroinflammatory property of MWP in BV2 microglia activated by β-amyloid (Aβ), which is a characteristic feature of Alzheimer's disease (AD). The composition of MWP was studied using HPLC. BV2 microglia cells were then treated with Aβ in the presence or absence of MWP. The effects of MWP treatment on Aβ-activated neuroinflammation were determined using PCR, western blotting, and immunofluorescence staining. MWP significantly inhibited the mRNA expression of inflammatory mediators such as IL-1β, IL-6, TNF-α, and MCP-1, as well as the expression of inducible nitric oxide synthase (iNOS) in Aβ-activated BV2 microglia. MWP also inhibited the nuclear translocation and signaling pathway of nuclear factor kappa B (NF-κB) by suppressing inhibitor of nuclear factor-κB (IκB) degradation and downregulating IκB kinase β (IKKβ) phosphorylation. Moreover, MWP decreased extracellular regulated protein kinase (ERK)/p38 mitogen-activated protein kinase (MAPK) phosphorylation, which is an important signaling pathway for proinflammatory gene expression. We concluded that MWP could suppress neuroinflammatory responses in Aβ-activated BV2 microglia via the NF-κB and ERK/p38 MAPK signaling cascades and could prove an effective therapeutic agent for the prevention and treatment of neuroinflammatory diseases such as AD.
Collapse
|
25
|
Li K, Wei Q, Liu FF, Hu F, Xie AJ, Zhu LQ, Liu D. Synaptic Dysfunction in Alzheimer's Disease: Aβ, Tau, and Epigenetic Alterations. Mol Neurobiol 2017; 55:3021-3032. [PMID: 28456942 DOI: 10.1007/s12035-017-0533-3] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Accepted: 04/06/2017] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) is a complex neurodegenerative disorder characterized in the early stages by loss of learning and memory. However, the mechanism underlying these symptoms remains unclear. The best correlation between cognitive decline and pathological changes is in synaptic dysfunction. Histopathological hallmarks of AD are the abnormal aggregation of Aβ and Tau. Evidence suggests that Aβ and Tau oligomers contribute to synaptic loss in AD. Recently, direct links between epigenetic alterations, such as dysfunction in non-coding RNAs (ncRNAs), and synaptic pathologies have emerged, raising interest in exploring the potential roles of ncRNAs in the synaptic deficits in AD. In this paper, we summarize the potential roles of Aβ, Tau, and epigenetic alterations (especially by ncRNAs) in the synaptic dysfunction of AD and discuss the novel findings in this area.
Collapse
Affiliation(s)
- Ke Li
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Qing Wei
- Department of Blood Transfusion, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Fang-Fang Liu
- Department of Pathology, Central Hospital of Wuhan, Wuhan, 430014, People's Republic of China
| | - Fan Hu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ao-Ji Xie
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China
| | - Ling-Qiang Zhu
- Department of Pathophysiology, Key Lab of Neurological Disorder of Education Ministry, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| | - Dan Liu
- Department of Medical Genetics, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, People's Republic of China.
| |
Collapse
|
26
|
Zhao C, Zhang H, Li H, Lv C, Liu X, Li Z, Xin W, Wang Y, Zhang W. Geniposide ameliorates cognitive deficits by attenuating the cholinergic defect and amyloidosis in middle-aged Alzheimer model mice. Neuropharmacology 2016; 116:18-29. [PMID: 27940040 DOI: 10.1016/j.neuropharm.2016.12.002] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 11/30/2016] [Accepted: 12/03/2016] [Indexed: 01/18/2023]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by memory deficits and cognitive decline. Amyloid-β (Aβ) deposition and cholinergic defect are widely thought to be the underlying mechanism of learning and memory impairment. Geniposide, which is the main active component of the traditional Chinese herbal Gardenia jasminoides Ellis, elicits neuroprotective effects by alleviating inflammation responses and oxidative damages. In this study, we investigated the protective effect of geniposide on levels of cholinergic markers, RAGE, RAGE-dependent signalling pathways and amyloid accumulation in the APPswe/PS1dE9 AD model mouse. Geniposide suppressed MAPK signaling over-activation mediated by Aβ-RAGE interaction, resulting in reduced Aβ accumulation and amelioration of cholinergic deficits in the cerebral hippocampus. Furthermore, geniposide inhibited the toxic effect of oligomeric Aβ1-42 induced cholinergic deficit by increasing ChAT levels and activity but decreasing AChE activity in cultured primary hippocampal neurons. These results indicated that geniposide enhanced cholinergic neurotransmission, which likely contributes to its memory enhancing effect.
Collapse
Affiliation(s)
- Chunhui Zhao
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100875, China; Institute of Chinese Materia Medica, China Academy of Chinese Medical Sciences, Beijing 100700, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China.
| | - Haijing Zhang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China.
| | - Hang Li
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China.
| | - Cui Lv
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100875, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China; Laboratory of Immunology for Environment and Health, Shandong Analysis and Test Center, Shandong Academy of Science, Jinan 250014, China.
| | - Xiaoli Liu
- Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China.
| | - Zhi Li
- Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China.
| | - Wenfeng Xin
- Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Sanqi Biotechnology and Pharmaceutical, Yun Nan Province, Kunming 650000, China.
| | - Yongyan Wang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China.
| | - Wensheng Zhang
- Beijing Area Major Laboratory of Protection and Utilization of Traditional Chinese Medicine, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Natural Medicine, Ministry of Education, Beijing Normal University, Beijing 100875, China; College of Resources Science Technology, Beijing Normal University, Beijing 100875, China; Engineering Research Center of Sanqi Biotechnology and Pharmaceutical, Yun Nan Province, Kunming 650000, China.
| |
Collapse
|
27
|
Jang WY, Lee BR, Jeong J, Sung Y, Choi M, Song P, Kim H, Jang S, Kim H, Joo KI, Lee JW, Choo YS, Kim E, Ryoo ZY. Overexpression of serum amyloid a 1 induces depressive-like behavior in mice. Brain Res 2016; 1654:55-65. [PMID: 27608955 DOI: 10.1016/j.brainres.2016.09.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Revised: 08/30/2016] [Accepted: 09/04/2016] [Indexed: 11/27/2022]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by loss of memory and cognitive abilities. In AD, amyloid β (Aβ) protein aggregates in the brain of patients, forming amyloid plaques. Aβ plaques are known to be surrounded by activated microglial cells. Serum amyloid A (SAA) is elevated from several hundred to 1000-fold as part of the immune response against various injuries, including trauma, infection, and inflammation. Additionally, continuous elevation of SAA is related to the development of amyloidosis. This study was designed to identify the relationship between SAA1 and AD using liver specific SAA1 overexpressing mice (TG), because SAA1 is expressed in the liver during the acute phase. We detected exogenous SAA1 expression in the brain of TG mice. This result implies that liver-derived SAA1 migrates to the brain tissues. Thus, we confirmed that the blood brain barrier (BBB) functioned normally using Evans-blue staining and CARS. Furthermore, our results show an increase in the accumulation of the 87kDa form of Aβ in TG mice compared to wild type mice (WT). Additionally, the number of microglial cells and levels of pro-inflammatory cytokines were increased. Next, we investigated the relationship between SAA1 and depression by performing social interaction tests. The results showed that TG mice have a tendency to avoid stranger mice and an impaired social recognition. In conclusion, the SAA1 TG mouse model is a valuable model to study depression.
Collapse
Affiliation(s)
- Woo Young Jang
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Bo-Ram Lee
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea; Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea
| | - Jain Jeong
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Younghun Sung
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Minjee Choi
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Park Song
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Hyerim Kim
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Soyoung Jang
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Hyunmin Kim
- Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea
| | - Kyung-Il Joo
- Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea
| | - Jeong-Woong Lee
- Biotherapeutics Translational Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| | - Yeon Sik Choo
- Department of Biology, Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea
| | - Eunjoo Kim
- Division of Nano & Energy Convergence Research, Daegu Gyeongbuk Institute of Science and Technology (DGIST), Daegu 711-873, Republic of Korea.
| | - Zae Young Ryoo
- School of Life Science and Biotechnology, KNU Creative BioResearch Group (BK21 plus project), Kyungpook National University, 1370 Sankyuk-dong, Buk-ku, Daegu 702-701, Republic of Korea.
| |
Collapse
|
28
|
microRNA-132/212 deficiency enhances Aβ production and senile plaque deposition in Alzheimer's disease triple transgenic mice. Sci Rep 2016; 6:30953. [PMID: 27484949 PMCID: PMC4971468 DOI: 10.1038/srep30953] [Citation(s) in RCA: 104] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Accepted: 07/11/2016] [Indexed: 01/02/2023] Open
Abstract
The abnormal regulation of amyloid-β (Aβ) metabolism (e.g., production, cleavage, clearance) plays a central role in Alzheimer’s disease (AD). Among endogenous factors believed to participate in AD progression are the small regulatory non-coding microRNAs (miRs). In particular, the miR-132/212 cluster is severely reduced in the AD brain. In previous studies we have shown that miR-132/212 deficiency in mice leads to impaired memory and enhanced Tau pathology as seen in AD patients. Here we demonstrate that the genetic deletion of miR-132/212 promotes Aβ production and amyloid (senile) plaque formation in triple transgenic AD (3xTg-AD) mice. Using RNA-Seq and bioinformatics, we identified genes of the miR-132/212 network with documented roles in the regulation of Aβ metabolism, including Tau, Mapk, and Sirt1. Consistent with these findings, we show that the modulation of miR-132, or its target Sirt1, can directly regulate Aβ production in cells. Finally, both miR-132 and Sirt1 levels correlated with Aβ load in humans. Overall, our results support the hypothesis that the miR-132/212 network, including Sirt1 and likely other target genes, contributes to abnormal Aβ metabolism and senile plaque deposition in AD. This study strengthens the importance of miR-dependent networks in neurodegenerative disorders, and opens the door to multifactorial drug targets of AD by targeting Aβ and Tau.
Collapse
|
29
|
An early dysregulation of FAK and MEK/ERK signaling pathways precedes the β-amyloid deposition in the olfactory bulb of APP/PS1 mouse model of Alzheimer's disease. J Proteomics 2016; 148:149-58. [PMID: 27498392 DOI: 10.1016/j.jprot.2016.07.032] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 06/28/2016] [Accepted: 07/31/2016] [Indexed: 01/25/2023]
Abstract
UNLABELLED Olfactory dysfunction is an early event of Alzheimer's disease (AD). However, the mechanisms associated to AD neurodegeneration in olfactory areas are unknown. Here we used double-transgenic amyloid precursor protein/presenilin 1 (APPswe/PS1dE9) mice and label-free quantitative proteomics to analyze early pathological effects on the olfactory bulb (OB) during AD progression. Prior to β-amyloid plaque formation, 9 modulated proteins were detected on 3-month-old APP/PS1 mice while 16 differential expressed proteins were detected at 6months, when β-amyloid plaques appear, indicating a moderate imbalance in cytoskeletal rearrangement, and synaptic plasticity in APP/PS1 OBs. Moreover, β-amyloid induced an inactivation of focal adhesion kinase (FAK) together with a transient activation of MEK1/2, leading to inactivation of ERK1/2 in 6-months APP/PS1 OBs. In contrast, the analysis of human OBs revealed a late activation of FAK in advanced AD stages, whereas ERK1/2 activation was enhanced across AD staging respect to controls. This survival potential was accompanied by the inhibition of the proapototic factor BAD in the OB across AD phenotypes. Our data contribute to a better understanding of the early molecular mechanisms that are modulated in AD neurodegeneration, highlighting significant differences in the regulation of survival pathways between APP/PS1 mice and sporadic human AD. SIGNIFICANCE Loss of smell is involved in early stages of Alzheimer's disease (AD), usually preceding classic disease symptoms. However, the mechanisms governing this dysfunction are still poorly understood, losing its potential as a useful tool for clinical diagnosis. Our study characterizes potential AD-associated molecular changes in APP/PS1 mice olfactory bulb (OB) using MS-quantitative proteomics, revealing early cytoskeletal disruption and synaptic plasticity impairment. Moreover, an opposite pattern was found when comparing the activation status of specific survival pathways between APP/PS1 OBs and OBs derived from sAD subjects with different neuropathological grading. Our data reflect, in part, the progressive effect of APP overproduction and Aβ accumulation on the OB proteome during AD progression.
Collapse
|
30
|
Gastrodin Protects Neural Progenitor Cells Against Amyloid β (1-42)-Induced Neurotoxicity and Improves Hippocampal Neurogenesis in Amyloid β (1-42)-Injected Mice. J Mol Neurosci 2016; 60:21-32. [PMID: 27112440 DOI: 10.1007/s12031-016-0758-z] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 04/11/2016] [Indexed: 12/13/2022]
Abstract
The aim of this study was to investigate the neuroprotective effects of gastrodin (GAS), one of the major bioactive components of Gastrodia elata Blume (Tian Ma), against amyloid β (Aβ) (1-42)-induced neurotoxicity in primary neural progenitor cells (NPCs). We found that pretreatment with GAS not only prevents a loss in cell viability following treatment with Aβ (1-42) but also counteracts Aβ (1-42)-triggered release of pro-inflammatory cytokines and nitric oxide (NO) in a dose-dependent manner. Additionally, GAS was able to attenuate Aβ (1-42)-induced apoptosis in NPCs, evidenced by the decreased percentage of apoptotic cells and altered expression of apoptosis-related proteins in response to GAS pretreatment prior to Aβ (1-42) exposure. Furthermore, in Aβ (1-42)-injected C57BL/6 mice, we found that systemic administration of GAS could improve hippocampal neurogenesis, manifested by the increased number of SOX-2 and doublecortin (DCX)-positive cells in the DG area. Mechanistic studies revealed that in NPCs, GAS could reverse the Aβ (1-42)-induced increase in phosphorylation of MEK-1/2, extracellular signal-regulated kinases (ERK), and c-Jun N-terminal kinase (JNK). When combining GAS with the MEK inhibitor U0126 or the JNK inhibitor SP600125, we observed a synergistic effect against Aβ (1-42)-induced reduction in cell viability of NPCs. Taken together, these results show the efficacy and underlying mechanism of GAS against amyloid β (1-42)-induced neurotoxicity and provide substantial insight into the potential merits of GAS for its clinical application in the treatment of Alzheimer's disease.
Collapse
|
31
|
Yang H, Wang S, Yu L, Zhu X, Xu Y. Esculentoside A suppresses Aβ1–42-induced neuroinflammation by down-regulating MAPKs pathwaysin vivo. Neurol Res 2015; 37:859-66. [DOI: 10.1179/1743132815y.0000000066] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
32
|
Wan W, Cao L, Liu L, Zhang C, Kalionis B, Tai X, Li Y, Xia S. Aβ(1-42) oligomer-induced leakage in an in vitro blood-brain barrier model is associated with up-regulation of RAGE and metalloproteinases, and down-regulation of tight junction scaffold proteins. J Neurochem 2015; 134:382-93. [PMID: 25866188 DOI: 10.1111/jnc.13122] [Citation(s) in RCA: 122] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 03/13/2015] [Accepted: 03/31/2015] [Indexed: 12/20/2022]
Abstract
Accumulating evidence indicates that abnormal deposition of amyloid-β (Aβ) peptide in the brain is responsible for endothelial cell damage and consequently leads to blood-brain barrier (BBB) leakage. However, the mechanisms underlying BBB disruption are not well described. We employed an monolayer BBB model comprising bEnd.3 cell and found that BBB leakage was induced by treatment with Aβ(1-42), and the levels of tight junction (TJ) scaffold proteins (ZO-1, Claudin-5, and Occludin) were decreased. Through comparisons of the effects of the different components of Aβ(1-42), including monomer (Aβ(1-42)-Mono), oligomer (Aβ(1-42)-Oligo), and fibril (Aβ(1-42)-Fibril), our data confirmed that Aβ(1-42)-Oligo is likely to be the most important damage factor that results in TJ damage and BBB leakage in Alzheimer's disease. We found that the incubation of bEnd.3 cells with Aβ(1-42) significantly up-regulated the level of receptor for advanced glycation end-products (RAGE). Co-incubation of a polyclonal antibody to RAGE and Aβ(1-42)-Oligo in bEnd.3 cells blocked RAGE suppression of Aβ(1-42)-Oligo-induced alterations in TJ scaffold proteins and reversed Aβ(1-42)-Oligo-induced up-regulation of RAGE, matrix metalloproteinase (MMP)-2, and MMP-9. Furthermore, we found that these effects induced by Aβ(1-42)-Oligo treatment were effectively suppressed by knockdown of RAGE using small interfering RNA (siRNA) transfection. We also found that GM 6001, a broad-spectrum MMP inhibitor, partially reversed the Aβ(1-42)-Oligo-induced inhibitor effects in bEnd.3 cells. Thus, these results suggested that RAGE played an important role in Aβ-induced BBB leakage and alterations of TJ scaffold proteins, through a mechanism that involved up-regulation of MMP-2 and MMP-9.
Collapse
Affiliation(s)
- Wenbin Wan
- Department of Neurology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lan Cao
- State Key Laboratory of Medical Neurobiology, Department of Neurobiology and Institutes of Brain Science, School of Basic Medical Science, Fudan University, Shanghai, China
| | - Lumei Liu
- Geriatrics Department of Chinese Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Chinese Medicine, Dongfang Hospital, Tongji University, Shanghai, China
| | - Bill Kalionis
- Department of Perinatal Medicine Pregnancy Research Centre and University of Melbourne Department of Obstetrics and Gynaecology, Royal Women's Hospital, Parkville, VIC, Australia
| | - Xiantao Tai
- School of Acupuncture, Massage and Rehabilitation, Yunnan University of Traditional Chinese Medicine, Kunming, China
| | - Yaming Li
- Geriatrics Department of Chinese Medicine, Huadong Hospital, Fudan University, Shanghai, China
| | - Shijin Xia
- Shanghai Institute of Geriatrics, Huadong Hospital, Fudan University, Shanghai, China
| |
Collapse
|
33
|
Teich AF, Nicholls RE, Puzzo D, Fiorito J, Purgatorio R, Fa’ M, Arancio O. Synaptic therapy in Alzheimer's disease: a CREB-centric approach. Neurotherapeutics 2015; 12:29-41. [PMID: 25575647 PMCID: PMC4322064 DOI: 10.1007/s13311-014-0327-5] [Citation(s) in RCA: 118] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Therapeutic attempts to cure Alzheimer's disease (AD) have failed, and new strategies are desperately needed. Motivated by this reality, many laboratories (including our own) have focused on synaptic dysfunction in AD because synaptic changes are highly correlated with the severity of clinical dementia. In particular, memory formation is accompanied by altered synaptic strength, and this phenomenon (and its dysfunction in AD) has been a recent focus for many laboratories. The molecule cyclic adenosine monophosphate response element-binding protein (CREB) is at a central converging point of pathways and mechanisms activated during the processes of synaptic strengthening and memory formation, as CREB phosphorylation leads to transcription of memory-associated genes. Disruption of these mechanisms in AD results in a reduction of CREB activation with accompanying memory impairment. Thus, it is likely that strategies aimed at these mechanisms will lead to future therapies for AD. In this review, we will summarize literature that investigates 5 possible therapeutic pathways for rescuing synaptic dysfunction in AD: 4 enzymatic pathways that lead to CREB phosphorylation (the cyclic adenosine monophosphate cascade, the serine/threonine kinases extracellular regulated kinases 1 and 2, the nitric oxide cascade, and the calpains), as well as histone acetyltransferases and histone deacetylases (2 enzymes that regulate the histone acetylation necessary for gene transcription).
Collapse
Affiliation(s)
- Andrew F. Teich
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Russell E. Nicholls
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Daniela Puzzo
- />Department of Bio-Medical Sciences, Section of Physiology, University of Catania, Catania, 95125 Italy
| | - Jole Fiorito
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Rosa Purgatorio
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Mauro Fa’
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| | - Ottavio Arancio
- />Department of Pathology & Cell Biology, Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032 USA
| |
Collapse
|
34
|
Intracellular accumulation of amyloid-β (Aβ) protein plays a major role in Aβ-induced alterations of glutamatergic synaptic transmission and plasticity. J Neurosci 2014; 34:12893-903. [PMID: 25232124 DOI: 10.1523/jneurosci.1201-14.2014] [Citation(s) in RCA: 90] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Intracellular accumulation of amyloid-β (Aβ) protein has been proposed as an early event in AD pathogenesis. In patients with mild cognitive impairment, intraneuronal Aβ immunoreactivity was found especially in brain regions critically involved in the cognitive deficits of AD. Although a large body of evidence demonstrates that Aβ42 accumulates intraneuronally ((in)Aβ), the action and the role of Aβ42 buildup on synaptic function have been poorly investigated. Here, we demonstrate that basal synaptic transmission and LTP were markedly depressed following Aβ42 injection into the neuron through the patch pipette. Control experiments performed with the reverse peptide (Aβ42-1) allowed us to exclude that the effects of (in)Aβ depended on changes in oncotic pressure. To further investigate (in)Aβ synaptotoxicity we used an Aβ variant harboring oxidized methionine in position 35 that does not cross the neuronal plasma membrane and is not uploaded from the extracellular space. This Aβ42 variant had no effects on synaptic transmission and plasticity when applied extracellularly, but induced synaptic depression and LTP inhibition after patch-pipette dialysis. Finally, the injection of an antibody raised against human Aβ42 (6E10) in CA1 pyramidal neurons of mouse hippocampal brain slices and autaptic microcultures did not, per se, significantly affect LTP and basal synaptic transmission, but it protected against the toxic effects of extracellular Aβ42. Collectively, these findings suggest that Aβ42-induced impairment of glutamatergic synaptic function depends on its internalization and intracellular accumulation thus paving the way to a systemic proteomic analysis of intracellular targets/partners of Aβ42.
Collapse
|
35
|
RAGE inhibition in microglia prevents ischemia-dependent synaptic dysfunction in an amyloid-enriched environment. J Neurosci 2014; 34:8749-60. [PMID: 24966375 DOI: 10.1523/jneurosci.0141-14.2014] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Ischemia is known to increase the deleterious effect of β-amyloid (Aβ), contributing to early cognitive impairment in Alzheimer's disease. Here, we investigated whether transient ischemia may function as a trigger for Aβ-dependent synaptic impairment in the entorhinal cortex (EC), acting through specific cellular signaling. We found that synaptic depression induced by oxygen glucose deprivation (OGD) was enhanced in EC slices either in presence of synthetic oligomeric Aβ or in slices from mutant human amyloid precursor protein transgenic mice (mhAPP J20). OGD-induced synaptic depression was ameliorated by functional suppression of RAGE. In particular, overexpression of the dominant-negative form of RAGE targeted to microglia (DNMSR) protects against OGD-induced synaptic impairment in an amyloid-enriched environment, reducing the activation of stress-related kinases (p38MAPK and JNK) and the release of IL-1β. Our results demonstrate a prominent role for the RAGE-dependent neuroinflammatory pathway in the synaptic failure induced by Aβ and triggered by transient ischemia.
Collapse
|
36
|
Zhang H, Wang Y, Yan S, Du F, Wu L, Yan S, Yan SS. Genetic deficiency of neuronal RAGE protects against AGE-induced synaptic injury. Cell Death Dis 2014; 5:e1288. [PMID: 24922072 PMCID: PMC4611721 DOI: 10.1038/cddis.2014.248] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 04/28/2014] [Accepted: 05/05/2014] [Indexed: 01/11/2023]
Abstract
Synaptic dysfunction and degeneration is an early pathological feature of aging and age-related diseases, including Alzheimer's disease (AD). Aging is associated with increased generation and deposition of advanced glycation endproducts (AGEs), resulting from nonenzymatic glycation (or oxidation) proteins and lipids. AGE formation is accelerated in diabetes and AD-affected brain, contributing to cellular perturbation. The extent of AGEs' involvement, if at all, in alterations in synaptic structure and function is currently unknown. Here we analyze the contribution of neuronal receptor of AGEs (RAGE) signaling to AGE-mediated synaptic injury using novel transgenic neuronal RAGE knockout mice specifically targeted to the forebrain and transgenic mice expressing neuronal dominant-negative RAGE (DN-RAGE). Addition of AGEs to brain slices impaired hippocampal long-term potentiation (LTP). Similarly, treatment of hippocampal neurons with AGEs significantly decreases synaptic density. Such detrimental effects are largely reversed by genetic RAGE depletion. Notably, brain slices from mice with neuronal RAGE deficiency or DN-RAGE are resistant to AGE-induced LTP deficit. Further, RAGE deficiency or DN-RAGE blocks AGE-induced activation of p38 signaling. Taken together, these data show that neuronal RAGE functions as a signal transducer for AGE-induced synaptic dysfunction, thereby providing new insights into a mechanism by which the AGEs–RAGE-dependent signaling cascade contributes to synaptic injury via the p38 MAP kinase signal transduction pathway. Thus, RAGE blockade may be a target for development of interventions aimed at preventing the progression of cognitive decline in aging and age-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Hongju Zhang
- 1] Departments of Pharmacology and Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA [2] School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Yongfu Wang
- Departments of Pharmacology and Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| | - Shijun Yan
- Departments of Pharmacology and Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| | - Fang Du
- Departments of Pharmacology and Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| | - Long Wu
- Departments of Pharmacology and Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| | - Shiqiang Yan
- School of Chemistry and Chemical Engineering, Lanzhou University, Lanzhou, Gansu 730000, China
| | - Shirley S Yan
- Departments of Pharmacology and Toxicology and Higuchi Bioscience Center, School of Pharmacy, University of Kansas, Lawrence, KS 66047, USA
| |
Collapse
|
37
|
Wan W, Chen H, Li Y. The potential mechanisms of Aβ-receptor for advanced glycation end-products interaction disrupting tight junctions of the blood-brain barrier in Alzheimer's disease. Int J Neurosci 2013; 124:75-81. [PMID: 23855502 DOI: 10.3109/00207454.2013.825258] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
The receptor for advanced glycation end-products (RAGE) is a multiligand membrane receptor that has been implicated in the cytotoxicity effects of β-amyloid protein (Aβ) in AD. Positive feedback mechanism of RAGE within blood-brain barrier (BBB) and/or cells inside the brain is proposed, including interaction with Aβ stimulating activation of proinflammatory cytokines, release of reactive oxygen species (ROS), which leads to neuron damage and BBB dysfunction. RAGE is the main factor mediating Aβ cytotoxicity. Attenuation of RAGE activity may inhibit Aβ from accumulation in the cerebral blood vessels and prevent neurotoxicity. Furthermore, RAGE may serve as a therapeutic target for Alzheimer's disease by inhibiting pathophysiological consequences of Aβ-RAGE interaction. Tight junctions (TJ) are identified as the basic structure of the BBB and RAGE-mediated Aβ cytotoxicity to the brain microvascular endothelial cells (BMEC), resulting in damaged BBB structural integrity. However, the potential mechanism is poorly studied.
Collapse
Affiliation(s)
- Wenbin Wan
- Huadong Hospital, Fudan University , Shanghai , PR China
| | | | | |
Collapse
|
38
|
Enhanced Aβ(1-40) production in endothelial cells stimulated with fibrillar Aβ(1-42). PLoS One 2013; 8:e58194. [PMID: 23505467 PMCID: PMC3591408 DOI: 10.1371/journal.pone.0058194] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2012] [Accepted: 02/04/2013] [Indexed: 12/25/2022] Open
Abstract
Amyloid accumulation in the brain of Alzheimer’s patients results from altered processing of the 39- to 43-amino acid amyloid β protein (Aβ). The mechanisms for the elevated amyloid (Aβ1–42) are considered to be over-expression of the amyloid precursor protein (APP), enhanced cleavage of APP to Aβ, and decreased clearance of Aβ from the central nervous system (CNS). We report herein studies of Aβ stimulated effects on endothelial cells. We observe an interesting and as yet unprecedented feedback effect involving Aβ1–42 fibril-induced synthesis of APP by Western blot analysis in the endothelial cell line Hep-1. We further observe an increase in the expression of Aβ1–40 by flow cytometry and fluorescence microscopy. This phenomenon is reproducible for cultures grown both in the presence and absence of serum. In the former case, flow cytometry reveals that Aβ1–40 accumulation is less pronounced than under serum-free conditions. Immunofluorescence staining further corroborates these observations. Cellular responses to fibrillar Aβ1–42 treatment involving eNOS upregulation and increased autophagy are also reported.
Collapse
|
39
|
Effect of α-synuclein on amyloid β-induced toxicity: relevance to Lewy body variant of Alzheimer disease. Neurochem Res 2013; 38:797-806. [PMID: 23389658 DOI: 10.1007/s11064-013-0982-7] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2012] [Revised: 12/30/2012] [Accepted: 01/25/2013] [Indexed: 10/27/2022]
Abstract
Alzheimer's disease, the most prevalent age-related neurodegenerative disease, is characterized by the presence of extracellular senile plaques composed of amyloid-beta (Aβ) peptide and intracellular neurofibrillary tangles. More than 50 % of Alzheimer's disease (AD) patients also exhibit abundant accumulation of α-synuclein (α-Syn)-positive Lewy bodies. This Lewy body variant of AD (LBV-AD) is associated with accelerated cognitive dysfunction and progresses more rapidly than pure AD. In addition, it has been suggested that Aβ and α-Syn can directly interact. In this study we investigated the effect of α-Syn on Aβ-induced toxicity in cortical neurons. In order to mimic the intracellular accumulation of α-Syn observed in the brain of LBV-AD patients, we used valproic acid (VPA) to increase its endogenous expression levels. The release of α-Syn from damaged presynaptic terminals that occurs during the course of the disease was simulated by challenging cells with recombinant α-Syn. Our results showed that either VPA-induced α-Syn upregulation or addition of recombinant α-Syn protect primary cortical neurons from soluble Aβ1-42 decreasing the caspase-3-mediated cell death. It was also found that neuroprotection against Aβ-induced toxicity mediated by α-Syn overexpression involves the PI3K/Akt cell survival pathway. Furthermore, recombinant α-Syn was shown to directly interact with Aβ1-42 and to decrease the levels of Aβ1-42 oligomers, which might explain its neuroprotective effect. In conclusion, we demonstrate that either endogenous or exogenous α-Syn can be neuroprotective against Aβ-induced cell death, suggesting a cell defence mechanism during the initial stages of the mixed pathology.
Collapse
|
40
|
Zhao L, Wang JL, Wang YR, Fa XZ. Apigenin attenuates copper-mediated β-amyloid neurotoxicity through antioxidation, mitochondrion protection and MAPK signal inactivation in an AD cell model. Brain Res 2012. [PMID: 23178511 DOI: 10.1016/j.brainres.2012.11.019] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Apigenin, belonging to a less toxic and non-mutagenic flavone subclass of flavonoids, has been reported to possess numerous biological activities beneficial to health. Although evidence has shown apigenin might exert its protective effects by reducing the toxicity induced by amyloid-β peptides (Aβ), the precise mechanism is unclear. In the present study, we investigated the in vitro neuroprotective activity of apigenin interrelated with amyloid toxicity and mental homeostasis in an Alzheimer's disease (AD) cell model and explored its potential signal transduction. Our results showed that apigenin protected neurons against Aβ-mediated toxicity induced by copper, which was characterized by increasing neuronal viability and relieving mitochondrial membrane dissipation and neuronal nuclear condensation. Further, we demonstrated that apigenin did not provide sufficient effect on decreasing β-amyloid precursor protein (AβPP) expression and lowering Aβ(1-42) secretion, but conserved redox balance by increasing intracellular glutathione levels and enhancing cellular superoxide dismutase and glutathione peroxidase activities, reduced intracellular reactive oxygen species (ROS) generation, blocked ROS-induced p38 mitogen-activated protein kinases (p38 MAPK)- MAPKAP kinase-2 (MK2)-heat shock protein 27 (Hsp27) and stress-activated protein kinase (SAPK)/c-Jun N-terminal kinase (JNK)-c-Jun signaling pathways, preserved mitochondrial function, and then regulated apoptotic pathways. In conclusion, apigenin could exert neuroprotection against Aβ-induced toxicity in the presence of copper mainly through the mechanisms that regulate redox imbalance, preserve mitochondrial function, inhibit MAPK pathways, and depress neuronal apoptosis.
Collapse
Affiliation(s)
- Le Zhao
- College of Life and Environmental Sciences, Minzu University of China, 27 South Street, Zhongguancun Beijing 100081, PR China
| | | | | | | |
Collapse
|
41
|
Mazargui H, Lévêque C, Bartnik D, Fantini J, Gouget T, Melone MAB, Funke SA, Willbold D, Perrone L. A synthetic amino acid substitution of Tyr10 in Aβ peptide sequence yields a dominant negative variant in amyloidogenesis. Aging Cell 2012; 11:530-41. [PMID: 22385841 DOI: 10.1111/j.1474-9726.2012.00814.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia in elderly people, and age is the major nongenetic risk factor for sporadic AD. A hallmark of AD is the accumulation of amyloid in the brain, which is composed mainly of the amyloid beta-peptide (Aβ) in the form of oligomers and fibrils. However, how aging induces Aβ aggregation is not yet fully determined. Some residues in the Aβ sequence seem to promote Aβ-induced toxicity in association with age-dependent risk factors for AD, such as (i) increased GM1 brain membrane content, (ii) altered lipid domain in brain membrane, (iii) oxidative stress. However, the role of Aβ sequence in promoting aggregation following interaction with the plasma membrane is not yet demonstrated. As Tyr10 is implicated in the induction of oxidative stress and stabilization of Aβ aggregation, we substituted Tyr 10 with a synthetic amino acid that abolishes Aβ-induced oxidative stress and shows an accelerated interaction with GM1. This variant peptide shows impaired aggregation properties and increased affinity for GM1. It has a dominant negative effect on amyloidogenesis in vitro, in cellulo, and in isolated synaptosomes. The present study shed new light in the understanding of Aβ-membrane interactions in Aβ-induced neurotoxicity. It demonstrates the relevance of Aβ sequence in (i) Aβ-membrane interaction, underlining the role of age-dependent enhanced GM1 content in promoting Aβ aggregation, (ii) Aβ aggregation, and (iii) Aβ-induced oxidative stress. Our results open the way for the design of peptides aimed to inhibit Aβ aggregation and neurotoxicity.
Collapse
|
42
|
Piccinni A, Origlia N, Veltri A, Vizzaccaro C, Marazziti D, Catena-Dell'osso M, Conversano C, Moroni I, Domenici L, Dell'osso L. Plasma β-amyloid peptides levels: a pilot study in bipolar depressed patients. J Affect Disord 2012; 138:160-4. [PMID: 22310032 DOI: 10.1016/j.jad.2011.12.042] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2011] [Revised: 12/21/2011] [Accepted: 12/22/2011] [Indexed: 10/14/2022]
Abstract
BACKGROUND Patients with mood disorders present a great risk for dementia and generally for cognitive decline. Low levels of β-amyloid peptide 1-42 (Aβ42) and high Aβ40/Aβ42 ratio have been associated with this risk and have been reported also in geriatric patients suffering from depression. The aim of the present study was to compare the plasma levels of Aβ40 and Aβ42 in patients with bipolar depression and healthy subjects, and to correlate them with the characteristics of clinical course. METHODS Levels of Aβ40 and Aβ42 were measured by using specific ELISA kits in 16 patients with bipolar depression and in 16 control subjects with a negative history for somatic, psychiatric, neurological and substance abuse disorders. RESULTS Patients presented significantly lower plasma Aβ42 levels and higher Aβ40/Aβ42 ratio, as compared with control subjects. Moreover, a significant negative correlation was found between Aβ42 plasma levels and the duration of the illness, while a positive correlation was detected between the Aβ40/Aβ42 ratio and the number of affective episodes. LIMITATIONS The major limitations of the study are the small sample size, the scanty characterization of the illness episodes and the fact that all the patients were under psychopharmacological treatment. CONCLUSION Although further research is necessary to establish firm conclusions, the present data would suggest that changes in plasma levels of different Aβ peptides might represent a useful tool to identify the risk for cognitive decline in bipolar patients.
Collapse
Affiliation(s)
- Armando Piccinni
- Department of Psychiatry, Neurobiology, Pharmacology and Biotechnologies, University of Pisa, Via Roma 67, 56126 Pisa, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
The Complexity of Sporadic Alzheimer's Disease Pathogenesis: The Role of RAGE as Therapeutic Target to Promote Neuroprotection by Inhibiting Neurovascular Dysfunction. Int J Alzheimers Dis 2012; 2012:734956. [PMID: 22482078 PMCID: PMC3310161 DOI: 10.1155/2012/734956] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 12/02/2011] [Indexed: 01/08/2023] Open
Abstract
Alzheimer's disease (AD) is the most common cause of dementia. Amyloid plaques and neurofibrillary tangles are prominent pathological features of AD. Aging and age-dependent oxidative stress are the major nongenetic risk factors for AD. The beta-amyloid peptide (Aβ), the major component of plaques, and advanced glycation end products (AGEs) are key activators of plaque-associated cellular dysfunction. Aβ and AGEs bind to the receptor for AGEs (RAGE), which transmits the signal from RAGE via redox-sensitive pathways to nuclear factor kappa-B (NF-κB). RAGE-mediated signaling is an important contributor to neurodegeneration in AD. We will summarize the current knowledge and ongoing studies on RAGE function in AD. We will also present evidence for a novel pathway induced by RAGE in AD, which leads to the expression of thioredoxin interacting protein (TXNIP), providing further evidence that pharmacological inhibition of RAGE will promote neuroprotection by blocking neurovascular dysfunction in AD.
Collapse
|
44
|
Yang W, Wu Q, Yuan C, Gao J, Xiao M, Gu M, Ding J, Hu G. Aquaporin-4 mediates astrocyte response to β-amyloid. Mol Cell Neurosci 2012; 49:406-14. [PMID: 22365952 DOI: 10.1016/j.mcn.2012.02.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2011] [Revised: 01/17/2012] [Accepted: 02/06/2012] [Indexed: 12/31/2022] Open
Abstract
It has been demonstrated that the water channel protein aquaporin-4 (AQP4) plays an important role in astrocyte plasticity in response to a variety of injuries or stimuli. However, the potential role of AQP4 in astrocyte response to β-amyloid (Aβ) has not been studied. The purpose of this study was to investigate this issue. Compared to media control, the lower concentrations of Aβ(1-42) (0.1-1 μM) increased AQP4 expression in cultured mouse cortical astrocytes, while the higher concentrations of Aβ(1-42) (10 μM) decreased AQP4 expression. AQP4 gene knockout reduced Aβ(1-42)-induced astrocyte activation and apoptosis, which was associated with a reduction in the uptake of Aβ via decreased upregulation of low-density lipoprotein receptor related protein-1. Moreover, time-course and levels of Aβ(1-42)-induced mitogen-activated protein kinase phosphorylation were altered in AQP4 null astrocytes compared with wild-type controls. Our data reveal a novel role of AQP4 in the uptake of Aβ by astrocytes, indicating that AQP4 is a molecular target for Alzheimer's disease.
Collapse
Affiliation(s)
- Wei Yang
- Jiangsu Key Laboratory of Neurodegeneration, Nanjing Medical University, Nanjing, Jiangsu Province 210029, PR China
| | | | | | | | | | | | | | | |
Collapse
|
45
|
Askarova S, Yang X, Sheng W, Sun GY, Lee JCM. Role of Aβ-receptor for advanced glycation endproducts interaction in oxidative stress and cytosolic phospholipase A₂ activation in astrocytes and cerebral endothelial cells. Neuroscience 2011; 199:375-85. [PMID: 21978883 DOI: 10.1016/j.neuroscience.2011.09.038] [Citation(s) in RCA: 73] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Revised: 08/23/2011] [Accepted: 09/14/2011] [Indexed: 12/22/2022]
Abstract
Blood-brain barrier (BBB) dysfunctions have been implicated in the progression of Alzheimer's disease. Cerebral endothelial cells (CECs) and astrocytes are the main cell components of the BBB. Although amyloid-β oligomers (Aβ₄₂) have been reported to mediate oxidative damage to the CECs and astrocytes and trigger the downstream mitogen-activated protein kinase/extracellular signal-regulated kinase (MAPK/ERK) pathway, the cell surface binding site for Aβ₄₂ and exact sequence of these events have yet to be elucidated. In this study, the receptor for advanced glycation endproducts (RAGE) was postulated to function as a signal transducing cell surface receptor for Aβ₄₂ to induce reactive oxygen species (ROS) generation from NADPH oxidase and trigger downstream pathways for the phosphorylation of extracellular signal-regulated kinases (ERK1/2) and cytosolic phospholipase A₂ (cPLA₂). We found that Aβ₄₂ competed with the anti-RAGE antibody (Ab(RAGE)) to bind to RAGE on the surfaces of CECs and primary astrocytes. In addition, Ab(RAGE) abrogate Aβ₄₂-induced ROS production and the colocalization between the cytosolic (p47-phox) and membrane (gp91-phox) subunits of NADPH oxidase in both cell types. Ab(RAGE) as well as NADPH oxidase inhibitor and ROS scavenger suppressed Aβ₄₂-induced ERK1/2 and cPLA₂ phosphorylation in CECs. At the same time, only Ab(RAGE), but neither NADPH oxidase inhibitor nor ROS scavenger, inhibited the ERK1/2 pathway and cPLA₂ phosphorylation in primary astrocytes. Therefore, this study demonstrates that NADPH oxidase complex assembly and ROS production are not required for Aβ₄₂ binding to RAGE at astrocytic surface leading to sequential phosphorylation of ERK1/2 and cPLA₂, and suggests the presence of two different RAGE-dependent downstream pathways in the CECs and astrocytes.
Collapse
Affiliation(s)
- S Askarova
- Department of Biological Engineering, University of Missouri, Columbia, Missouri 65211, USA
| | | | | | | | | |
Collapse
|
46
|
Ablation of cellular prion protein does not ameliorate abnormal neural network activity or cognitive dysfunction in the J20 line of human amyloid precursor protein transgenic mice. J Neurosci 2011; 31:10427-31. [PMID: 21775587 DOI: 10.1523/jneurosci.1459-11.2011] [Citation(s) in RCA: 100] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Previous studies suggested that the cellular prion protein (PrP(c)) plays a critical role in the pathogenesis of Alzheimer's disease (AD). Specifically, amyloid-β (Aβ) oligomers were proposed to cause synaptic and cognitive dysfunction by binding to PrP(c). To test this hypothesis, we crossed human amyloid precursor protein (hAPP) transgenic mice from line J20 onto a PrP(c)-deficient background. Ablation of PrP(c) did not prevent the premature mortality and abnormal neural network activity typically seen in hAPPJ20 mice. Furthermore, hAPPJ20 mice with or without PrP(c) expression showed comparably robust abnormalities in learning and memory and in other behavioral domains at 6-8 months of age. Notably, these abnormalities are not refractory to therapeutic manipulations in general: they can be effectively prevented by interventions that prevent Aβ-dependent neuronal dysfunction also in other lines of hAPP transgenic mice. Thus, at least in this model, PrP(c) is not an important mediator of Aβ-induced neurological impairments.
Collapse
|
47
|
|
48
|
Cuevas E, Lantz SM, Tobón-Velasco JC, Newport GD, Wu Q, Virmani A, Ali SF, Santamaría A. On the in vivo early toxic properties of A-beta 25-35 peptide in the rat hippocampus: involvement of the Receptor-for-Advanced Glycation-End-Products and changes in gene expression. Neurotoxicol Teratol 2011; 33:288-96. [PMID: 21216281 DOI: 10.1016/j.ntt.2010.12.002] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2010] [Revised: 12/23/2010] [Accepted: 12/23/2010] [Indexed: 11/19/2022]
Abstract
Amyloid-beta peptide (Aβ) deposition is assumed to play a pathogenic role in the brain of Alzheimer's disease patients. To date, the precise mechanisms underlying Aβ toxicity are not fully understood. A recent hypothesis suggesting that the Receptor-for-Advanced-Glycation-End-Products (RAGE)-a trans-membrane protein signaling for oxidative stress-is involved in Aβ toxicity is gaining attention. Early Aβ toxicity could indeed help to explain the deleterious events further produced by this molecule in the brain. In this work, we evaluated the pattern of early expression of RAGE in the toxic model induced by Aß₂₅₋₃₅ in rat CA1 region. Intrahippocampal injections of Aβ₂₅₋₃₅ in rats increased the RAGE expression at 24 h post-injection; this event was accompanied by increased components of RAGE downstream signaling in hippocampal cells, such as enhanced expression of the pro-apoptotic factor NF-κB, increased nitric oxide production, LDH leakage, mitochondrial dysfunction, increased TNF-α expression, antioxidant genes down-regulation, and augmented neurodegeneration. Our findings support an active role of RAGE during the early stages of Aβ₂₅₋₃₅ toxicity in the hippocampus.
Collapse
Affiliation(s)
- Elvis Cuevas
- Neurochemistry Laboratory, Division of Neurotoxicology, National Center for Toxicological Research, Jefferson, AR 72079, USA
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Palop JJ, Mucke L. Amyloid-beta-induced neuronal dysfunction in Alzheimer's disease: from synapses toward neural networks. Nat Neurosci 2010; 13:812-8. [PMID: 20581818 DOI: 10.1038/nn.2583] [Citation(s) in RCA: 1252] [Impact Index Per Article: 83.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease is the most frequent neurodegenerative disorder and the most common cause of dementia in the elderly. Diverse lines of evidence suggest that amyloid-beta (Abeta) peptides have a causal role in its pathogenesis, but the underlying mechanisms remain uncertain. Here we discuss recent evidence that Abeta may be part of a mechanism controlling synaptic activity, acting as a positive regulator presynaptically and a negative regulator postsynaptically. The pathological accumulation of oligomeric Abeta assemblies depresses excitatory transmission at the synaptic level, but also triggers aberrant patterns of neuronal circuit activity and epileptiform discharges at the network level. Abeta-induced dysfunction of inhibitory interneurons likely increases synchrony among excitatory principal cells and contributes to the destabilization of neuronal networks. Strategies that block these Abeta effects may prevent cognitive decline in Alzheimer's disease. Potential obstacles and next steps toward this goal are discussed.
Collapse
Affiliation(s)
- Jorge J Palop
- Gladstone Institute of Neurological Disease and Department of Neurology, University of California, San Francisco, California, USA.
| | | |
Collapse
|