1
|
Zhang YP, Li YY, Zhang C, Li YJ, Liu BP, Zhang Y, Lin JD, Song C. Interleukin-10 Attenuates Behavioral, Immune and Neurotrophin Changes Induced by Chronic Central Administration of Interleukin-1β in Rats. Neuroimmunomodulation 2022; 29:380-390. [PMID: 35316814 DOI: 10.1159/000521710] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 12/20/2021] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Activated microglia can trigger pro-inflammatory cytokine releases and neuroinflammation, which may inhibit astrocytes to produce neurotrophins and anti-inflammatory factors. Both eventually lead to neuron apoptosis or death. Furthermore, effective antidepressant or anti-dementia treatments can reduce pro-inflammatory cytokines, while enhance interleukin (IL)-10 production. However, the underline mechanism by which IL-10 modulates glial cell function, hence improves cognitive impairment or depression-like behavior is unknown. This study evaluated whether and how IL-10 attenuated chronic IL-1β administration-induced behavioral changes and the possible involved mechanisms. METHODS Rats received intracerebroventricular injection of IL-1β and/or IL-10 for 14 days. Then animal memory and depression-like behavior, pro-inflammatory cytokines, glial activities, expression of brain-derived neurotrophic factor (BDNF), Trk B, p75, and apoptosis-related genes were studied. RESULTS Compared to controls, significantly increased latent time and swimming distance in the Morris-water-maze, decreased sucrose consumption, and decreased locomotor and center zone entries in the open-field were found in rats administrated with IL-1β. These changes were associated with the reduction of GFAP expression, and concentrations of BDNF and anti-inflammatory cytokine IL-10, but the increase in the expressions of CD11b, TrkB, p75, and Caspase-3, the ratio of Bax/Bcl-2, and the concentrations of IL-1β, tumor necrosis factor-α, and IL-6. IL-10 treatment markedly attenuated IL-1β-induced above changes, except for the expressions of neurotrophin receptors. CONCLUSION IL-10-improved behavioral changes may be through suppressing microglia activity and inflammation, while restoring astrocyte function and BDNF expression.
Collapse
Affiliation(s)
- Yong-Ping Zhang
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Marine Medicine Research and Development Center of Shenzhen Institutes, Guangdong Ocean University, Shenzhen, China
| | - Yu-Yu Li
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Cai Zhang
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Ya-Jun Li
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Bai-Ping Liu
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Yan Zhang
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
| | - Ju-Da Lin
- Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Cai Song
- Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang, China
- Marine Medicine Research and Development Center of Shenzhen Institutes, Guangdong Ocean University, Shenzhen, China
| |
Collapse
|
2
|
Anwar H, Rasul A, Iqbal J, Ahmad N, Imran A, Malik SA, Ijaz F, Akram R, Maqbool J, Sajid F, Sun T, Hussain G, Manzoor MF. Dietary biomolecules as promising regenerative agents for peripheral nerve injury: An emerging nutraceutical-based therapeutic approach. J Food Biochem 2021; 45:e13989. [PMID: 34719796 DOI: 10.1111/jfbc.13989] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/29/2021] [Accepted: 10/10/2021] [Indexed: 12/22/2022]
Abstract
Peripheral nerve damage is a debilitating condition that can result in partial or complete functional loss as a result of axonal degeneration, as well as lifelong dependence. Many therapies have been imbued with a plethora of positive features while posing little risks. It is worth noting that these biomolecules work by activating several intrinsic pathways that are known to be important in peripheral nerve regeneration. Although the underlying mechanism is used for accurate and speedy functional recovery, none of them are without side effects. As a result, it is believed that effective therapy is currently lacking. The dietary biomolecules-based intervention, among other ways, is appealing, safe, and effective. Upregulation of transcription factors, neurotrophic factors, and growth factors such as NGF, GDNF, BDNF, and CTNF may occur as a result of these substances' dietary intake. Upregulation of the signaling pathways ERK, JNK, p38, and PKA has also been seen, which aids in axonal regeneration. Although several mechanistic approaches to understanding their involvement have been suggested, more work is needed to reveal the amazing properties of these biomolecules. We have discussed in this article that how different dietary biomolecules can help with functional recovery and regeneration after an injury. PRACTICAL APPLICATIONS: Based on the information known to date, we may conclude that treatment techniques for peripheral nerve injury have downsides, such as complications, donor shortages, adverse effects, unaffordability, and a lack of precision in efficacy. These difficulties cast doubt on their efficacy and raise severe concerns about the prescription. In this situation, the need for safe and effective therapeutic techniques is unavoidable, and dietary biomolecules appear to be a safe, cost-efficient, and effective way to promote nerve regeneration following an injury. The information on these biomolecules has been summarized here. Upregulation of transcription factors, neurotrophic factors, and growth factors, such as NGF, GDNF, BDNF, and CTNF, as well as the ERK, JNK, p38, and PKA, signaling pathways, may stimulate axonal regeneration.
Collapse
Affiliation(s)
- Haseeb Anwar
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Javed Iqbal
- Department of Neurology, Allied Hospital, Faisalabad Medical University, Faisalabad, Pakistan
| | - Nazir Ahmad
- Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Ali Imran
- Institute of Home and Food Sciences, Government College University, Faisalabad, Pakistan
| | - Shoaib Ahmad Malik
- Department of Biochemistry, Sargodha Medical College, University of Sargodha, Sargodha, Pakistan
| | - Fazeela Ijaz
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Rabia Akram
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Javeria Maqbool
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Faiqa Sajid
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Tao Sun
- Center for Precision Medicine, School of Medicine and School of Biomedical Sciences, Huaqiao University, Xiamen, China
| | - Ghulam Hussain
- Neurochemicalbiology and Genetics Laboratory (NGL), Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | | |
Collapse
|
3
|
Huin C, Cronier S, Guégan P, Béringue V, Rezaei H, Noinville S. Conformation-dependent membrane permeabilization by neurotoxic PrP oligomers: The role of the H2H3 oligomerization domain. Arch Biochem Biophys 2020; 692:108517. [PMID: 32738196 DOI: 10.1016/j.abb.2020.108517] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/08/2020] [Accepted: 07/26/2020] [Indexed: 12/11/2022]
Abstract
The relationship between prion propagation and the generation of neurotoxic species and clinical onset remains unclear. Several converging lines of evidence suggest that interactions with lipids promote various precursors to form aggregation-prone states that are involved in amyloid fibrils. Here, we compared the cytotoxicities of different soluble isolated oligomeric constructs from murine full-length PrP and from the restricted helical H2H3 domain with their effects on lipid vesicles. The helical H2H3 domain is suggested to be the minimal region of PrP involved in the oligomerization process. The discrete PrP oligomers of both the full-length sequence and the H2H3 domain have de novo β-sheeted structure when interacting with the membrane. They were shown to permeabilize synthetic negatively charged vesicles in a dose-dependent manner. Restricting the polymerization domain of the full-length PrP to the H2H3 helices strongly diminished the ability of the corresponding oligomers to associate with the lipid vesicles. Furthermore, the membrane impairment mechanism occurs differently for the full-length PrP oligomers and the H2H3 helices, as shown by dye-release and black lipid membrane experiments. The membrane damage caused by the full-length PrP oligomers is correlated to their neuronal toxicity at submicromolar concentrations, as shown by cell culture assays. Although oligomers of synthetic H2H3 could compromise in vitro cell homeostasis, they followed a membrane-disruptive pattern that was different from the full-length oligomers, as revealed by the role of PrPC in cell viability assays.
Collapse
Affiliation(s)
- Cécile Huin
- Sorbonne Universités, CNRS, Institut Parisien de Chimie Moléculaire, Equipe Chimie des Polymères, 4 Place Jussieu, F-75005, Paris, France; University of Evry, F-91025, Evry, France
| | - Sabrina Cronier
- UR892, Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - Philippe Guégan
- Sorbonne Universités, CNRS, Institut Parisien de Chimie Moléculaire, Equipe Chimie des Polymères, 4 Place Jussieu, F-75005, Paris, France
| | - Vincent Béringue
- UR892, Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - Human Rezaei
- UR892, Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France
| | - Sylvie Noinville
- UR892, Virologie et Immunologie Moléculaires, Institut National de la Recherche Agronomique (INRA), Jouy-en-Josas, France; Sorbonne Universités, UPMC Univ Paris 06, CNRS, UMR8233, MONARIS, Université Pierre et Marie Curie, Paris, France.
| |
Collapse
|
4
|
Corrêa-Velloso JC, Gonçalves MC, Naaldijk Y, Oliveira-Giacomelli Á, Pillat MM, Ulrich H. Pathophysiology in the comorbidity of Bipolar Disorder and Alzheimer's Disease: pharmacological and stem cell approaches. Prog Neuropsychopharmacol Biol Psychiatry 2018; 80:34-53. [PMID: 28476640 DOI: 10.1016/j.pnpbp.2017.04.033] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 04/28/2017] [Indexed: 12/22/2022]
Abstract
Neuropsychiatric disorders involve various pathological mechanisms, resulting in neurodegeneration and brain atrophy. Neurodevelopmental processes have shown to be critical for the progression of those disorders, which are based on genetic and epigenetic mechanisms as well as on extrinsic factors. We review here common mechanisms underlying the comorbidity of Bipolar Disorders and Alzheimer's Disease, such as aberrant neurogenesis and neurotoxicity, reporting current therapeutic approaches. The understanding of these mechanisms precedes stem cell-based strategies as a new therapeutic possibility for treatment and prevention of Bipolar and Alzheimer's Disease progression. Taking into account the difficulty of studying the molecular basis of disease progression directly in patients, we also discuss the importance of stem cells for effective drug screening, modeling and treating psychiatric diseases, once in vitro differentiation of patient-induced pluripotent stem cells provides relevant information about embryonic origins, intracellular pathways and molecular mechanisms.
Collapse
Affiliation(s)
- Juliana C Corrêa-Velloso
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Maria Cb Gonçalves
- Departamento de Neurologia e Neurociências, Escola Paulista de Medicina, Universidade Federal de São Paulo, Rua Pedro de Toledo 669, São Paulo, SP 04039-032, Brazil
| | - Yahaira Naaldijk
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Ágatha Oliveira-Giacomelli
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Micheli M Pillat
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Av. Prof. Lineu Prestes 748, São Paulo, SP 05508-000, Brazil.
| |
Collapse
|
5
|
Zhang Y, Jiao G, Song C, Gu S, Brown RE, Zhang J, Zhang P, Gagnon J, Locke S, Stefanova R, Pelletier C, Zhang Y, Lu H. An Extract from Shrimp Processing By-Products Protects SH-SY5Y Cells from Neurotoxicity Induced by Aβ 25-35. Mar Drugs 2017; 15:md15030083. [PMID: 28327516 PMCID: PMC5367040 DOI: 10.3390/md15030083] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/07/2017] [Accepted: 03/15/2017] [Indexed: 12/14/2022] Open
Abstract
Increased evidence suggests that marine unsaturated fatty acids (FAs) can protect neurons from amyloid-β (Aβ)-induced neurodegeneration. Nuclear magnetic resonance (NMR), high performance liquid chromatography (HPLC) and gas chromatography (GC) assays showed that the acetone extract 4-2A obtained from shrimp Pandalus borealis industry processing wastes contained 67.19% monounsaturated FAs and 16.84% polyunsaturated FAs. The present study evaluated the anti-oxidative and anti-inflammatory effects of 4-2A in Aβ25–35-insulted differentiated SH-SY5Y cells. Cell viability and cytotoxicity were measured by using 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and lactate dehydrogenase (LDH) assays. Quantitative PCR and Western blotting were used to study the expression of neurotrophins, pro-inflammatory cytokines and apoptosis-related genes. Administration of 20 μM Aβ25–35 significantly reduced SH-SY5Y cell viability, the expression of nerve growth factor (NGF) and its tyrosine kinase TrkA receptor, as well as the level of glutathione, while increased reactive oxygen species (ROS), nitric oxide, tumor necrosis factor (TNF)-α, brain derived neurotrophic factor (BDNF) and its TrkB receptor. Aβ25–35 also increased the Bax/Bcl-2 ratio and Caspase-3 expression. Treatment with 4-2A significantly attenuated the Aβ25–35-induced changes in cell viability, ROS, GSH, NGF, TrkA, TNF-α, the Bax/Bcl-2 ratio and Caspase-3, except for nitric oxide, BDNF and TrKB. In conclusion, 4-2A effectively protected SH-SY5Y cells against Aβ-induced neuronal apoptosis/death by suppressing inflammation and oxidative stress and up-regulating NGF and TrKA expression.
Collapse
Affiliation(s)
- Yongping Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Guangling Jiao
- Coastal Zones Research Institute Inc., 232B, avenue de l'Église, Shippagan, NB E8S 1J2, Canada.
- Aquatic and Crop Resource Development, National Research Council of Canada, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Cai Song
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
- Graduate Institute of Neural and Cognitive Sciences, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Shelly Gu
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Richard E Brown
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Junzeng Zhang
- Aquatic and Crop Resource Development, National Research Council of Canada, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Pingcheng Zhang
- Department of Psychology and Neuroscience, Dalhousie University, Halifax, NS B3H 4R2, Canada.
| | - Jacques Gagnon
- Coastal Zones Research Institute Inc., 232B, avenue de l'Église, Shippagan, NB E8S 1J2, Canada.
| | - Steven Locke
- Aquatic and Crop Resource Development, National Research Council of Canada, 550 University Avenue, Charlottetown, PE C1A 4P3, Canada.
| | - Roumiana Stefanova
- Aquatic and Crop Resource Development, National Research Council of Canada, 1411 Oxford Street, Halifax, NS B3H 3Z1, Canada.
| | - Claude Pelletier
- Coastal Zones Research Institute Inc., 232B, avenue de l'Église, Shippagan, NB E8S 1J2, Canada.
| | - Yi Zhang
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| | - Hongyu Lu
- Research Institute for Marine Drugs and Nutrition, College of Food Science and Technology, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
6
|
Psychosocial stress on neuroinflammation and cognitive dysfunctions in Alzheimer's disease: the emerging role for microglia? Neurosci Biobehav Rev 2017; 77:148-164. [PMID: 28185874 DOI: 10.1016/j.neubiorev.2017.01.046] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 01/20/2017] [Accepted: 01/31/2017] [Indexed: 01/22/2023]
Abstract
Chronic psychosocial stress is increasingly recognized as a risk factor for late-onset Alzheimer's disease (LOAD) and associated cognitive deficits. Chronic stress also primes microglia and induces inflammatory responses in the adult brain, thereby compromising synapse-supportive roles of microglia and deteriorating cognitive functions during aging. Substantial evidence demonstrates that failure of microglia to clear abnormally accumulating amyloid-beta (Aβ) peptide contributes to neuroinflammation and neurodegeneration in AD. Moreover, genome-wide association studies have linked variants in several immune genes, such as TREM2 and CD33, the expression of which in the brain is restricted to microglia, with cognitive dysfunctions in LOAD. Thus, inflammation-promoting chronic stress may create a vicious cycle of aggravated microglial dysfunction accompanied by increased Aβ accumulation, collectively exacerbating neurodegeneration. Surprisingly, however, little is known about whether and how chronic stress contributes to microglia-mediated neuroinflammation that may underlie cognitive impairments in AD. This review aims to summarize the currently available clinical and preclinical data and outline potential molecular mechanisms linking stress, microglia and neurodegeneration, to foster future research in this field.
Collapse
|
7
|
Zhu D, Yang N, Liu YY, Zheng J, Ji C, Zuo PP. M2 Macrophage Transplantation Ameliorates Cognitive Dysfunction in Amyloid-β-Treated Rats Through Regulation of Microglial Polarization. J Alzheimers Dis 2017; 52:483-95. [PMID: 27003214 DOI: 10.3233/jad-151090] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly population. Neuroinflammation induced by amyloid-β (Aβ) aggregation is considered to be the critical factor underlying AD pathological mechanisms. Alternatively activated (M2) macrophages/microglia have been reported to have neuroprotective effects in neurodegenerative disease. In this study, we characterized the neuroprotective effects of M2 macrophage transplantation in AD model rats and investigated the underlying mechanisms. Intracerebroventricular injection of Aβ1 - 42 to rats was used to model AD and resulted in cognitive impairment, neuronal damage, and inflammatory changes in the brain microenvironment. We observed an increased interferon regulatory factor (IRF) 5/IRF4 ratio, resulting in greater production of classically activated (M1) versus M2 microglia. M2 macrophage transplantation attenuated inflammation in the brain, reversed Aβ1 - 42-induced changes in the IRF4-IRF5 ratio, drove endogenous microglial polarization toward the M2 phenotype, and ameliorated cognitive impairment. Nerve growth factor (NGF) treatment reduced the IRF5/IRF4 ratio and induced primary microglial polarization to the M2 phenotype in vitro; these effects were prevented by tyrosine Kinase Receptor A (TrkA) inhibition. M2 macrophage transplantation restored the balance of IRF4-IRF5 by affecting the expression of NGF and inflammatory cytokines in the brains of AD model rats. This drove microglial polarization to the M2 phenotype, promoted termination of neuroinflammation, and resulted in improved cognitive abilities.
Collapse
|
8
|
de Miranda AS, Brant F, Vieira LB, Rocha NP, Vieira ÉLM, Rezende GHS, de Oliveira Pimentel PM, Moraes MFD, Ribeiro FM, Ransohoff RM, Teixeira MM, Machado FS, Rachid MA, Teixeira AL. A Neuroprotective Effect of the Glutamate Receptor Antagonist MK801 on Long-Term Cognitive and Behavioral Outcomes Secondary to Experimental Cerebral Malaria. Mol Neurobiol 2016; 54:7063-7082. [PMID: 27796746 DOI: 10.1007/s12035-016-0226-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2016] [Accepted: 10/13/2016] [Indexed: 02/06/2023]
Abstract
Cerebral malaria (CM) is a life-threatening complication of Plasmodium falciparum infection, which can result in long-term cognitive and behavioral deficits despite successful anti-malarial therapy. Due to the substantial social and economic burden of CM, the development of adjuvant therapies is a scientific goal of highest priority. Apart from vascular and immune responses, changes in glutamate system have been reported in CM pathogenesis suggesting a potential therapeutic target. Based on that, we hypothesized that interventions in the glutamatergic system induced by blockage of N-methyl-D-aspartate (NMDA) receptors could attenuate experimental CM long-term cognitive and behavioral outcomes. Before the development of evident CM signs, susceptible mice infected with Plasmodium berghei ANKA (PbA) strain were initiated on treatment with dizocilpine maleate (MK801, 0.5 mg/kg), a noncompetitive NMDA receptor antagonist. On day 5 post-infection, mice were treated orally with a 10-day course chloroquine (CQ, 30 mg/kg). Control mice also received saline, CQ or MK801 + CQ therapy. After 10 days of cessation of CQ treatment, magnetic resonance images (MRI), behavioral and immunological assays were performed. Indeed, MK801 combined with CQ prevented long-term memory impairment and depressive-like behavior following successful PbA infection resolution. In addition, MK801 also modulated the immune system by promoting a balance of TH1/TH2 response and upregulating neurotrophic factors levels in the frontal cortex and hippocampus. Moreover, hippocampus abnormalities observed by MRI were partially prevented by MK801 treatment. Our results indicate that NMDA receptor antagonists can be neuroprotective in CM and could be a valuable adjuvant strategy for the management of the long-term impairment observed in CM.
Collapse
Affiliation(s)
- Aline Silva de Miranda
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Laboratory of Neurobiology, Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil. .,Departamento de Morfologia, ICB, UFMG, Av. Antônio Carlos, 6627, Pampulha, Belo Horizonte, 31270-901, Brazil.
| | - Fátima Brant
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Luciene Bruno Vieira
- Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Natália Pessoa Rocha
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Pharmacology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Érica Leandro Marciano Vieira
- Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Gustavo Henrique Souza Rezende
- Department of Physiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Marcio F D Moraes
- Department of Physiology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabíola Mara Ribeiro
- Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | | | - Mauro Martins Teixeira
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Fabiana Simão Machado
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Milene Alvarenga Rachid
- Department of Pathology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| | - Antônio Lúcio Teixeira
- Postgraduate Program in Health Sciences: Infectious Diseases and Tropical Medicine, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Interdisciplinary Laboratory of Medical Investigation, School of Medicine, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil.,Department of Biochemistry and Immunology, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte, MG, Brazil
| |
Collapse
|
9
|
Eugenín J, Vecchiola A, Murgas P, Arroyo P, Cornejo F, von Bernhardi R. Expression Pattern of Scavenger Receptors and Amyloid-β Phagocytosis of Astrocytes and Microglia in Culture are Modified by Acidosis: Implications for Alzheimer’s Disease. J Alzheimers Dis 2016; 53:857-73. [DOI: 10.3233/jad-160083] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Affiliation(s)
- Jaime Eugenín
- Laboratory of Neural Systems, Department of Biology, Faculty of Chemistry and Biology, Universidad de Santiago de Chile (USACH), Santiago, Chile
| | - Andrea Vecchiola
- Laboratory of Neuroscience, Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
- Department of Endocrinology, Faculty of Medicine, Pontificia Universidad Catolica de Chile, Santiago, Chile
| | - Paola Murgas
- Laboratory of Neuroscience, Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Pablo Arroyo
- Laboratory of Neuroscience, Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Francisca Cornejo
- Laboratory of Neuroscience, Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Rommy von Bernhardi
- Laboratory of Neuroscience, Department of Neurology, Faculty of Medicine, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
10
|
Liang W, Zhao X, Feng J, Song F, Pan Y. Ursolic acid attenuates beta-amyloid-induced memory impairment in mice. ARQUIVOS DE NEURO-PSIQUIATRIA 2016; 74:482-8. [PMID: 27332074 DOI: 10.1590/0004-282x20160065] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/05/2015] [Accepted: 01/26/2016] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Increasing evidence demonstrates that oxidative stress and inflammatory are involved in amyloid β (Aβ)-induced memory impairments. Ursolic acid (UA), a triterpenoid compound, has potent anti-inflammatory and antioxidant activities. However, it remains unclear whether UA attenuates Aβ-induced neurotoxicity. METHOD The aggregated Aβ25-35 was intracerebroventricularly administered to mice. RESULTS We found that UA significantly reversed the Aβ25-35-induced learning and memory deficits. Our results indicated that one of the potential mechanisms of the neuroprotective effect was attenuating the Aβ25-35-induced accumulation of malondialdehyde (MDA) and depletion of glutathione (GSH) in the hippocampus. Furthermore, UA significantly suppressed the upregulation of IL-1β, IL-6, and tumor necrosis-α factor levels in the hippocampus of Aβ25-35-treated mice. CONCLUSION These findings suggest that UA prevents memory impairment through amelioration of oxidative stress, inflammatory response and may offer a novel therapeutic strategy for the treatment of Alzheimer's disease.
Collapse
Affiliation(s)
- Wenna Liang
- The Third People's Hospital, Liaocheng, Department of Neurology, Shandong , China, The Third People's Hospital of Liaocheng, Department of Neurology, Shandong, China
| | - Xiaoyang Zhao
- The Third People's Hospital, Liaocheng, Department of Neurology, Shandong , China, The Third People's Hospital of Liaocheng, Department of Neurology, Shandong, China
| | - Jinping Feng
- The Third People's Hospital, Liaocheng, Department of Neurology, Shandong , China, The Third People's Hospital of Liaocheng, Department of Neurology, Shandong, China
| | - Fenghua Song
- The Third People's Hospital, Liaocheng, Department of Pharmacy, Shandong , China, The Third People's Hospital of Liaocheng, Department of Pharmacy, Shandong, China
| | - Yunzhi Pan
- The Third Affiliated Hospital, Qiqihar Medical University, Department of Neurology, Heilong Jiang , China, The Third Affiliated Hospital of Qiqihar Medical University, Department of Neurology, Heilong Jiang, China
| |
Collapse
|
11
|
Budni J, Bellettini-Santos T, Mina F, Garcez ML, Zugno AI. The involvement of BDNF, NGF and GDNF in aging and Alzheimer's disease. Aging Dis 2015; 6:331-41. [PMID: 26425388 DOI: 10.14336/ad.2015.0825] [Citation(s) in RCA: 263] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/25/2015] [Indexed: 12/21/2022] Open
Abstract
Aging is a normal physiological process accompanied by cognitive decline. This aging process has been the primary risk factor for development of aging-related diseases such as Alzheimer's disease (AD). Cognitive deficit is related to alterations of neurotrophic factors level such as brain-derived neurotrophic factor (BDNF), nerve growth factor (NGF) and glial cell-derived neurotrophic factor (GDNF). These strong relationship between aging and AD is important to investigate the time which they overlap, as well as, the pathophysiological mechanism in each event. Considering that aging and AD are related to cognitive impairment, here we discuss the involving these neurotrophic factors in the aging process and AD.
Collapse
Affiliation(s)
- Josiane Budni
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Tatiani Bellettini-Santos
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Francielle Mina
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Michelle Lima Garcez
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| | - Alexandra Ioppi Zugno
- Laboratório de Neurociências, Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde, Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
| |
Collapse
|
12
|
Liu J, Ma Y, Tian S, Zhang L, Zhao M, Zhang Y, Xu D. T cells promote the regeneration of neural precursor cells in the hippocampus of Alzheimer's disease mice. Neural Regen Res 2014; 9:1541-7. [PMID: 25317172 PMCID: PMC4192972 DOI: 10.4103/1673-5374.139481] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2014] [Indexed: 01/07/2023] Open
Abstract
Alzheimer's disease is closely associated with disorders of neurogenesis in the brain, and growing evidence supports the involvement of immunological mechanisms in the development of the disease. However, at present, the role of T cells in neuronal regeneration in the brain is unknown. We injected amyloid-beta 1–42 peptide into the hippocampus of six BALB/c wild-type mice and six BALB/c-nude mice with T-cell immunodeficiency to establish an animal model of Alzheimer's disease. A further six mice of each genotype were injected with same volume of normal saline. Immunohistochemistry revealed that the number of regenerated neural progenitor cells in the hippocampus of BALB/c wild-type mice was significantly higher than that in BALB/c-nude mice. Quantitative fluorescence PCR assay showed that the expression levels of peripheral T cell-associated cytokines (interleukin-2, interferon-γ) and hippocampal microglia-related cytokines (interleukin-1β, tumor necrosis factor-α) correlated with the number of regenerated neural progenitor cells in the hippocampus. These results indicate that T cells promote hippocampal neurogenesis in Alzheimer's disease and T-cell immunodeficiency restricts neuronal regeneration in the hippocampus. The mechanism underlying the promotion of neuronal regeneration by T cells is mediated by an increased expression of peripheral T cells and central microglial cytokines in Alzheimer's disease mice. Our findings provide an experimental basis for understanding the role of T cells in Alzheimer's disease.
Collapse
Affiliation(s)
- Jing Liu
- Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China ; Department of Human Anatomy, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Yuxin Ma
- Department of Human Anatomy, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Sumin Tian
- Department of Human Anatomy, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Li Zhang
- Department of Human Anatomy, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Mengmeng Zhao
- Department of Human Anatomy, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Yaqiong Zhang
- Department of Human Anatomy, School of Basic Courses, Guangdong Pharmaceutical University, Guangzhou, Guangdong Province, China
| | - Dachuan Xu
- Department of Human Anatomy, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
13
|
Compound danshen tablet ameliorated aβ25-35-induced spatial memory impairment in mice via rescuing imbalance between cytokines and neurotrophins. Altern Ther Health Med 2014; 14:23. [PMID: 24422705 PMCID: PMC3898400 DOI: 10.1186/1472-6882-14-23] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 01/08/2014] [Indexed: 12/18/2022]
Abstract
Background Compound Danshen Tablet (CDT), a Traditional Chinese Medicine, has recently been reported to improve spatial cognition in a rat model of Alzheimer’s disease. However, in vivo neuroprotective mechanism of the CDT in models of spatial memory impairment is not yet evaluated. The present study is aimed to elucidate the cellular mechanism of CDT on Aβ25-35-induced cognitive impairment in mice. Methods Mice were randomly divided into 5 groups: the control group (sham operated), the Aβ25-35 treated group, the positive drug group, and large and small dosage of the CDT groups, respectively. CDT was administered at a dose of 0.81 g/kg and 0.405 g/kg for 3 weeks. The mice in the positive drug group were treated with 0.4 mg/kg of Huperzine A, whereas the mice of the control and Aβ25-35 treated groups were administrated orally with equivalent saline. After 7 days of preventive treatment, mice were subjected to lateral ventricle injection of Aβ25-35 to establish the mice model of Alzheimer’s disease. Spatial memory impairment was evaluated by Morris water maze test. Choline acetyltransferase (ChAT) contents in hippocampus and cortex were quantified by ELISA. The levels of cytokines, receptor of activated protein kinase C1 (RACK1) and brain-derived neurotrophic factor (BDNF) in hippocampus were measured by RT-PCR and ELISA. Results The results showed that Aβ25-35 caused spatial memory impairment as demonstrated by performance in the Morris water maze test. CDT was able to confer a significant improvement in spatial memory, and protect mice from Aβ25-35-induced neurotoxicity. Additionally, CDT also inhibited the increase of TNF-α and IL-6 level, and increased the expression of choline acetyltransferase (ChAT), receptor of activated protein kinase C1 (RACK1) and brain-derived neurotrophic factor (BDNF) in brain as compared to model mice. Conclusion These findings strongly implicate that CDT may be a useful treatment against learning and memory deficits in mice by rescuing imbalance between cytokines and neurotrophins.
Collapse
|
14
|
Hayley S, Litteljohn D. Neuroplasticity and the next wave of antidepressant strategies. Front Cell Neurosci 2013; 7:218. [PMID: 24312008 PMCID: PMC3834236 DOI: 10.3389/fncel.2013.00218] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2013] [Accepted: 10/29/2013] [Indexed: 12/13/2022] Open
Abstract
Depression is a common chronic psychiatric disorder that is also often co-morbid with numerous neurological and immune diseases. Accumulating evidence indicates that disturbances of neuroplasticity occur with depression, including reductions of hippocampal neurogenesis and cortical synaptogenesis. Improper trophic support stemming from stressor-induced reductions of growth factors, most notably brain derived neurotrophic factor (BDNF), likely drives such aberrant neuroplasticity. We posit that psychological and immune stressors can interact upon a vulnerable genetic background to promote depression by disturbing BDNF and neuroplastic processes. Furthermore, the chronic and commonly relapsing nature of depression is suggested to stem from "faulty wiring" of emotional circuits driven by neuroplastic aberrations. The present review considers depression in such terms and attempts to integrate the available evidence indicating that the efficacy of current and "next wave" antidepressant treatments, whether used alone or in combination, is at least partially tied to their ability to modulate neuroplasticity. We particularly focus on the N-methyl-D-aspartate (NMDA) antagonist, ketamine, which already has well documented rapid antidepressant effects, and the trophic cytokine, erythropoietin (EPO), which we propose as a potential adjunctive antidepressant agent.
Collapse
Affiliation(s)
- Shawn Hayley
- Department of Neuroscience, Carleton University Ottawa, ON, Canada
| | | |
Collapse
|
15
|
Mengel D, Röskam S, Neff F, Balakrishnan K, Deuster O, Gold M, Oertel WH, Bacher M, Bach JP, Dodel R. Naturally occurring autoantibodies interfere with β-amyloid metabolism and improve cognition in a transgenic mouse model of Alzheimer's disease 24 h after single treatment. Transl Psychiatry 2013; 3:e236. [PMID: 23462987 PMCID: PMC3625912 DOI: 10.1038/tp.2012.151] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
There is evidence that naturally occurring antibodies directed against Aβ (nAbs-Aβ) have a role in Aβ-metabolism and Aβ-clearance. The presence of nAbs-Aβ leads to a reduction in amyloid fibrillation and thus a reduction in their toxicity. We investigated the effects of nAbs-Aβ in respect to oligomerization and used the Tg2576 transgenic mouse model in order to investigate the rapid effect with a single-dose (24 h) on oligomer breakdown and cytokine secretion along with immunohistochemical characterization of synaptic plasticity. nAbs-Aβ were able to reduce toxic oligomer concentration with an increase in Aβ-monomers. Cytokine secretion was significantly reduced. Synaptic plasticity was also improved after administration of nAbs. Finally, single treatment lead to a significant improvement in cognition. This study demonstrates the efficacy of nAbs-Aβ and presents evidence that several hallmarks of the disease are targeted by nAbs-Aβ.
Collapse
Affiliation(s)
- D Mengel
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - S Röskam
- Department of Neurology, Philipps-University Marburg, Marburg, Germany,Department of Animal Physiology, Philipps-University, Marburg, Germany
| | - F Neff
- Department of Neurology, Philipps-University Marburg, Marburg, Germany,Institute of Pathology, Helmholtz Zentrum, Deutsches Forschungszentrum für Gesundheit und Umwelt (GmbH), München, Germany
| | - K Balakrishnan
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - O Deuster
- Department of Neurology, Philipps-University Marburg, Marburg, Germany,IZKS-Mainz, University Medical Center, Mainz, Germany
| | - M Gold
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - W H Oertel
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - M Bacher
- Department of Neurology, Philipps-University Marburg, Marburg, Germany,Institute of Immunology, Philipps-University, Marburg, Germany
| | - J-P Bach
- Department of Neurology, Philipps-University Marburg, Marburg, Germany
| | - R Dodel
- Department of Neurology, Philipps-University Marburg, Marburg, Germany,Department of Neurology, Philipps-University Marburg, Baldingerstrasse, 35041 Marburg, Germany. E-mail:
| |
Collapse
|
16
|
Lin LF, Liao MJ, Xue XY, Zhang W, Yan L, Cai L, Zhou XW, Zhou X, Luo HM. Combination of Aβ clearance and neurotrophic factors as a potential treatment for Alzheimer's disease. Neurosci Bull 2013; 29:111-20. [PMID: 23179066 PMCID: PMC5561854 DOI: 10.1007/s12264-012-1287-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2012] [Accepted: 06/12/2012] [Indexed: 12/31/2022] Open
Abstract
There is no effective drug to treat Alzheimer's disease (AD), a neurodegenerative disease affecting an estimated 30 million people around the world. Strongly supported by preclinical and clinical studies, amyloid-beta (Aβ) may be a target for developing drugs against AD. Meanwhile, the fact that localized neuronal death/loss and synaptic impairment occur in AD should also be considered. Neuronal regeneration, which does not occur normally in the mammalian central nervous system, can be promoted by neurotrophic factors (NTFs). Evidence from clinical trials has shown that both Aβ clearance and NTFs are potentially effective in treating AD, thus a new approach combining Aβ clearance and administration of NTFs may be an effective therapeutic strategy.
Collapse
Affiliation(s)
- Lian-Feng Lin
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Min-Jing Liao
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
- Department of Laboratory Medicine, Medical College of Hunan Normal University, Changsha, 410013 China
| | - Xiao-Yan Xue
- Ganzhou People’s Hospital, Ganzhou, 341000 China
| | - Wei Zhang
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Li Yan
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Liang Cai
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Xiao-Wen Zhou
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Xing Zhou
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
| | - Huan-Min Luo
- Department of Pharmacology, Jinan University, Guangzhou, 510632 China
- Institute of Brain Sciences, School of Medicine, Jinan University, Guangzhou, 510632 China
- The Joint Laboratory of Brain Function and Health, Jinan University and The University of Hong Kong, Jinan University, Guangzhou, 510632 China
| |
Collapse
|
17
|
Neurotoxicity and memory deficits induced by soluble low-molecular-weight amyloid-β1-42 oligomers are revealed in vivo by using a novel animal model. J Neurosci 2012; 32:7852-61. [PMID: 22674261 DOI: 10.1523/jneurosci.5901-11.2012] [Citation(s) in RCA: 133] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Neuronal and synaptic degeneration are the best pathological correlates for memory decline in Alzheimer's disease (AD). Although the accumulation of soluble low-molecular-weight amyloid-β (Aβ) oligomers has been suggested to trigger neurodegeneration in AD, animal models overexpressing or infused with Aβ lack neuronal loss at the onset of memory deficits. Using a novel in vivo approach, we found that repeated hippocampal injections of small soluble Aβ(1-42) oligomers in awake, freely moving mice were able to induce marked neuronal loss, tau hyperphosphorylation, and deficits in hippocampus-dependent memory. The neurotoxicity of small Aβ(1-42) species was observed in vivo as well as in vitro in association with increased caspase-3 activity and reduced levels of the NMDA receptor subunit NR2B. We found that the sequestering agent transthyretin is able to bind the toxic Aβ(1-42) species and attenuated the loss of neurons and memory deficits. Our novel mouse model provides evidence that small, soluble Aβ(1-42) oligomers are able to induce extensive neuronal loss in vivo and initiate a cascade of events that mimic the key neuropathological hallmarks of AD.
Collapse
|
18
|
Bagheri M, Roghani M, Joghataei MT, Mohseni S. Genistein inhibits aggregation of exogenous amyloid-beta1–40 and alleviates astrogliosis in the hippocampus of rats. Brain Res 2012; 1429:145-54. [DOI: 10.1016/j.brainres.2011.10.020] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2011] [Revised: 09/28/2011] [Accepted: 10/10/2011] [Indexed: 11/17/2022]
|
19
|
Shindo T, Takasaki K, Uchida K, Onimura R, Kubota K, Uchida N, Irie K, Katsurabayashi S, Mishima K, Nishimura R, Fujiwara M, Iwasaki K. Ameliorative Effects of Telmisartan on the Inflammatory Response and Impaired Spatial Memory in a Rat Model of Alzheimer’s Disease Incorporating Additional Cerebrovascular Disease Factors. Biol Pharm Bull 2012. [DOI: 10.1248/bpb.b12-00387] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Taro Shindo
- Department of Psychiatry, Faculty of Medicine, Fukuoka University
| | - Kotaro Takasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | - Kanako Uchida
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | - Rika Onimura
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | - Kaori Kubota
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | - Naoki Uchida
- Department of Psychiatry, Faculty of Medicine, Fukuoka University
| | - Keiichi Irie
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | | | - Kenichi Mishima
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | - Ryoji Nishimura
- Department of Psychiatry, Faculty of Medicine, Fukuoka University
| | - Michihiro Fujiwara
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| | - Katsunori Iwasaki
- Department of Neuropharmacology, Faculty of Pharmaceutical Science, Fukuoka University
| |
Collapse
|