1
|
Harris BN, Yavari M, Ramalingam L, Mounce PL, Alers Maldonado K, Chavira AC, Thomas S, Scoggin S, Biltz C, Moustaid-Moussa N. Impact of Long-Term Dietary High Fat and Eicosapentaenoic Acid on Behavior and Hypothalamic-Pituitary-Adrenal Axis Activity in Amyloidogenic APPswe/PSEN1dE9 Mice. Neuroendocrinology 2024; 114:553-576. [PMID: 38301617 PMCID: PMC11153005 DOI: 10.1159/000536586] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 01/30/2024] [Indexed: 02/03/2024]
Abstract
INTRODUCTION Alzheimer's disease (AD) alters neurocognitive and emotional function and causes dysregulation of multiple homeostatic processes. The leading AD framework pins amyloid beta plaques and tau tangles as primary drivers of dysfunction. However, many additional variables, including diet, stress, sex, age, and pain tolerance, interact in ways that are not fully understood to impact the onset and progression of AD pathophysiology. We asked: (1) does high-fat diet, compared to low-fat diet, exacerbate AD pathophysiology and behavioral decline? And, (2) can supplementation with eicosapentaenoic (EPA)-enriched fish oil prevent high-fat-diet-induced changes? METHODS Male and female APPswePSdE9 mice, and their non-transgenic littermates, were randomly assigned to a diet condition (low-fat, high-fat, high-fat with EPA) and followed from 2 to 10 months of age. We assessed baseline corticosterone concentration during aging, pain tolerance, cognitive function, stress coping, and corticosterone response to a stressor. RESULTS Transgenic mice were consistently more active than non-transgenic mice but did not perform worse on either cognitive task, even though we recently reported that these same transgenic mice exhibited metabolic changes and had increased amyloid beta. Mice fed high-fat diet had higher baseline and post-stressor corticosterone, but diet did not impact cognition or pain tolerance. Sex had the biggest influence, as female mice were consistently more active and had higher corticosterone than males. CONCLUSION Overall, diet, genotype, and sex did not have consistent impacts on outcomes. We found little support for predicted interactions and correlations, suggesting diet impacts metabolic function and amyloid beta levels, but these outcomes do not translate to changes in behaviors measured here.
Collapse
Affiliation(s)
- Breanna N. Harris
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
| | - Mahsa Yavari
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
- Current address: Department of Molecular Metabolism, School of Public Health, Harvard University, Boston, MA
| | - Latha Ramalingam
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
- Current address: Department of Nutritional and Food Studies Syracuse University, Syracuse, NY
| | - P. Logan Mounce
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
| | | | - Angela C. Chavira
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Sarah Thomas
- Department of Biological Sciences, Texas Tech University, Lubbock, TX
| | - Shane Scoggin
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Caroline Biltz
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
| | - Naima Moustaid-Moussa
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX
- Obesity Research Institute, Office of Research & Innovation, Texas Tech University
| |
Collapse
|
2
|
Samir SM, Hassan HM, Elmowafy R, ElNashar EM, Alghamdi MA, AlSheikh MH, Al-Zahrani NS, Alasiri FM, Elhadidy MG. Neuroprotective effect of ranolazine improves behavioral discrepancies in a rat model of scopolamine-induced dementia. Front Neurosci 2024; 17:1267675. [PMID: 38323121 PMCID: PMC10845649 DOI: 10.3389/fnins.2023.1267675] [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] [Received: 07/27/2023] [Accepted: 12/15/2023] [Indexed: 02/08/2024] Open
Abstract
Background Ranolazine (Rn), an antianginal agent, acts in the central nervous system and has been used as a potential treatment agent for pain and epileptic disorders. Alzheimer's disease (AD) is one of the most prevalent neurodegenerative diseases and the leading factor in dementia in the elderly. Aim We examined the impact of Rn on scopolamine (Sco)-induced dementia in rats. Methods Thirty-two albino male rats were divided into four groups: control, Rn, Sco, and Rn + Sco. Results A significant decrease in the escape latency in the Morris water maze test after pre-treatment with Rn explained better learning and memory in rats. Additionally, Rn significantly upregulated the activities of the antioxidant enzymes in the treated group compared to the Sco group but substantially reduced acetylcholinesterase activity levels in the hippocampus. Moreover, Rn dramatically reduced interleukin-1 β (IL-1β) and IL-6 and upregulated the gene expression of brain-derived neurotrophic factor (BDNF). Furthermore, in the Sco group, the hippocampal tissue's immunohistochemical reaction of Tau and glial factor activating protein (GFAP) was significantly increased in addition to the upregulation of the Caspase-3 gene expression, which was markedly improved by pre-treatment with Rn. The majority of pyramidal neurons had large vesicular nuclei with prominent nucleoli and appeared to be more or less normal, reflecting the all-beneficial effects of Rn when the hippocampal tissue was examined under a microscope. Conclusion Our findings indicated that Rn, through its antioxidative, anti-inflammatory, and anti-apoptotic effects, as well as the control of the expression of GFAP, BDNF, and Tau proteins, has a novel neuroprotective impact against scopolamine-induced dementia in rats.
Collapse
Affiliation(s)
- Shereen M. Samir
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Hend M. Hassan
- Department of Human Anatomy and Embryology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Rasha Elmowafy
- Department of Medical Biochemistry and Molecular Biology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Eman Mohamed ElNashar
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mansour Abdullah Alghamdi
- Department of Anatomy, College of Medicine, King Khalid University, Abha, Saudi Arabia
- Genomics and Personalized Medicine Unit, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Mona Hmoud AlSheikh
- Department of Physiology, College of Medicine, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Norah Saeed Al-Zahrani
- Department of Clinical Biochemistry, College of Medicine, King Khalid University, Abha, Saudi Arabia
| | - Faten Mohammed Alasiri
- Pharmacist in King Fahad Armed Forces Hospital Khamis Mushait, Khamis Mushait, Saudi Arabia
| | - Mona G. Elhadidy
- Department of Medical Physiology, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Department of Medical Physiology, Faculty of Medicine, Al-Baha University, Al-Baha, Saudi Arabia
| |
Collapse
|
3
|
Huang F, Marungruang N, Martinsson I, Camprubí Ferrer L, Nguyen TD, Gondo TF, Karlsson EN, Deierborg T, Öste R, Heyman-Lindén L. A mixture of Nordic berries improves cognitive function, metabolic function and alters the gut microbiota in C57Bl/6J male mice. Front Nutr 2023; 10:1257472. [PMID: 37854349 PMCID: PMC10580983 DOI: 10.3389/fnut.2023.1257472] [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] [Received: 07/12/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Our diets greatly influence our health. Multiple lines of research highlight the beneficial properties of eating berries and fruits. In this study, a berry mixture of Nordic berries previously identified as having the potential to improve memory was supplemented to young C57Bl/6J male mice to investigate effects on cognition function, metabolic health, markers of neuroinflammation, and gut microbiota composition. C57Bl/6J male mice at the age of 8 weeks were given standard chow, a high-fat diet (HF, 60%E fat), or a high-fat diet supplemented with freeze-dried powder (20% dwb) of a mixture of Nordic berries and red grape juice (HF + Berry) for 18 weeks (n = 12 animals/diet group). The results show that supplementation with the berry mixture may have beneficial effects on spatial memory, as seen by enhanced performance in the T-maze and Barnes maze compared to the mice receiving the high-fat diet without berries. Additionally, berry intake may aid in counteracting high-fat diet induced weight gain and could influence neuroinflammatory status as suggested by the increased levels of the inflammation modifying IL-10 cytokine in hippocampal extracts from berry supplemented mice. Furthermore, the 4.5-month feeding with diet containing berries resulted in significant changes in cecal microbiota composition. Analysis of cecal bacterial 16S rRNA revealed that the chow group had significantly higher microbial diversity, as measured by the Shannon diversity index and total operational taxonomic unit richness, than the HF group. The HF diet supplemented with berries resulted in a strong trend of higher total OTU richness and significantly increased the relative abundance of Akkermansia muciniphila, which has been linked to protective effects on cognitive decline. In conclusion, the results of this study suggest that intake of a Nordic berry mixture is a valuable strategy for maintaining and improving cognitive function, to be further evaluated in clinical trials.
Collapse
Affiliation(s)
- Fang Huang
- Division of Biotechnology, Department of Chemistry, Lund University, Lund, Sweden
- Aventure AB, Lund, Sweden
| | | | - Isak Martinsson
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Lluís Camprubí Ferrer
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Thao Duy Nguyen
- Department of Food Technology, Engineering and Nutrition, Lund University, Lund, Sweden
| | - Thamani Freedom Gondo
- Department of Chemistry, Centre for Analysis and Synthesis, Lund University, Lund, Sweden
| | | | - Tomas Deierborg
- Experimental Neuroinflammation Laboratory, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | | | - Lovisa Heyman-Lindén
- Berry Lab AB, Lund, Sweden
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
4
|
Xu D, Guo Q. miR-26a Improves Microglial Activation and Neuronal Apoptosis in a Rat Model of Cerebral Infarction by Regulating the TREM1-TLR4/MyD88/NF-κB Axis. Dev Neurosci 2023; 46:221-236. [PMID: 37703835 DOI: 10.1159/000533813] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 08/24/2023] [Indexed: 09/15/2023] Open
Abstract
Emerging studies have indicated that abnormally expressed microRNAs (miRNAs) are related to the pathogenesis of cerebral ischemia. Nevertheless, the function of miR-26a in neuronal damage and microglial activation during cerebral infarction remains elusive. It was revealed that miR-26a was downregulated in oxygen-glucose deprivation (OGD)-treated microglia and neurons. Overexpressing miR-26a reduced the inflammatory reaction in BV2 cells and decreased neuronal apoptosis following OGD stimulation. miR-26a upregulation inactivated the TLR4/MyD88/NF-κB pathway and inhibited TREM1 expression. Repressing NF-κB phosphorylation inhibited the miR-26a level. As supported by the dual-luciferase reporter assay, TREM1 was directly targeted by miR-26a. Furthermore, a rat model of middle cerebral artery occlusion (MCAO) was built. We discovered that miR-26a improved cognitive, learning, and motor functions and reduced cerebral edema in MCAO rats. Mechanistically, upregulating miR-26a reduced inflammation and neuronal apoptosis by mitigating the TREM1-TLR4/MyD88/NF-κB pathway in the MCAO rat model. Collectively, this study verified that the miR-26a-TREM1-TLR4/MyD88/NF-κB axis contributes to modulating OGD-mediated microglial activation and neuronal injury.
Collapse
Affiliation(s)
- Daxiong Xu
- Department of Neurology, Bazhong Central Hospital, Bazhong, China
| | - Qi'an Guo
- Department of Anesthesiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China,
| |
Collapse
|
5
|
Steuer EL, Kemper LJ, Hlynialuk CJW, Leinonen-Wright K, Montonye ML, Lapcinski IP, Forster CL, Ashe KH, Liu P. Blocking Site-Specific Cleavage of Human Tau Delays Progression of Disease-Related Phenotypes in Genetically Matched Tau-Transgenic Mice Modeling Frontotemporal Dementia. J Neurosci 2022; 42:4737-4754. [PMID: 35508385 PMCID: PMC9186797 DOI: 10.1523/jneurosci.0543-22.2022] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/22/2022] [Accepted: 04/28/2022] [Indexed: 11/21/2022] Open
Abstract
Studies have recently demonstrated that a caspase-2-mediated cleavage of human tau (htau) at asparate-314 (D314) is responsible for cognitive deficits and neurodegeneration in mice modeling frontotemporal dementia (FTD). However, these animal studies may be confounded by flaws in their model systems, such as endogenous functional gene disruption and inequivalent transgene expression. To avoid these weaknesses, we examined the pathogenic role of this site-specific htau cleavage in FTD using genetically matched htau targeted-insertion mouse lines: rT2 and rT3. Both male and female mice were included in this study. rT2 mice contain a single copy of the FTD-linked htau proline-to-leucine mutation at amino acid 301 (htau P301L), inserted into a neutral site to avoid dysregulation of host gene expression. The similarly constructed rT3 mice harbor an additional D314-to-glutamate (D314E) mutation that blocks htau cleavage. We demonstrate that htau transgene expression occurs primarily in the forebrain at similar levels in rT2 and rT3 mice. Importantly, expression of the cleavage-resistant D314E mutant delays transgene-induced tau accumulation in the postsynaptic density, brain atrophy, hippocampal neurodegeneration, and spatial memory impairment, without altering age-related progression of pathologic tau conformation and phosphorylation. Our comprehensive investigation of age-dependent disease phenotypes associated with the htau P301L variant in precisely engineered FTD-modeling mice unveils a transiently protective effect of blocking htau cleavage at D314. Findings of this study advance our understanding of the contribution of this tau cleavage to the pathogenesis of FTD, and aid the development of effective dementia-targeting therapies.SIGNIFICANCE STATEMENT A site-specific and caspase-2-mediated cleavage of human tau plays a pathologic role in dementia. In this study, we investigate the contribution of this cleavage to the pathogenesis of frontotemporal dementia (FTD) using two genetically matched, tau-transgene targeted-insertion mouse lines that differ only by a cleavage-resistant mutation. The use of these mice avoids confounding effects associated with the random integration of tau transgenes to the mouse genome and allows us to comprehensively evaluate the impact of the tau cleavage on FTD phenotypes. Our data reveal that blocking this tau cleavage delays memory impairment and neurodegeneration of FTD-modeling mice. These findings improve our understanding of the pathogenic mechanisms underlying FTD and will facilitate the development of effective therapeutics.
Collapse
Affiliation(s)
- Elizabeth L Steuer
- N. Bud Grossman Center for Memory Research and Care
- Department of Neurology
| | - Lisa J Kemper
- N. Bud Grossman Center for Memory Research and Care
- Department of Neurology
| | | | | | | | - Ian P Lapcinski
- N. Bud Grossman Center for Memory Research and Care
- Department of Neurology
| | - Colleen L Forster
- N. Bud Grossman Center for Memory Research and Care
- UMN Academic Health Center Biological Materials Procurement Network, University of Minnesota, Minneapolis, Minnesota 55455
| | - Karen H Ashe
- N. Bud Grossman Center for Memory Research and Care
- Department of Neurology
- Geriatric Research, Education, and Clinical Centers, Veterans Affairs Medical Center, Minneapolis, Minnesota 55417
| | - Peng Liu
- N. Bud Grossman Center for Memory Research and Care
- Department of Neurology
| |
Collapse
|
6
|
Spirulina platensis alleviates high fat diet-induced cognitive impairment in mice via the gut-brain axis. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104706] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
7
|
Differential Effects of Human P301L Tau Expression in Young versus Aged Mice. Int J Mol Sci 2021; 22:ijms222111637. [PMID: 34769068 PMCID: PMC8583766 DOI: 10.3390/ijms222111637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 11/17/2022] Open
Abstract
The greatest risk factor for developing Alzheimer’s disease (AD) is increasing age. Understanding the changes that occur in aging that make an aged brain more susceptible to developing AD could result in novel therapeutic targets. In order to better understand these changes, the current study utilized mice harboring a regulatable mutant P301L human tau transgene (rTg(TauP301L)4510), in which P301L tau expression can be turned off or on by the addition or removal of doxycycline in the drinking water. This regulatable expression allowed for assessment of aging independent of prolonged mutant tau expression. Our results suggest that P301L expression in aged mice enhances memory deficits in the Morris water maze task. These behavioral changes may be due to enhanced late-stage tau pathology, as evidenced by immunoblotting and exacerbated hippocampal dysregulation of glutamate release and uptake measured by the microelectrode array technique. We additionally observed changes in proteins important for the regulation of glutamate and tau phosphorylation that may mediate these age-related changes. Thus, age and P301L tau interact to exacerbate tau-induced detrimental alterations in aged animals.
Collapse
|
8
|
An enriched environment prevents cognitive impairment in an Alzheimer's disease model by enhancing the secretion of exosomal microRNA-146a from the choroid plexus. Brain Behav Immun Health 2021; 9:100149. [PMID: 34589894 PMCID: PMC8474441 DOI: 10.1016/j.bbih.2020.100149] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Accepted: 09/16/2020] [Indexed: 02/08/2023] Open
Abstract
Alzheimer’s disease (AD) is characterized by the extensive deposition of amyloid-β plaques and neurofibrillary tangles. We previously found that preserved function of astrocytes is associated with cognitively normal subjects with AD pathology. Here we show that an enriched environment (EE) can prevent cognitive impairment in AD model mice by ameliorating astrocytic inflammation and increasing synaptic density in the subiculum area of the hippocampus. In AD model mice treated with an EE, increased levels of microRNA (miR)-146a and down-regulation of NF-κB were observed in the hippocampus. In addition, increased levels of interferon (IFN)-γ were seen in serum from mice exposed to an EE. In vitro, enhanced miR-146a expression was observed in exosomes derived from the choroid plexus (CP) after IFN-γ treatment. In further in vitro experiments, we transfected miR-146a into Aβ/lipopolysaccharide-induced inflammatory astrocytes and showed that miR-146a ameliorated astrocytic inflammation by down-regulating tumor necrosis factor receptor-associated factor 6 and NF-κB. The present study indicates that following an EE, exosomal miR-146a derived from the CP cells is a key factor in ameliorating astrocytic inflammation, leading to synaptogenesis and correction of cognitive impairment. An enriched environment (EE) prevented the cognitive impairment in 5 × FAD mice. An EE inhibited astrocytic inflammation and increased miR-146a in hippocampus. An EE increased the levels of interferon-γ (IFN-γ) in serum. IFN-γ increased the secretion of exosomal miR-146a from cultured choroid plexus. Transfection of miR-146a down-regulated NF-κB in cultured astrocytes.
Collapse
|
9
|
Brady RD, Bird S, Sun M, Yamakawa GR, Major BP, Mychasiuk R, O'Brien TJ, McDonald SJ, Shultz SR. Activation of the Protein Kinase R-Like Endoplasmic Reticulum Kinase (PERK) Pathway of the Unfolded Protein Response after Experimental Traumatic Brain Injury and Treatment with a PERK Inhibitor. Neurotrauma Rep 2021; 2:330-342. [PMID: 34318301 PMCID: PMC8310749 DOI: 10.1089/neur.2021.0001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Neurodegeneration after traumatic brain injury (TBI) is increasingly recognized as a key factor contributing to poor chronic outcomes. Activation (i.e., phosphorylation) of the protein kinase R-like endoplasmic reticulum kinase (PERK) pathway has been implicated in neurodegenerative conditions with pathological similarities to TBI and may be a potential target to improve TBI outcomes. Here, we aimed to determine whether a moderate TBI would induce activation of the PERK pathway and whether treatment with the PERK inhibitor, GSK2606414, would improve TBI recovery. Male mice were administered a lateral fluid percussion injury (FPI) or sham injury and were euthanized at either 2 h, 24 h, or 1 week post-injury (n = 5 per injury group and time point) to assess changes in the PERK pathway. In the injured cortex, there was increased phosphorylated-PERK at 2 h post-FPI and increased phosphorylation of eukaryotic translation initiation factor α at 24 h post-FPI. We next examined the effect of acute treatment with GSK2606414 on pathological and behavioral outcomes at 4 weeks post-injury. Thus, there were a total of four groups: sham + VEH (n = 9); sham + GSK4606414 (n = 10); FPI + VEH (n = 9); and FPI + GSK2606414 (n = 9). GSK2606414 (50 mg/kg) or vehicle treatment was delivered by oral gavage beginning at 30 min post-injury, followed by two further treatments at 12-h increments. There were no significant effects of GSK2606414 on any of the outcomes assessed, which could be attributable to several reasons. For example, activation of PERK may not be a significant contributor to the neurological consequences 4 weeks post-FPI in mice. Further research is required to elucidate the role of the PERK pathway in TBI and whether interventions that target this pathway are beneficial.
Collapse
Affiliation(s)
- Rhys D Brady
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stefanie Bird
- Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Mujun Sun
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Glenn R Yamakawa
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Brendan P Major
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Richelle Mychasiuk
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Terence J O'Brien
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| | - Stuart J McDonald
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Physiology, Anatomy, and Microbiology, La Trobe University, Bundoora, Victoria, Australia
| | - Sandy R Shultz
- Department of Neuroscience, Central Clinical School, Monash University, Melbourne, Victoria, Australia.,Department of Medicine, The University of Melbourne, Parkville, Victoria, Australia
| |
Collapse
|
10
|
Akbar L, Juliandi B, Boediono A, Batubara I, Subangkit M. Effects of Eugenol on Memory Performance, Neurogenesis, and Dendritic Complexity of Neurons in Mice Analyzed by Behavioral Tests and Golgi Staining of Brain Tissue. J Stem Cells Regen Med 2021; 17:35-41. [PMID: 34434006 PMCID: PMC8372414 DOI: 10.46582/jsrm.1701005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 12/31/2020] [Indexed: 11/19/2022]
Abstract
Eugenol, as the main component in clove, has neuroprotective abilities, including its effect to learning memory of mice. However, there is no evidence showing whether eugenol can expand the growth of dendrites in the brain. The objective of this research was to examine the effects of eugenol towards dendritic complexity of neurons, neurogenesis, and memory performance in hippocampus. A total of 21 mice were divided into three groups; (i) mice were administered 30 mg/kg bw eugenol orally, (ii) mice were administered 100 mg/kg bw eugenol orally, and (iii) mice were administered distilled water as control. Mice were kept for 30 consecutive days following the standard animal housing. The memory performance was observed through the Y-arm maze alternation, Novel Object Recognition (NOR), and Morris Water Maze (MWM) test. The brain was dissected and stained with FD Rapid Golgi StainingTM kit to observe dendrites in the dentate gyrus (DG) and cornu ammonis 1 (CA1) region; and Haematoxylin-Eosin (HE) staining to assess neurogenesis in the DG. Our results showed that eugenol enhanced putative neural stem cells (NPCs) and granular cells (GC) number, and also decrease neuronal cell death in DG (p<0.0001). Eugenol also increased dendritic complexity of neurons in DG region; while in CA1, eugenol has given a positive effect only on the basal area. Eugenol increased spatial and recognition memory in mice, indicated by a higher number of correct alternations and discrimination ratio compared to the control group (p<0.05), although escape latency in MWM did not show significant effect (p>0.05). As analyzed by behavioral tests and Golgi staining of brain tissue, eugenol can increase memory performance, neurogenesis, and dendritic complexity of neurons in the DG and CA1 basal region of brain in mice.
Collapse
Affiliation(s)
- Latiful Akbar
- Graduate Program of Animal Bioscience, Department of Biology, Faculty of Mathematics and Natural Sciences
| | - Berry Juliandi
- Department of Biology, Faculty of Mathematics and Natural Sciences
| | - Arief Boediono
- Department of Anatomy, Physiology, and Pharmacology, Faculty of Veterinary Medicine
| | - Irmanida Batubara
- Department of Chemistry, Faculty of Mathematics and Natural Sciences
- Tropical Biopharmaca Research Center
| | - Mawar Subangkit
- Department of Veterinary Clinic, Reproduction, and Pathology, Faculty of Veterinary Medicine, IPB University (Bogor Agricultural University), Bogor, Indonesia
| |
Collapse
|
11
|
Wang B, Lin X, Zhou J, Xie C, Li C, Dong R, Zhang G, Sun X, Wang M, Bi Y. Insulin-like growth factor-1 improves postoperative cognitive dysfunction following splenectomy in aged rats. Exp Ther Med 2021; 21:215. [PMID: 33574912 PMCID: PMC7818527 DOI: 10.3892/etm.2021.9647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2019] [Accepted: 09/29/2020] [Indexed: 11/15/2022] Open
Abstract
Postoperative cognitive dysfunction (POCD) is a serious complication following anesthesia and operations in aged patients undergoing surgical intervention. It is characterized by temporary or permanent cognitive decline, memory impairment and deterioration in language comprehension and social adaption ability. Therefore, the development of POCD prevention and treatment tools has become an area of interest. The current study assessed the therapeutic effects of insulin-like growth factor-1 (IGF-1) on POCD in aged rats and explored the underlying mechanisms. Model rats underwent splenectomy under 1.5-2% isoflurane and mechanical ventilation. IGF-1 (50 µg/kg) was diluted in normal saline and administered by abdominal hypodermic injection daily from the operation to day 7 post-operation. Following splenectomy, the animals showed marked cognitive impairment as determined by the Morris water maze test. Hippocampal protein levels of amyloid precursor protein (APP), β-site APP-cleaving enzyme-1 (BACE-1), amyloid-β (Aβ), capase3, Bax and Bcl-2 were assessed by immunoblotting. Neuronal apoptosis in the hippocampus was analyzed using a TUNEL assay. The results demonstrated that the levels of APP, BACE-1, Aβ, caspase3 and Bax were increased following splenectomy, while the levels of Bcl2 were reduced at days 1, 3 and 7 post-operation in aged rats. However, IGF-1 downregulated APP, BACE-1, Aβ, capase3 and Bax, and upregulated Bcl2 at these time points following splenectomy. TUNEL staining revealed that administration of IGF-1 significantly reduced neuronal apoptosis in the hippocampal CA1 region following splenectomy. These results indicated that IGF-1 decreased Aβ-protein production and inhibited neuronal apoptosis in the hippocampus following splenectomy, subsequently alleviating POCD.
Collapse
Affiliation(s)
- Bin Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xu Lin
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Jiahui Zhou
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Chunhui Xie
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Chuan Li
- Department of Anesthesiology, Weifang Medical University, Weifang, Shandong 261042, P.R. China
| | - Rui Dong
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Gaofeng Zhang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Xiaopeng Sun
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Mingshan Wang
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| | - Yanlin Bi
- Department of Anesthesiology, Qingdao Municipal Hospital Affiliated to Qingdao University, Qingdao, Shandong 266071, P.R. China
| |
Collapse
|
12
|
Wang A, Zou X, Wu J, Ma Q, Yuan N, Ding F, Li X, Chen J. Early-Life Stress Alters Synaptic Plasticity and mTOR Signaling: Correlation With Anxiety-Like and Cognition-Related Behavior. Front Genet 2021; 11:590068. [PMID: 33381149 PMCID: PMC7767996 DOI: 10.3389/fgene.2020.590068] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 11/24/2020] [Indexed: 12/28/2022] Open
Abstract
Early-life stress (ELS) predisposes individuals to psychiatric disorders, including anxiety and depression, and cognitive impairments later in life. However, the underlying molecular mechanisms are not completely understood. Developmental deficits in hippocampal synaptic plasticity are among the primary detrimental alterations in brain function induced by ELS. Impaired synaptic plasticity is usually accompanied by decreased synaptic proteins, such as postsynaptic density 95 (PSD95) and synaptophysin, which are important for synaptic function. The mTOR signaling pathway plays a vital role in regulating protein translation, and mTOR activation is functionally associated with synaptic protein synthesis. In the present study, we observed whether ELS impacts synaptic protein synthesis and mTOR signaling, which is involved in synaptic plasticity. Herein, we established a maternal separation (MS) and chronic restraint stress (CRS) model and evaluated anxiety-like behavior and cognitive function (e.g., learning and memory) in adulthood through behavioral examination and analyzed hippocampal expression levels of PSD95 and synaptophysin. To explore whether the mTOR signaling pathway was associated with ELS, we also examined the activity of mTOR and s6. The behavior tests indicated that maternally separated mice showed increased anxiety-like behavior and cognitive impairments. PSD95 and synaptophysin mRNA and protein expression levels were decreased in the hippocampus, and phosphorylated mTOR and phosphorylated s6 were significantly decreased in maternally separated mice vs. those not exposed to MS. Our data demonstrate that MS impairs synaptic plasticity and inhibits mTOR signaling, specifically via s6. Therefore, we speculate that ELS decreased synaptic plasticity via the inhibition of the mTOR pathway in the hippocampus, which may underlie vulnerability to stress and mental disorders in adulthood.
Collapse
Affiliation(s)
- Anfeng Wang
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaojuan Zou
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan, China
| | - Jiajia Wu
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan, China
| | - Qingyu Ma
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Naijun Yuan
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Fengmin Ding
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan, China
| | - Xiaojuan Li
- Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China
| | - Jiaxu Chen
- School of Basic Medical Science, Hubei University of Chinese Medicine, Wuhan, China.,Formula-Pattern Research Center, School of Traditional Chinese Medicine, Jinan University, Guangzhou, China.,School of Traditional Chinese Medicine, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
13
|
Chataigner M, Mortessagne P, Lucas C, Pallet V, Layé S, Mehaignerie A, Bouvret E, Dinel AL, Joffre C. Dietary fish hydrolysate supplementation containing n-3 LC-PUFAs and peptides prevents short-term memory and stress response deficits in aged mice. Brain Behav Immun 2021; 91:716-730. [PMID: 32976934 DOI: 10.1016/j.bbi.2020.09.022] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 12/12/2022] Open
Abstract
Brain aging is characterized by a decline in cognitive functions, which can lead to the development of neurodegenerative pathologies. Age-related spatial learning and memory deficits are associated with a chronic low-grade inflammation. Anxiety disorders and stress response alterations, occurring for a part of the elderly, have also been linked to an increased neuroinflammation and thus, an accelerated cognitive decline. Nutrition is an innovative strategy to prevent age-related cognitive impairments. Among the nutrients, n-3 long chain polyunsaturated fatty acids (LC-PUFAs) and low molecular weight peptides from proteins, especially those from marine resources, are good candidates for their immunomodulatory, anxiolytic and neuroprotective properties. The aim of this study is to determine the combined effect of n-3 LC-PUFAs and low molecular weight peptides on cognitive functions, and their mechanism of action. We are the first to show that a dietary supplementation with a fish hydrolysate containing n-3 LC-PUFAs and low molecular weight peptides prevented the age-related spatial short-term memory deficits and modulated navigation strategies adopted during spatial learning. In addition, the fish hydrolysate displayed anxiolytic activities with the reduction of anxiety-like behaviour in aged mice, restored the plasmatic corticosterone levels similar to adult animals following an acute stress and modulated the hypothalamic stress response. These effects on behaviour can be explained by the immunomodulatory and neuroprotective properties of the fish hydrolysate that limited microgliosis in vivo, decreased LPS-induced expression of pro-inflammatory cytokines and increased the expression of growth factors such as BDNF and NGF in vitro. Thus, n-3 LC-PUFAs and low molecular weight peptides contained in the fish hydrolysate can play an important role in the limitation of neuroinflammation and stress response alterations during aging and represent a potential strategy for the prevention of age-related cognitive decline.
Collapse
Affiliation(s)
- M Chataigner
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France; Abyss Ingredients, 56850 Caudan, France
| | - P Mortessagne
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - C Lucas
- NutriBrain Research and Technology Transfer, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - V Pallet
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - S Layé
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France
| | | | - E Bouvret
- Abyss Ingredients, 56850 Caudan, France
| | - A L Dinel
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France; NutriBrain Research and Technology Transfer, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France
| | - C Joffre
- Université de Bordeaux, INRAE, Bordeaux INP, NutriNeuro, 146 rue Léo Saignat, 33076 Bordeaux, France.
| |
Collapse
|
14
|
Direct inhibition of Keap1-Nrf2 Protein-Protein interaction as a potential therapeutic strategy for Alzheimer's disease. Bioorg Chem 2020; 103:104172. [DOI: 10.1016/j.bioorg.2020.104172] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 07/24/2020] [Accepted: 08/02/2020] [Indexed: 12/11/2022]
|
15
|
Long-term probiotic intervention mitigates memory dysfunction through a novel H3K27me3-based mechanism in lead-exposed rats. Transl Psychiatry 2020; 10:25. [PMID: 32066679 PMCID: PMC7026181 DOI: 10.1038/s41398-020-0719-8] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Revised: 12/07/2019] [Accepted: 12/20/2019] [Indexed: 02/06/2023] Open
Abstract
Chronic lead exposure is associated with the development of neurodegenerative diseases, characterized by the long-term memory decline. However, whether this pathogenesis could be prevented through adjusting gut microbiota is not yet understood. To address the issue, pregnant rats and their female offspring were treated with lead (125 ppm) or separately the extra probiotics (1010 organisms/rat/day) till adulthood. For results, memory dysfunction was alleviated by the treatment of multispecies probiotics. Meanwhile, the gut microbiota composition was partially normalized against lead-exposed rats, which in turn mediated the memory repairment via fecal transplantation trials. In the molecular aspect, the decreased H3K27me3 (trimethylation of histone H3 Lys 27) in the adult hippocampus was restored with probiotic intervention, an epigenetic event mediated by EZH2 (enhancer of zeste homolog 2) at early developmental stage. In a neural cellular model, EZH2 overexpression showed the similar rescue effect with probiotics, whereas its blockade led to the neural re-damages. Regarding the gut-brain inflammatory mediators, the disrupted IL-6 (interleukin 6) expression was resumed by probiotic treatment. Intraperitoneal injection of tocilizumab, an IL-6 receptor antagonist, upregulated the hippocampal EZH2 level and consequently alleviated the memory injuries. In conclusion, reshaping gut microbiota could mitigate memory dysfunction caused by chronic lead exposure, wherein the inflammation-hippocampal epigenetic pathway of IL-6-EZH2-H3K27me3, was first proposed to mediate the studied gut-brain communication. These findings provided insight with epigenetic mechanisms underlying a unique gut-brain interaction, shedding light on the safe and non-invasive treatment of neurodegenerative disorders with environmental etiology.
Collapse
|
16
|
RNF34 overexpression exacerbates neurological deficits and brain injury in a mouse model of intracerebral hemorrhage by potentiating mitochondrial dysfunction-mediated oxidative stress. Sci Rep 2019; 9:16296. [PMID: 31704983 PMCID: PMC6841714 DOI: 10.1038/s41598-019-52494-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 10/07/2019] [Indexed: 12/27/2022] Open
Abstract
Intracerebral hemorrhage (ICH) is a common neurological condition associated with high disability and mortality. Alterations in protein ubiquitination have emerged as a key mechanism in the pathogenesis of neurological diseases. Here, we investigated the effects of the E3 ubiquitin ligase ring finger protein 34 (RNF34) on neurological deficits and brain injury in ICH mice. An ICH model was established via intracerebral injection of autologous blood into wild-type and RNF34 transgenic mice. Brain injury, neurological function, neuronal activity, and oxidative stress levels were measured, respectively. The underlying mechanisms were explored by molecular and cellular approaches. Our results showed that RNF34 overexpression in mice significantly aggravated the ICH-induced memory impairment, brain edema, infarction, hematoma volume, and loss of neuronal activity. RNF34 and oxidative stress levels gradually increased from 6 to 48 h after the ICH challenge and were positively correlated. The ICH-induced increase in intracellular ROS, superoxide anion, and mROS generation and the decrease in adenosine triphosphate production were exacerbated in RNF34 transgenic mice, but NADPH oxidase activity was unaffected. Moreover, RNF34 upregulation potentiated the ICH-induced decrease in PGC-1α, UCP2, and MnSOD expressions. RNF34 interacted with PGC-1α and targeted it for ubiquitin-dependent degradation. This study reveals that RNF34 exacerbates neurological deficits and brain injury by facilitating PGC-1α protein degradation and promoting mitochondrial dysfunction-mediated oxidative stress.
Collapse
|
17
|
Developmental Pathogenicity of 4-Repeat Human Tau Is Lost with the P301L Mutation in Genetically Matched Tau-Transgenic Mice. J Neurosci 2019; 40:220-236. [PMID: 31685653 DOI: 10.1523/jneurosci.1256-19.2019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 10/16/2019] [Accepted: 10/20/2019] [Indexed: 12/25/2022] Open
Abstract
Tau is a microtubule-associated protein that becomes dysregulated in a group of neurodegenerative diseases called tauopathies. Differential tau isoforms, expression levels, promoters, and disruption of endogenous genes in transgenic mouse models of tauopathy make it difficult to draw definitive conclusions about the biological role of tau in these models. We addressed this shortcoming by characterizing the molecular and cognitive phenotypes associated with the pathogenic P301L tau mutation (rT2 mice) in relation to a genetically matched transgenic mouse overexpressing nonmutant (NM) 4-repeat (4R) human tau (rT1 mice). Both male and female mice were included in this study. Unexpectedly, we found that 4R NM human tau (hTau) exhibited abnormal dynamics in young mice that were lost with the P301L mutation, including elevated protein stability and hyperphosphorylation, which were associated with cognitive impairment in 5-month-old rT1 mice. Hyperphosphorylation of NM hTau was observed as early as 4 weeks of age, and transgene suppression for the first 4 or 12 weeks of life prevented abnormal molecular and cognitive phenotypes in rT1, demonstrating that NM hTau pathogenicity is specific to postnatal development. We also show that NM hTau exhibits stronger binding to microtubules than P301L hTau, and is associated with mitochondrial abnormalities. Overall, our genetically matched mice have revealed that 4R NM hTau overexpression is pathogenic in a manner distinct from classical aging-related tauopathy, underlining the importance of assaying the effects of transgenic disease-related proteins at appropriate stages in life.SIGNIFICANCE STATEMENT Due to differences in creation of transgenic lines, the pathological properties of the P301L mutation confers to the tau protein in vivo have remained elusive, perhaps contributing to the lack of disease-modifying therapies for tauopathies. In an attempt to characterize P301L-specific effects on tau biology and cognition in novel genetically matched transgenic mouse models, we surprisingly found that nonmutant human tau has development-specific pathogenic properties of its own. Our findings indicate that overexpression of 4-repeat human tau during postnatal development is associated with excessive microtubule binding, which may disrupt important cellular processes, such as mitochondrial dynamics, leading to elevated stability and hyperphosphorylation of tau, and eventual cognitive impairments.
Collapse
|
18
|
Frame AK, Lone A, Harris RA, Cumming RC. Simple Protocol for Distinguishing Drug-induced Effects on Spatial Memory Acquisition, Consolidation and Retrieval in Mice Using the Morris Water Maze. Bio Protoc 2019; 9:e3376. [PMID: 33654872 DOI: 10.21769/bioprotoc.3376] [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: 06/11/2019] [Revised: 08/13/2019] [Accepted: 08/19/2019] [Indexed: 11/02/2022] Open
Abstract
The Morris water maze (MWM) is one of the most commonly used tests for assessing spatial learning and memory in mice. While the MWM is highly amenable to testing the effects of memory modifying drugs, most studies do not consider the timing or duration of drug exposure when conducting the MWM assay; factors that can strongly influence the effect of the drug on different stages of memory and interfere with data interpretation. Herein we describe a MWM protocol which offers the advantage of distinguishing the impact of a fast acting intraperitoneally (IP) injected drug on the different stages of spatial memory: acquisition, consolidation, and retrieval. Mice initially undergo habituation to both the MWM apparatus and IP injection procedure over the course of three days. For assessing the effect of a drug on memory acquisition, mice are injected with the drug prior to training sessions over four consecutive days, where mice learn to find an escape platform in a circular water tank using distal spatial cues. To determine the effect of the drug on memory consolidation, mice are injected with the drug immediately after each training session. For testing the effect of a drug on memory retrieval, mice receive mock IP injections on each training day and the drug is IP injected only once, prior to a probe trial, where mice attempt to locate the platform following its removal from the tank. This protocol provides a simple strategy for distinguishing the effect(s) of a CNS acting drug on the different stages of memory.
Collapse
Affiliation(s)
- Ariel K Frame
- Department of Biology, Western University, London, Ontario N6A 5B7, Canada
| | - Asad Lone
- Department of Biology, Western University, London, Ontario N6A 5B7, Canada
| | - Richard A Harris
- Department of Biology, Western University, London, Ontario N6A 5B7, Canada
| | - Robert C Cumming
- Department of Biology, Western University, London, Ontario N6A 5B7, Canada
| |
Collapse
|
19
|
Sankowski R, Huerta TS, Kalra R, Klein TJ, Strohl JJ, Al-Abed Y, Robbiati S, Huerta PT. Large-Scale Validation of the Paddling Pool Task in the Clockmaze for Studying Hippocampus-Based Spatial Cognition in Mice. Front Behav Neurosci 2019; 13:121. [PMID: 31231197 PMCID: PMC6568215 DOI: 10.3389/fnbeh.2019.00121] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 05/20/2019] [Indexed: 11/30/2022] Open
Abstract
Rationally designed behavioral tests are important tools to assess the function of specific brain regions. The hippocampus is a crucial neural substrate for spatial cognition, and many studies have linked hippocampal dysfunction with defects on spatial learning and memory in neurological conditions ranging from Alzheimer's disease to autoimmune syndromes, such as neuropsychiatric lupus. While our understanding of hippocampal function, from the molecular to the system levels, has increased dramatically over the last decades, this effort has not yet translated into efficacious therapies for cognitive impairment. We think that the availability of highly validated behavioral paradigms to measure cognition in mouse models is likely to enhance the potential success of preclinical therapeutic modalities. Here, we present an extensive study of the paddling pool task (PPT), first reported by Deacon and Rawlins, in which mice learn to escape from shallow water through a peripheral exit in a circular arena dubbed the clockmaze. We show that the PPT provides highly reliable results when assaying spatial cognition in C57/BL6 mice (120 males, 40 females) and BALB/c mice (40 males, 90 females). Additionally, we develop a robust algorithm for the assessment of escape strategies with clearly quantifiable readouts, enabling fine-granular phenotyping. Notably, the use of spatial strategy increases linearly across trials in the PPT. In a separate cohort of mice, we apply muscimol injections to silence the dorsal CA1 region of the hippocampus and show that the use of the spatial strategy in the PPT relies on the integrity of the dorsal hippocampus. Additionally, we compare directly the PPT and the Morris water maze (MWM) task in C57/BL6 mice (20 males, 20 females) and BALB/c mice (20 males, 20 females) and we find that the PPT induces significantly lower anxiety, exhaustion and hypothermia than the MWM. We conclude that the PPT provides a robust assessment of spatial cognition in mice, which can be applied in conjunction with other tests, to facilitate hypothesis testing and drug development to combat cognitive impairment.
Collapse
Affiliation(s)
- Roman Sankowski
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Faculty of Medicine, Institute of Neuropathology, University of Freiburg, Freiburg, Germany
| | - Tomás S. Huerta
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Rishi Kalra
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Toby J. Klein
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Joshua J. Strohl
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Yousef Al-Abed
- Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| | - Sergio Robbiati
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
| | - Patricio T. Huerta
- Laboratory of Immune & Neural Networks, Institute of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Institute of Bioelectronic Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Elmezzi Graduate School of Molecular Medicine, Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY, United States
- Department of Molecular Medicine, Zucker School of Medicine at Hofstra/Northwell, Manhasset, NY, United States
| |
Collapse
|
20
|
Swarup V, Hinz FI, Rexach JE, Noguchi KI, Toyoshiba H, Oda A, Hirai K, Sarkar A, Seyfried NT, Cheng C, Haggarty SJ, Grossman M, Van Deerlin VM, Trojanowski JQ, Lah JJ, Levey AI, Kondou S, Geschwind DH. Identification of evolutionarily conserved gene networks mediating neurodegenerative dementia. Nat Med 2019; 25:152-164. [PMID: 30510257 PMCID: PMC6602064 DOI: 10.1038/s41591-018-0223-3] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 09/18/2018] [Indexed: 02/02/2023]
Abstract
Identifying the mechanisms through which genetic risk causes dementia is an imperative for new therapeutic development. Here, we apply a multistage, systems biology approach to elucidate the disease mechanisms in frontotemporal dementia. We identify two gene coexpression modules that are preserved in mice harboring mutations in MAPT, GRN and other dementia mutations on diverse genetic backgrounds. We bridge the species divide via integration with proteomic and transcriptomic data from the human brain to identify evolutionarily conserved, disease-relevant networks. We find that overexpression of miR-203, a hub of a putative regulatory microRNA (miRNA) module, recapitulates mRNA coexpression patterns associated with disease state and induces neuronal cell death, establishing this miRNA as a regulator of neurodegeneration. Using a database of drug-mediated gene expression changes, we identify small molecules that can normalize the disease-associated modules and validate this experimentally. Our results highlight the utility of an integrative, cross-species network approach to drug discovery.
Collapse
Affiliation(s)
- Vivek Swarup
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Co-first author
| | - Flora I. Hinz
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Co-first author
| | - Jessica E. Rexach
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Ken-ichi Noguchi
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Hiroyoshi Toyoshiba
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Akira Oda
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Keisuke Hirai
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Arjun Sarkar
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Nicholas T. Seyfried
- Department of Biochemistry, Emory University School of Medicine, Atlanta, GA,Alzheimer’s Disease Research Center and Department of Neurology, Emory University School of Medicine, Atlanta, GA
| | - Chialin Cheng
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - IFGC
- International FTD-Genomics Consortium, a list of members and affiliations appears at the end of the paper
| | - Murray Grossman
- Center for Neurodegenerative Disease Research, Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Vivianna M. Van Deerlin
- The Penn FTD Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - John Q. Trojanowski
- The Penn FTD Center, Department of Neurology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - James J. Lah
- Alzheimer’s Disease Research Center and Department of Neurology, Emory University School of Medicine, Atlanta, GA
| | - Allan I. Levey
- Alzheimer’s Disease Research Center and Department of Neurology, Emory University School of Medicine, Atlanta, GA
| | - Shinichi Kondou
- CNS Drug Discovery Unit, Pharmaceutical Research Division, Takeda Pharmaceutical Company Limited, Fujisawa, Kanagawa 251-8555, Japan
| | - Daniel H. Geschwind
- Program in Neurogenetics, Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA,Institute of Precision Health, University of California, Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
21
|
Input-Specific Synaptic Location and Function of the α5 GABA A Receptor Subunit in the Mouse CA1 Hippocampal Neurons. J Neurosci 2018; 39:788-801. [PMID: 30523065 DOI: 10.1523/jneurosci.0567-18.2018] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 10/01/2018] [Accepted: 10/30/2018] [Indexed: 12/21/2022] Open
Abstract
Hippocampus-dependent learning processes are coordinated via a large diversity of GABAergic inhibitory mechanisms. The α5 subunit-containing GABAA receptor (α5-GABAAR) is abundantly expressed in the hippocampus populating primarily the extrasynaptic domain of CA1 pyramidal cells, where it mediates tonic inhibitory conductance and may cause functional deficits in synaptic plasticity and hippocampus-dependent memory. However, little is known about synaptic expression of the α5-GABAAR and, accordingly, its location site-specific function. We examined the cell- and synapse-specific distribution of the α5-GABAAR in the CA1 stratum oriens/alveus (O/A) using a combination of immunohistochemistry, whole-cell patch-clamp recordings and optogenetic stimulation in hippocampal slices obtained from mice of either sex. In addition, the input-specific role of the α5-GABAAR in spatial learning and anxiety-related behavior was studied using behavioral testing and chemogenetic manipulations. We demonstrate that α5-GABAAR is preferentially targeted to the inhibitory synapses made by the vasoactive intestinal peptide (VIP)- and calretinin-positive terminals onto dendrites of somatostatin-expressing interneurons. In contrast, synapses made by the parvalbumin-positive inhibitory inputs to O/A interneurons showed no or little α5-GABAAR. Inhibiting the α5-GABAAR in control mice in vivo improved spatial learning but also induced anxiety-like behavior. Inhibiting the α5-GABAAR in mice with inactivated CA1 VIP input could still improve spatial learning and was not associated with anxiety. Together, these data indicate that the α5-GABAAR-mediated phasic inhibition via VIP input to interneurons plays a predominant role in the regulation of anxiety while the α5-GABAAR tonic inhibition via this subunit may control spatial learning.SIGNIFICANCE STATEMENT The α5-GABAAR subunit exhibits high expression in the hippocampus, and regulates the induction of synaptic plasticity and the hippocampus-dependent mnemonic processes. In CA1 principal cells, this subunit occupies mostly extrasynaptic sites and mediates tonic inhibition. Here, we provide evidence that, in CA1 somatostatin-expressing interneurons, the α5-GABAAR subunit is targeted to synapses formed by the VIP- and calretinin-expressing inputs, and plays a specific role in the regulation of anxiety-like behavior.
Collapse
|
22
|
Bali P, Bammidi S, Banik A, Nehru B, Anand A. CD34 and CD117 Stemness of Lineage-Negative Cells Reverses Memory Loss Induced by Amyloid Beta in Mouse Model. Front Behav Neurosci 2018; 12:222. [PMID: 30443207 PMCID: PMC6222267 DOI: 10.3389/fnbeh.2018.00222] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2018] [Accepted: 09/04/2018] [Indexed: 12/19/2022] Open
Abstract
A majority of the neurodegenerative disorders including Alzheimer's disease are untreatable and occur primarily due to aging and rapidly changing lifestyles. The rodent Alzheimer's disease models are critical for investigating the underlying disease pathology and screening of novel therapeutic targets in preclinical settings. We aimed to characterize the stemness properties of human umbilical cord blood (hUCB) derived lineage-negative (Lin−) stem cells based on CD34 and CD117 expression as well as surface morphology using flow cytometry and scanning electron microscopy, respectively. The efficacy of the stem cells was tested by its capacity to rescue the injury caused by intrahippocampal delivery of varying doses of amyloid beta. The hUCB Lin− stem cells reversed memory loss due to Aβ42-induced injury more effectively at micromolar concentration, and not picomolar concentration. More studies are required to delineate the underlying molecular events associated with hUCB Lin− stem cells.
Collapse
Affiliation(s)
- Parul Bali
- Department of Biophysics, Panjab University, Chandigarh, India.,Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sridhar Bammidi
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Avijit Banik
- Department of Pharmacology, Rollins Research Center, Emory University School of Medicine, Atlanta, GA, United States
| | - Bimla Nehru
- Department of Biophysics, Panjab University, Chandigarh, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
23
|
Huber CM, Yee C, May T, Dhanala A, Mitchell CS. Cognitive Decline in Preclinical Alzheimer's Disease: Amyloid-Beta versus Tauopathy. J Alzheimers Dis 2018; 61:265-281. [PMID: 29154274 PMCID: PMC5734131 DOI: 10.3233/jad-170490] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We perform a large-scale meta-analysis of 51 peer-reviewed 3xTg-AD mouse publications to compare Alzheimer’s disease (AD) quantitative clinical outcome measures, including amyloid-β (Aβ), total tau, and phosphorylated tau (pTau), with cognitive performance in Morris water maze (MWM) and Novel Object Recognition (NOR). “High” levels of Aβ (Aβ40, Aβ42) showed significant but weak trends with cognitive decline (MWM: slope = 0.336, R2 = 0.149, n = 259, p < 0.001; NOR: slope = 0.156, R2 = 0.064, n = 116, p < 0.05); only soluble Aβ or directly measured Aβ meaningfully contribute. Tau expression in 3xTg-AD mice was within 10–20% of wild type and not associated with cognitive decline. In contrast, increased pTau is directly and significantly correlated with cognitive decline in MWM (slope = 0.408, R2 = 0.275, n = 371, p < < 0.01) and NOR (slope = 0.319, R2 = 0.176, n = 113, p < 0.05). While a variety of pTau epitopes (AT8, AT270, AT180, PHF-1) were examined, AT8 correlated most strongly with cognition (slope = 0.586, R2 = 0.521, n = 185, p < < 0.001). Multiple linear regression confirmed pTau is a stronger predictor of MWM performance than Aβ. Despite pTau’s lower physical concentration than Aβ, pTau levels more directly and quantitatively correlate with 3xTg-AD cognitive decline. pTau’s contribution to neurofibrillary tangles well after Aβ levels plateau makes pTau a viable treatment target even in late-stage clinical AD. Principal component analysis, which included hyperphosphorylation induced by kinases (pGSK3β, GSK3β, CDK5), identified phosphorylated ser9 GSK3β as the primary contributor to MWM variance. In summary, meta-analysis of cognitive decline in preclinical AD finds tauopathy more impactful than Aβ. Nonetheless, complex AD interactions dictate successful therapeutics harness synergy between Aβ and pTau, possibly through the GSK3 pathway.
Collapse
Affiliation(s)
- Colin M Huber
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA.,Department of Bioengineering, University of Pennsylvania School of Engineering and Applied Sciences, Philadelphia, PA, USA
| | - Connor Yee
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Taylor May
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Apoorva Dhanala
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| | - Cassie S Mitchell
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University School of Medicine, Atlanta, GA, USA
| |
Collapse
|
24
|
Lu C, Wang Y, Wang D, Zhang L, Lv J, Jiang N, Fan B, Liu X, Wang F. Neuroprotective Effects of Soy Isoflavones on Scopolamine-Induced Amnesia in Mice. Nutrients 2018; 10:E853. [PMID: 29966363 PMCID: PMC6073222 DOI: 10.3390/nu10070853] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2018] [Revised: 06/26/2018] [Accepted: 06/28/2018] [Indexed: 01/02/2023] Open
Abstract
In the recent years, interest in soybean as a neuroprotective nutrient in the management of Alzheimer’s disease (AD) has increased and soy isoflavones (SI), as kinds of soybean phytochemicals, are thought to be biologically active components that confer this beneficial effect against neurodegenerative diseases. However, the neuroprotective effect of SI is not well understood. Therefore, the present study (30 days) was conducted to investigate the neuroprotective effects of soy isoflavones (SI) on scopolamine (SCOP)-induced memory impairments in Institute of Cancer Research (ICR) mice (aged 4 weeks) and to elucidate its underlying mechanisms of action. SI (40 mg/kg) administration improved the cognitive performance of SCOP-treated mice in an object location recognition task and the Morris water maze test. SI (40 mg/kg) administration significantly enhanced cholinergic system function and suppressed oxidative stress levels in the hippocampus of SCOP-treated mice. Furthermore, SI (40 mg/kg) treatment markedly upregulated the phosphorylation levels of extracellular signal-regulated kinase (ERK), cAMP response element-binding protein (CREB) and brain-derived neurotrophic factor (BDNF) expression levels in the hippocampus. Taken together, these results demonstrated that soy isoflavones exerted a significant neuroprotective effect on cognitive dysfunctions induced by scopolamine, suggesting that soy isoflavones could be a good candidate for possible treatment of neurodegenerative diseases, such as Alzheimer’s disease (AD).
Collapse
Affiliation(s)
- Cong Lu
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100193, China.
| | - Yan Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| | - Donghui Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| | - Lijing Zhang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| | - Jingwei Lv
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100193, China.
| | - Ning Jiang
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100193, China.
| | - Bei Fan
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| | - Xinmin Liu
- Research Center for Pharmacology & Toxicology, Institute of Medicinal Plant Development (IMPLAD), Chinese Academy of Medical Sciences (CAMS) and Peking Union Medical College (PUMC), Beijing 100193, China.
| | - Fengzhong Wang
- Institute of Food Science and Technology, Chinese Academy of Agricultural Sciences (CAAS), Beijing 100193, China.
| |
Collapse
|
25
|
Wachiryah TA, Hathaipat L. Enhancing effect of Tiliacora triandra leaves extract on spatial learning, memory and learning flexibility as well as hippocampal choline acetyltransferase activity in mice. AVICENNA JOURNAL OF PHYTOMEDICINE 2018; 8:380-388. [PMID: 30377596 PMCID: PMC6204150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVE The present study investigates the effect of Tiliacora triandra leaf extract on spatial learning, memory, and learning flexibility as well as hippocampal choline acetyltransferase (ChAT) activity in mice. MATERIALS AND METHODS Thirty male ICR mice were randomly divided into three groups including 10% Tween 80, T. triandra 300 mg/kg and T. triandra 600 mg/kg. All administrations were done orally for 18 consecutive days. Spatial learning, memory and learning flexibility were assessed using the Morris water maze. ChAT activity and hippocampal neuronal cell number were assessed by immunohistochemistry and histological methods, respectively. RESULTS The results demonstrated that T. triandra leaf extract (300 and 600 mg/kg) significantly enhances spatial learning and learning flexibility. Only 300 mg/kg of T. triandra significantly improved the spatial memory. The hippocampal ChAT activity and total hippocampal cell number were significantly increased in T. triandra-treated groups. CONCLUSION The present study indicated that T. triandra leaf extract improves the spatial learning, memory and learning flexibility, exerts neuroprotective effects on hippocampal neurons and maintains ChAT activity in this brain area.
Collapse
Affiliation(s)
- Thong-asa Wachiryah
- Animal Toxicology and Physiology Specialty Research Unit (ATPSRU), Physiology Division, Department of Zoology, Faculty of Science, Kasetsart University, Bangkok, Thailand,Corresponding Author: Tel: +662 562-5555, Fax: +662 942-8695,
| | | |
Collapse
|
26
|
Zhu JD, Wang JJ, Zhang XH, Yu Y, Kang ZS. Panax ginseng extract attenuates neuronal injury and cognitive deficits in rats with vascular dementia induced by chronic cerebral hypoperfusion. Neural Regen Res 2018; 13:664-672. [PMID: 29722318 PMCID: PMC5950676 DOI: 10.4103/1673-5374.230292] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Panax ginseng is a slow-growing perennial plant. Panax ginseng extract has numerous biological activities, including antitumor, anti-inflammatory and antistress activities. Panax ginseng extract also has a cognition-enhancing effect in rats with alcohol-induced memory impairment. In this study, we partially occluded the bilateral carotid arteries in the rat to induce chronic cerebral hypoperfusion, a well-known model of vascular dementia. The rats were then intragastrically administered 50 or 100 mg/kg Panax ginseng extract. Morris water maze and balance beam tests were used to evaluate memory deficits and motor function, respectively. Protein quantity was used to evaluate cholinergic neurons. Immunofluorescence staining was used to assess the number of glial fibrillary acidic protein-positive cells. Western blot assay was used to evaluate protein levels of vascular endothelial growth factor, basic fibroblast growth factor, Bcl-2 and Bax. Treatment with Panax ginseng extract for 8 weeks significantly improved behavioral function and increased neuronal density and VEGF and bFGF protein expression in the hippocampal CA3 area. Furthermore, Panax ginseng extract reduced the number of glial fibrillary acidic protein-immunoreactive cells, and it decreased apoptosis by upregulating Bcl-2 and downregulating Bax protein expression. The effect of Panax ginseng extract was dose-dependent and similar to that of nimodipine, a commonly used drug for the treatment of vascular dementia. These findings suggest that Panax ginseng extract is neuroprotective against vascular dementia induced by chronic cerebral hypoperfusion, and therefore might have therapeutic potential for preventing and treating the disease.
Collapse
Affiliation(s)
- Jun-De Zhu
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guian New Area, Guizhou Province, China
| | - Jun-Jie Wang
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guian New Area, Guizhou Province, China
| | - Xian-Hu Zhang
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guian New Area, Guizhou Province, China
| | - Yan Yu
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guian New Area, Guizhou Province, China
| | - Zhao-Sheng Kang
- Department of Anatomy, School of Basic Medicine, Guizhou Medical University, Guian New Area, Guizhou Province, China
| |
Collapse
|
27
|
Neuroprotective effect of ipriflavone against scopolamine-induced memory impairment in rats. Psychopharmacology (Berl) 2017; 234:3037-3053. [PMID: 28733814 DOI: 10.1007/s00213-017-4690-x] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
Abstract
BACKGROUND Alzheimer's disease is an age-related neurodegenerative disorder characterized clinically by a progressive loss of memory and cognitive functions resulting in severe dementia. Ipriflavone (IPRI) is a non-hormonal, semi-synthetic isoflavone, clinically used in some countries for the treatment and prevention of postmenopausal osteoporosis. Moreover, ipriflavone is a non-peptidomimetic small molecule AChE inhibitor with an improved bioavailability after systemic administration, due to its efficient blood-brain barrier permeability in comparison with peptidomimetic inhibitors. OBJECTIVE The present study aimed to evaluate the possible enhancing effects of IPRI on memory impairments caused by scopolamine administration. METHODS Male rats were administered IPRI (50 mg/kg, oral) 2 h before scopolamine injection (2 mg/kg, intraperitoneally injected) daily for 4 weeks. Effects of IPRI on acetylcholinesterase activity, amyloid-β precursor processing, and neuroplasticity in the rats' hippocampus were investigated. RESULTS Daily administration of IPRI reverted memory impairment caused by scopolamine as measured by the reduction of the escape latency. IPRI significantly alleviated the oxidative stress and restored the mRNA expression of both cAMP-response element-binding protein and brain-derived neurotrophic factor in the hippocampus. Furthermore, it significantly increased the expression of ADAM10 and ADAM17 (two putative α-secretase enzymes) and phosphorylated extracellular signal-regulated kinase 1/2 (pERK1/2) that associated with decreased expression of β-secretase (BACE) in the hippocampus. Finally, both the amyloid-β (Aβ) and Tau pathologies were reduced. CONCLUSIONS IPRI showed promising neuroprotective effects against scopolamine-induced memory dysfunction in rats. These findings contributed to the stimulation of α-secretase enzymes, the activation of MAPK/ERK1/2, and the alleviation of oxidative stress.
Collapse
|
28
|
Improvement of spatial learning and memory, cortical gyrification patterns and brain oxidative stress markers in diabetic rats treated with Ficus deltoidea leaf extract and vitexin. J Tradit Complement Med 2017; 8:190-202. [PMID: 29322009 PMCID: PMC5755998 DOI: 10.1016/j.jtcme.2017.05.006] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2017] [Revised: 05/07/2017] [Accepted: 05/17/2017] [Indexed: 02/07/2023] Open
Abstract
Despite the fact that Ficus deltoidea and vitexin played important roles in controlling hyperglycemia, an effective mitigation strategy dealing with cognitive deficit observed in diabetes, little is known about its neuroprotective effects. The study is aimed to determine changes in behavioral, gyrification patterns and brain oxidative stress markers in streptozotocin (STZ)-induced diabetic rats following F. deltoidea and vitexin treatments. Diabetic rats were treated orally with metformin, methanolic extract of F. deltoidea leaves and vitexin for eight weeks. Morris water maze (MWM) test was performed to evaluate learning and memory functions. The patterns of cortical gyrification were subsequently visualized using micro-computed tomography (micro-CT). Quantification of brain oxidative stress biomarkers, insulin, amylin as well as serum testosterone were measured using a spectrophotometer. The brain fatty acid composition was determined using gas chromatography (GC). Biochemical variation in brain was estimated using Fourier transform infrared (FT-IR) spectroscopy. Results showed that oral administration of F. deltoidea extract and vitexin to diabetic rats attenuated learning and memory impairment, along with several clusters of improved gyrification. Both treatments also caused a significant increase in the superoxide dismutase (SOD) and glutathione peroxidase (GPx) values, as well as a significant reduction of TBARS. Strikingly, improvement of cortical gyrification, spatial learning and memory are supported by serum testosterone levels, fatty acid composition of brain and FT-IR spectra.
Collapse
|
29
|
Fang M, Zhang P, Zhao Y, Liu X. Bioinformatics and co-expression network analysis of differentially expressed lncRNAs and mRNAs in hippocampus of APP/PS1 transgenic mice with Alzheimer disease. Am J Transl Res 2017; 9:1381-1391. [PMID: 28386363 PMCID: PMC5376028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 02/01/2017] [Indexed: 06/07/2023]
Abstract
APP/PS1 transgenic mice with Alzheimer disease (AD) are widely used as a reliable animal model in studies about behaviors, physiology, biochemistry and histomorphology of AD, but few studies have been conducted to investigate the role of lncRNAs in this model. In this study, lncRNA microarray was employed to detect the gene expression profile and lncRNA expression profile in the mouse brain. Then, bioinformatics was used to predict the differentially expressed genes related to AD (n=20). Among different lncRNAs (n=249), 99 were downregulated and 150 upregulated. Co-expression network was applied to analyze the co-expression of differential lncRNAs and different genes. In network, lncRNA Gm13498 and lncRNA 1700030L20Rik correlated with the most genes and their degrees were 6 and 5, respectively. Then, the function and signal transduction pathways related to the differentially co-expressed lncRNAs were analyzed with bioinformatics, and results showed that these lncRNAs were involved in the systemic development of neurons, intercellular communication, regulation of action potential of neurons, development and differentiation of oligodendrocytes, neurotransmitters transmission, and neuronal regeneration. Realtime PCR was employed to detect the expression of relevant lncRNAs and differentially expressed RNAs in 10 samples, and results were consistent with above findings from microarray.
Collapse
Affiliation(s)
- Min Fang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University Shanghai, China
| | - Pei Zhang
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University Shanghai, China
| | - Yanxin Zhao
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University Shanghai, China
| | - Xueyuan Liu
- Department of Neurology, Shanghai Tenth People's Hospital, Tongji University Shanghai, China
| |
Collapse
|