1
|
Adeyemo SA, Ajao MY, Ogundeyi KJ, Femi-Akinlosotu O, Bakre AG. Ameliorative potential of ethanol extract of Calyptrochilum emarginatum leaves on scopolamine-induced amnesia in male swiss mice. JOURNAL OF ETHNOPHARMACOLOGY 2025; 336:118731. [PMID: 39182698 DOI: 10.1016/j.jep.2024.118731] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Calyptrochilum emarginatum (Afzel. Ex Sw.) Schltr. (Orchidaceae) is a traditional medicinal plant known for its antimicrobial properties and efficacy in managing convulsive fever and menstrual disorders and addressing conditions such as malaria, tuberculosis, and cough. AIM OF THE STUDY The study aims to examine the memory-enhancing and neuroprotective properties of ethanol extract of Calyptrochilum emarginatum leaves (EECEL) in scopolamine-induced amnesia mice model. MATERIALS AND METHODS Forty-two male mice were divided into six groups (n = 7). Group 1 served as control, administered distilled water (10 mL/kg, p. o), group 2 received scopolamine only (3 mg/kg, i. p.), groups 3 to 6 received pretreatments of EECEL (50, 100, and 200 mg/kg, p. o.) and donepezil (1 mg/kg, p. o.) 30 min before scopolamine (3 mg/kg), for seven days. Following treatments, behavioral (learning and memory) assessments were carried out, while biochemical (acetylcholinesterase activity, oxidative stress markers, inflammatory cytokines markers) and histological evaluations were done after euthanasia. RESULTS Scopolamine significantly impaired spatial, long term and recognition memory. Nevertheless, administration of EECEL (50, 100, and 200 mg/kg orally) enhanced memory function in mice, as observed in the Y maze [F (5, 30) = 20.23, p < 0.0001], Morris water maze [F (10, 90) = 3.105, p = 0.0019; [F (5, 30) = 21.13, p < 0.0001]], and novel object recognition tasks [F (5, 30) = 37.22, p < 0.0001)]. Scopolamine-treated mice exhibited significant dysfunction in the cholinergic system, as evidenced by elevated AChE activity [0.099 ± 0.005 vs. 0.063 ± 0.004 mol/min/g] with an elevation in oxidative stress. On the other hand, administration of EECEL counteracted these consequences by reducing AChE activity, mitigating oxidative damage, reducing pro-inflammatory cytokines, and preventing degeneration of neurons. CONCLUSION The results demonstrated that EECEL effectively mitigates scopolamine-induced memory impairment via an oxido-inflammatory mechanism and modulation of the central cholinergic system.
Collapse
Affiliation(s)
- Stella Afolakemi Adeyemo
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| | - Mutiu Yombo Ajao
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| | - Kehinde Joshua Ogundeyi
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| | - Omowunmi Femi-Akinlosotu
- Developmental Neurobiology and Forensic Anatomy Unit, Department of Anatomy, University of Ibadan, Ibadan, Nigeria.
| | - Adewale Ganiyu Bakre
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, University of Ibadan, Ibadan, Oyo-State, Nigeria.
| |
Collapse
|
2
|
Ribarič S. Detecting Early Cognitive Decline in Alzheimer's Disease with Brain Synaptic Structural and Functional Evaluation. Biomedicines 2023; 11:355. [PMID: 36830892 PMCID: PMC9952956 DOI: 10.3390/biomedicines11020355] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 01/22/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023] Open
Abstract
Early cognitive decline in patients with Alzheimer's (AD) is associated with quantifiable structural and functional connectivity changes in the brain. AD dysregulation of Aβ and tau metabolism progressively disrupt normal synaptic function, leading to loss of synapses, decreased hippocampal synaptic density and early hippocampal atrophy. Advances in brain imaging techniques in living patients have enabled the transition from clinical signs and symptoms-based AD diagnosis to biomarkers-based diagnosis, with functional brain imaging techniques, quantitative EEG, and body fluids sampling. The hippocampus has a central role in semantic and episodic memory processing. This cognitive function is critically dependent on normal intrahippocampal connections and normal hippocampal functional connectivity with many cortical regions, including the perirhinal and the entorhinal cortex, parahippocampal cortex, association regions in the temporal and parietal lobes, and prefrontal cortex. Therefore, decreased hippocampal synaptic density is reflected in the altered functional connectivity of intrinsic brain networks (aka large-scale networks), including the parietal memory, default mode, and salience networks. This narrative review discusses recent critical issues related to detecting AD-associated early cognitive decline with brain synaptic structural and functional markers in high-risk or neuropsychologically diagnosed patients with subjective cognitive impairment or mild cognitive impairment.
Collapse
Affiliation(s)
- Samo Ribarič
- Faculty of Medicine, Institute of Pathophysiology, University of Ljubljana, Zaloška 4, SI-1000 Ljubljana, Slovenia
| |
Collapse
|
3
|
Souza MA, Peres Bomfim LG, de Barros VL, Medeiros RC, Ginsicke DC, Colovati MES, Daly T, Zanesco A. The Differential Effect of Schooling and Physical Activity on Dementia in Older Women and Men from Brazil: Implications for Policymaking. J Alzheimers Dis 2022; 90:859-867. [DOI: 10.3233/jad-220517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Background: Modifiable risk factors exert crucial impact on dementia. Objective: We sought to answer the question: do two modifiable risk factors, schooling level and physical activity (PA), affect cognitive function similarly in each sex? Methods: This cross-sectional study was conducted in 2019 and 2021, and the survey was applied to the residents of the metropolitan area of Santos, a seashore of Sao Paulo State. Four hundred and twenty-two participants (women = 254 and men = 168) were eligible. Baecke questionnaire for the elderly was applied for the classification as physically inactive (PI) or active (PA). Cognitive function was assessed by the Mini-Mental State Examination (MMSE) and the Clinical Dementia Rating (CDR). Participants were also stratified by schooling status for both sexes. Results: Higher education had a sex-independent positive influence on MMSE and CDR (p < 0.001). PA influences positively MMSE in older women (PI: 25±5 and PA: 27±3, p < 0.03), but has no effect in older men (26±5 and 25±5, p > 0.05). Concordantly, older women who were PA (1.7 and 0 %) showed a lower prevalence of dementia compared with PI (6.2 and 2.1%), for mild and moderate respectively. Active older women had higher odds of improving the MMSE score (OR: 1.093; 95% CI: 1.008–1.186) than men (OR: 0.97 (95% CI: 0.896–1.051). Conclusion: Education affects cognitive function equally in Brazilian elderly whereas older women are more responsive to the beneficial effects of PA for dementia than men.
Collapse
Affiliation(s)
- Melyssa Alves Souza
- Postgraduate Program in Environmental Health, Metropolitan University of Santos (UNIMES), Santos, SP, Brazil
- Department of Biosciences, Institute of Health and Society, Federal University of Sao Paulo (UNIFESP), Santos, SP, Brazil
| | - Larissa Gomes Peres Bomfim
- Postgraduate Program in Environmental Health, Metropolitan University of Santos (UNIMES), Santos, SP, Brazil
| | - Vinicius Lúcio de Barros
- Postgraduate Program in Environmental Health, Metropolitan University of Santos (UNIMES), Santos, SP, Brazil
| | - Ronaldo Coelho Medeiros
- Postgraduate Program in Environmental Health, Metropolitan University of Santos (UNIMES), Santos, SP, Brazil
| | - Danielle Cristine Ginsicke
- Postgraduate Program in Environmental Health, Metropolitan University of Santos (UNIMES), Santos, SP, Brazil
| | | | - Timothy Daly
- Sorbonne Université, Science Norms Democracy, UMR 8011, Paris, France
| | - Angelina Zanesco
- Postgraduate Program in Environmental Health, Metropolitan University of Santos (UNIMES), Santos, SP, Brazil
| |
Collapse
|
4
|
Assessment of the In Vivo Relationship Between Cerebral Hypometabolism, Tau Deposition, TSPO Expression, and Synaptic Density in a Tauopathy Mouse Model: a Multi-tracer PET Study. Mol Neurobiol 2022; 59:3402-3413. [PMID: 35312967 PMCID: PMC9148291 DOI: 10.1007/s12035-022-02793-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 03/05/2022] [Indexed: 11/03/2022]
Abstract
Cerebral glucose hypometabolism is a typical hallmark of Alzheimer’s disease (AD), usually associated with ongoing neurodegeneration and neuronal dysfunction. However, underlying pathological processes are not fully understood and reproducibility in animal models is not well established. The aim of the present study was to investigate the regional interrelation of glucose hypometabolism measured by [18F]FDG positron emission tomography (PET) with various molecular targets of AD pathophysiology using the PET tracers [18F]PI-2620 for tau deposition, [18F]DPA-714 for TSPO expression associated with neuroinflammation, and [18F]UCB-H for synaptic density in a transgenic tauopathy mouse model. Seven-month-old rTg4510 mice (n = 8) and non-transgenic littermates (n = 8) were examined in a small animal PET scanner with the tracers listed above. Hypometabolism was observed throughout the forebrain of rTg4510 mice. Tau pathology, increased TSPO expression, and synaptic loss were co-localized in the cortex and hippocampus and correlated with hypometabolism. In the thalamus, however, hypometabolism occurred in the absence of tau-related pathology. Thus, cerebral hypometabolism was associated with two regionally distinct forms of molecular pathology: (1) characteristic neuropathology of the Alzheimer-type including synaptic degeneration and neuroinflammation co-localized with tau deposition in the cerebral cortex, and (2) pathological changes in the thalamus in the absence of other markers of AD pathophysiology, possibly reflecting downstream or remote adaptive processes which may affect functional connectivity. Our study demonstrates the feasibility of a multitracer approach to explore complex interactions of distinct AD-pathomechanisms in vivo in a small animal model. The observations demonstrate that multiple, spatially heterogeneous pathomechanisms can contribute to hypometabolism observed in AD mouse models and they motivate future longitudinal studies as well as the investigation of possibly comparable pathomechanisms in human patients.
Collapse
|
5
|
β-Glucan from Lentinula edodes prevents cognitive impairments in high-fat diet-induced obese mice: involvement of colon-brain axis. J Transl Med 2021; 19:54. [PMID: 33541370 PMCID: PMC7863530 DOI: 10.1186/s12967-021-02724-6] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2020] [Accepted: 01/28/2021] [Indexed: 12/15/2022] Open
Abstract
Background Long-term high fat (HF) diet intake can cause neuroinflammation and cognitive decline through the gut-brain axis. (1, 3)/(1, 6)-β-glucan, an edible polysaccharide isolated from medical mushroom, Lentinula edodes (L. edodes), has the potential to remodel gut microbiota. However, the effects of L. edodes derived β-glucan against HF diet-induced neuroinflammation and cognitive decline remain unknown. This study aimed to evaluate the neuroprotective effect and mechanism of dietary L edodes β-glucan supplementation against the obesity-associated cognitive decline in mice fed by a HF diet. Methods C57BL/6J male mice were fed with either a lab chow (LC), HF or HF with L. edodes β-glucan supplementation diets for 7 days (short-term) or 15 weeks (long-term). Cognitive behavior was examined; blood, cecum content, colon and brain were collected to evaluate metabolic parameters, endotoxin, gut microbiota, colon, and brain pathology. Results We reported that short-term and long-term L. edodes β-glucan supplementation prevented the gut microbial composition shift induced by the HF diet. Long-term L. edodes β-glucan supplementation prevented the HF diet-induced recognition memory impairment assessed by behavioral tests (the temporal order memory, novel object recognition and Y-maze tests). In the prefrontal cortex and hippocampus, the β-glucan supplementation ameliorated the alteration of synaptic ultrastructure, neuroinflammation and brain-derived neurotrophic factor (BDNF) deficits induced by HF diet. Furthermore, the β-glucan supplementation increased the mucosal thickness, upregulated the expression of tight junction protein occludin, decreased the plasma LPS level, and inhibited the proinflammatory macrophage accumulation in the colon of mice fed by HF diet. Conclusions This study revealed that L. edodes β-glucan prevents cognitive impairments induced by the HF diet, which may occur via colon-brain axis improvement. The finding suggested that dietary L. edodes β-glucan supplementation may be an effective nutritional strategy to prevent obesity-associated cognitive decline.
Collapse
|
6
|
John A, Reddy PH. Synaptic basis of Alzheimer's disease: Focus on synaptic amyloid beta, P-tau and mitochondria. Ageing Res Rev 2021; 65:101208. [PMID: 33157321 PMCID: PMC7770124 DOI: 10.1016/j.arr.2020.101208] [Citation(s) in RCA: 213] [Impact Index Per Article: 53.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/26/2022]
Abstract
Alzheimer's disease (AD) is a progressive and synaptic failure disease. Despite the many years of research, AD still harbors many secrets. As more of the world's population grows older, researchers are striving to find greater information on disease progression and pathogenesis. Identifying and treating the markers of this disease, or better yet, preventing it all together, are the hopes of those investing in this field of study. Several years of research revealed that synaptic pathology and mitochondrial oxidative damage are early events in disease progression. Loss of synapses and synaptic damage are the best correlates of cognitive deficits found in AD patients. As the disease progresses, there are significant changes at the synapse. These changes can both shed greater light onto the progression of the disease and serve as markers and therapeutic targets. This article addresses the mechanisms of synaptic action, mitochondrial regulation/dysregulation, resulting synaptic changes caused by amyloid beta and phosphorylated tau in AD progression. This article also highlights recent developments of risk factors, genetics and ApoE4 involvement, factors related to synaptic damage and loss, mislocalization of amyloid beta and phosphorylated tau, mitophagy, microglial activation and synapse-based therapies in AD. Furthermore, impairments in LTD and reactivation of microglia are discussed.
Collapse
Affiliation(s)
- Albin John
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - P Hemachandra Reddy
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Public Health, Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Speech, Language, and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
7
|
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer disease: a review. AIMS Neurosci 2020; 8:86-132. [PMID: 33490374 PMCID: PMC7815481 DOI: 10.3934/neuroscience.2021005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease is a progressive neurodegenerative disorder, mainly affecting older people, which severely impairs patients' quality of life. In the recent years, the number of affected individuals has seen a rapid increase. It is estimated that up to 107 million subjects will be affected by 2050 worldwide. Research in this area has revealed a lot about the biological and environmental underpinnings of Alzheimer, especially its correlation with β-Amyloid and Tau related mechanics; however, the precise molecular events and biological pathways behind the disease are yet to be discovered. In this review, we focus our attention on the biological mechanics that may lie behind Alzheimer development. In particular, we briefly describe the genetic elements and discuss about specific biological processes potentially associated with the disease.
Collapse
Affiliation(s)
| | | | | | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| |
Collapse
|
8
|
Zhao X, Li S, Gaur U, Zheng W. Artemisinin Improved Neuronal Functions in Alzheimer's Disease Animal Model 3xtg Mice and Neuronal Cells via Stimulating the ERK/CREB Signaling Pathway. Aging Dis 2020; 11:801-819. [PMID: 32765947 PMCID: PMC7390534 DOI: 10.14336/ad.2019.0813] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Accepted: 08/13/2019] [Indexed: 01/03/2023] Open
Abstract
The most common form of dementia is Alzheimer’s disease which is characterized by memory loss and cognitive disorders. The pathogenesis of Alzheimer’s disease is not known at present but toxicity of amyloid-beta is one of the central hypotheses. Amyloid-beta can stimulate the production of reactive oxygen species (ROS), cause oxidative stress, damage mitochondrial, cause inflammatory reactions and activate apoptosis related factors which lead to the neuronal death. In this study, we found that artemisinin, a first line antimalarial drug used in clinic for decades, improved the cognitive functions in Alzheimer’s disease animal model 3xTg mice. Further study showed that artemisinin reduced the deposition of amyloid-beta and tau protein, reduced the release of inflammation factors and apoptosis factors, and thereby reduced the neuronal cell death. Western blot assay showed that artemisinin stimulated the activation of ERK/CREB signaling pathway. Consistent with these results, artemisinin concentration-dependently promoted the survival of SH-SY5Y cell against toxicity of amyloid-beta protein 1-42 induced ROS accumulation, caspase activation and apoptosis. Artemisinin also stimulated the phosphorylation of ERK1/2 and CREB in SH-SY5Y cells in time and concentration-dependent manner. Inhibition of ERK/CREB pathway attenuated the protective effect of artemisinin. These data put together suggested that artemisinin has the potential to protect neuronal cells in vitro as well as in vivo animal model 3xTg mice via, at least in part, the activation of the ERK/CREB pathway. Our findings also strongly support the potential of artemisinin as a new multi-target drug that can be used for preventing and treating the Alzheimer’s disease.
Collapse
Affiliation(s)
- Xia Zhao
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Shuai Li
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Uma Gaur
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| | - Wenhua Zheng
- Center of Reproduction, Development & Aging and Institute of Translation Medicine, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
9
|
Guo Q, Xu S, Yang P, Wang P, Lu S, Sheng D, Qian K, Cao J, Lu W, Zhang Q. A dual-ligand fusion peptide improves the brain-neuron targeting of nanocarriers in Alzheimer's disease mice. J Control Release 2020; 320:347-362. [DOI: 10.1016/j.jconrel.2020.01.039] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2019] [Revised: 01/18/2020] [Accepted: 01/20/2020] [Indexed: 12/17/2022]
|
10
|
Wang K, Sun W, Zhang L, Guo W, Xu J, Liu S, Zhou Z, Zhang Y. Oleanolic Acid Ameliorates Aβ25-35 Injection-induced Memory Deficit in Alzheimer's Disease Model Rats by Maintaining Synaptic Plasticity. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2019; 17:389-399. [PMID: 29793416 PMCID: PMC6327117 DOI: 10.2174/1871527317666180525113109] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/07/2018] [Accepted: 05/22/2018] [Indexed: 12/13/2022]
Abstract
Background: Abnormal amyloid β (Aβ) accumulation and deposition in the hippocampus is an essential process in Alzheimer’s disease (AD). Objective: To investigate whether Oleanolic acid (OA) could improve memory deficit in AD model and its possible mechanism. Methods: Forty-five SD rats were randomly divided into sham operation group, model group, and OA group. AD models by injection of Aβ25-35 were built. Morris water maze (MWM) was applied to inves-tigate learning and memory, transmission electron microscope (TEM) to observe the ultrastructure of synapse, western blot to the proteins, electrophysiology for long-term potentiation (LTP), and Ca2+ con-centration in synapse was also measured. Results: The latency time in model group was significantly longer than that in sham operation group (P=0.0001); while it was significantly shorter in the OA group than that in model group (P=0.0001); compared with model group, the times of cross-platform in OA group significantly increased (P=0.0001). TEM results showed OA could alleviate neuron damage and synapses changes induced by Aβ25-35. The expressions of CaMKII, PKC, NMDAR2B, BDNF, TrkB, and CREB protein were signif-icantly improved by OA (P=0.0001, 0.036, 0.041, 0.0001, 0.0001, 0.026, respectively) compared with that in model group; the concentration of Ca2+ was significantly lower in OA group (1.11±0.42) than that in model group (1.68±0.18); and the slope rate (P=0.0001) and amplitude (P=0.0001) of f-EPSP significantly increased in OA group. Conclusion: The present results support that OA could ameliorate Aβ-induced memory loss of AD rats by maintaining synaptic plasticity of the hippocampus
Collapse
Affiliation(s)
- Kai Wang
- Graduate Institutes, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Weiming Sun
- Graduate Institutes, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Linlin Zhang
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Wei Guo
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Jiachun Xu
- Graduate Institutes, Tianjin University of Traditional Chinese Medicine, Tianjin, 300193, China
| | - Shuang Liu
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Zhen Zhou
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| | - Yulian Zhang
- Department of Neurology, the Second Hospital Affiliated to Tianjin University of Traditional Chinese Medicine, Tianjin, 300150, China
| |
Collapse
|
11
|
Zhou H, Tai J, Xu H, Lu X, Meng D. Xanthoceraside Could Ameliorate Alzheimer's Disease Symptoms of Rats by Affecting the Gut Microbiota Composition and Modulating the Endogenous Metabolite Levels. Front Pharmacol 2019; 10:1035. [PMID: 31572201 PMCID: PMC6753234 DOI: 10.3389/fphar.2019.01035] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 08/14/2019] [Indexed: 01/29/2023] Open
Abstract
Xanthoceraside (XAN) is a natural-derived compound with anti-Alzheimer activity from the husks of Xanthoceras sorbifolia. Although its therapeutic effect had been confirmed in previous studies, the mechanism was still unclear due to its poor solubility and low permeability. In this study, the pharmacological effect of XAN on Alzheimer's disease (AD) was confirmed by behavior experiments and H&E staining observation. Fecal microbiota transplantation (FMT) experiment also replicated the therapeutic effects, which indicates the potential targets of XAN on gut microbiota. The sequencing of 16S rRNA genes in fecal samples demonstrated that XAN reversed gut microbiota dysbiosis in AD animals. XAN could change the relative abundances of several phyla and genus of bacterial, particularly the ratio of Firmicutes/Bacteroidetes. Among them, Clostridium IV, Desulfovibrio, Corynebacterium, and Enterorhabdus had been reported to be involved in the pathologic developments of AD and other central nervous system disease. In metabolomics study, a series of host endogenous metabolites were detected, including amino acids, lysophosphatidylcholine, dihydrosphingosine, phytosphingosine, inosine, and hypoxanthine, which were all closely associated with the development of AD. Combined with the Spearman's correlation analysis, it was confirmed that the increases of five bacterial strains and decreases of six bacterial strains were closely correlated with the increases of nine host metabolites and the decreases of another five host metabolites. Therefore, XAN can modulate the structure of gut microbiota in AD rats; the changes of gut microbiota were significantly correlated with endogenous metabolites, and symptom of AD was ultimately alleviated. Our findings suggest that XAN may be a potential therapeutic drug for AD, and the gut microbiota may be potential targeting territory of XAN via microbiome-gut-brain pathway.
Collapse
Affiliation(s)
- Hongxu Zhou
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| | - Jingjie Tai
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Haiyan Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Xiumei Lu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, China
| | - Dali Meng
- School of Traditional Chinese Materia Medica, Shenyang Pharmaceutical University, Shenyang, China
| |
Collapse
|
12
|
Lu YB, Chen CX, Huang J, Tian YX, Xie X, Yang P, Wu M, Tang C, Zhang WP. Nicotinamide phosphoribosyltransferase secreted from microglia via exosome during ischemic injury. J Neurochem 2019; 150:723-737. [PMID: 31269239 DOI: 10.1111/jnc.14811] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Revised: 06/12/2019] [Accepted: 06/24/2019] [Indexed: 02/06/2023]
Abstract
Nicotinamide phosphoribosyltransferase (NAMPT) is the key enzyme of the salvage pathway of nicotinamide adenine dinucleotide synthesis. NAMPT can also be secreted and functions as a cytokine. We have previously shown that in the brain, NAMPT expression and secretion can be induced in microglia upon neuroinflammation and injury. Yet the mechanism for NAMPT secretion remains unclear. Here we show that NAMPT can be actively secreted from microglia upon the treatment of ischemia-like injury - oxygen-glucose deprivation and recovery (OGD/R). We confirmed that classical ER-Golgi pathway is not involved in NAMPT secretion. NAMPT secretion was further enhanced by ATP, and the secretion was mediated by P2X7 receptor and by intracellular Ca2+ . Importantly, we found that phospholipase D inhibitor, n-butanol, phospholipase D siRNA, and wortmannin significantly decreased OGD/R-induced and ATP-enhanced release of NAMPT in microglia. After excluding the mechanisms of involving secretory autophagy, endosomes, and secretory lysosome, we have concluded that microglial NAMPT is secreted mainly via exosome. Immune-electron microscopy identifies NAMPT in extracellular vesicles with the size and morphology characteristic of exosome. With the vesicles harvested by ultra-centrifugation, exosomal NAMPT is further confirmed by Western blotting analysis. Intriguingly, the amount of NAMPT relative to exosomal protein markers remains unchanged upon the treatment of OGD/R, suggesting a constant load of exosomal NAMPT in microglia. Taken together, we have identified NAMPT is actively secreted via exosome from microglia during neuroinflammation of ischemic injury.
Collapse
Affiliation(s)
- Yun-Bi Lu
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Chen-Xiang Chen
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Jing Huang
- Department of Pharmacy, Affiliated Hospital of Medical School of Ningbo University, Ningbo, Zhejiang, China
| | - Yu-Xin Tian
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xian Xie
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Ping Yang
- Core Medical Imaging Facility, Zhejiang University School of Medicine, Zhejiang, China
| | - Ming Wu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang, China
| | - Chun Tang
- CAS Key Laboratory of Magnetic Resonance in Biological Systems, State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Wuhan Institute of Physics and Mathematics of the Chinese Academy of Sciences, Wuhan, Hubei, China
| | - Wei-Ping Zhang
- Department of Pharmacology, Key Laboratory of Medical Neurobiology of Ministry of Health of China, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Zhejiang Province Key Laboratory of Mental Disorder's Management, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
13
|
Polis B, Srikanth KD, Gurevich V, Gil-Henn H, Samson AO. L-Norvaline, a new therapeutic agent against Alzheimer's disease. Neural Regen Res 2019; 14:1562-1572. [PMID: 31089055 PMCID: PMC6557086 DOI: 10.4103/1673-5374.255980] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Growing evidence highlights the role of arginase activity in the manifestation of Alzheimer’s disease (AD). Upregulation of arginase was shown to contribute to neurodegeneration. Regulation of arginase activity appears to be a promising approach for interfering with the pathogenesis of AD. Therefore, the enzyme represents a novel therapeutic target. In this study, we administered an arginase inhibitor, L-norvaline (250 mg/L), for 2.5 months to a triple-transgenic model (3×Tg-AD) harboring PS1M146V, APPSwe, and tauP301L transgenes. Then, the neuroprotective effects of L-norvaline were evaluated using immunohistochemistry, proteomics, and quantitative polymerase chain reaction assays. Finally, we identified the biological pathways activated by the treatment. Remarkably, L-norvaline treatment reverses the cognitive decline in AD mice. The treatment is neuroprotective as indicated by reduced beta-amyloidosis, alleviated microgliosis, and reduced tumor necrosis factor transcription levels. Moreover, elevated levels of neuroplasticity related postsynaptic density protein 95 were detected in the hippocampi of mice treated with L-norvaline. Furthermore, we disclosed several biological pathways, which were involved in cell survival and neuroplasticity and were activated by the treatment. Through these modes of action, L-norvaline has the potential to improve the symptoms of AD and even interferes with its pathogenesis. As such, L-norvaline is a promising neuroprotective molecule that might be tailored for the treatment of a range of neurodegenerative disorders. The study was approved by the Bar-Ilan University Animal Care and Use Committee (approval No. 82-10-2017) on October 1, 2017.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine; Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Kolluru D Srikanth
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Vyacheslav Gurevich
- Laboratory of Cancer Personalized Medicine and Diagnostic Genomics, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| |
Collapse
|
14
|
Polis B, Srikanth KD, Elliott E, Gil-Henn H, Samson AO. L-Norvaline Reverses Cognitive Decline and Synaptic Loss in a Murine Model of Alzheimer's Disease. Neurotherapeutics 2018; 15:1036-1054. [PMID: 30288668 PMCID: PMC6277292 DOI: 10.1007/s13311-018-0669-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The urea cycle is strongly implicated in the pathogenesis of Alzheimer's disease (AD). Arginase-I (ARGI) accumulation at sites of amyloid-beta (Aβ) deposition is associated with L-arginine deprivation and neurodegeneration. An interaction between the arginase II (ARGII) and mTOR-ribosomal protein S6 kinase β-1 (S6K1) pathways promotes inflammation and oxidative stress. In this study, we treated triple-transgenic (3×Tg) mice exhibiting increased S6K1 activity and wild-type (WT) mice with L-norvaline, which inhibits both arginase and S6K1. The acquisition of spatial memory was significantly improved in the treated 3×Tg mice, and the improvement was associated with a substantial reduction in microgliosis. In these mice, increases in the density of dendritic spines and expression levels of neuroplasticity-related proteins were followed by a decline in the levels of Aβ toxic oligomeric and fibrillar species in the hippocampus. The findings point to an association of local Aβ-driven and immune-mediated responses with altered L-arginine metabolism, and they suggest that arginase and S6K1 inhibition by L-norvaline may delay the progression of AD.
Collapse
Affiliation(s)
- Baruh Polis
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel.
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel.
| | - Kolluru D Srikanth
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
- Laboratory of Molecular and Behavioral Neuroscience, The Azrieli Faculty of Medicine, Bar-Ilan University, 8th Henrietta Szold Street, P.O. Box 1589, 1311502, Safed, Israel
| | - Evan Elliott
- Laboratory of Molecular and Behavioral Neuroscience, The Azrieli Faculty of Medicine, Bar-Ilan University, 8th Henrietta Szold Street, P.O. Box 1589, 1311502, Safed, Israel
| | - Hava Gil-Henn
- Laboratory of Cell Migration and Invasion, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| | - Abraham O Samson
- Drug Discovery Laboratory, The Azrieli Faculty of Medicine, Bar-Ilan University, 1311502, Safed, Israel
| |
Collapse
|
15
|
Calcagno E, Caudano F, Passalacqua M, Pronzato MA, Fedele E, Ricciarelli R. Investigating the amyloid-beta enhancing effect of cGMP in neuro2a cells. Mech Ageing Dev 2017; 166:1-5. [PMID: 28789837 DOI: 10.1016/j.mad.2017.08.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 10/24/2022]
Abstract
Long-term potentiation (LTP) and the process of memory formation require activation of cyclic guanosine monophosphate (cGMP) and cyclic adenosine monophosphate (cAMP) pathways. Notably, recent evidence indicated that both cyclic nucleotides boost the production of amyloid-beta (Aβ) peptides. In particular, cAMP was shown to favor hippocampal LTP by stimulating the synthesis of the amyloid precursor protein APP, whereas cGMP was found to enhance LTP and to improve memory by increasing Aβ levels without affecting the expression of APP. The results of the present study substantiate that cGMP has a role in the endocytic pathway of APP and suggest a scenario where the cyclic nucleotide enhances the production of Aβ by favoring the trafficking of APP from the cell cortex to the endolysosomal compartment.
Collapse
Affiliation(s)
- Elisa Calcagno
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Francesca Caudano
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Mario Passalacqua
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Maria A Pronzato
- Department of Experimental Medicine, University of Genoa, Genoa, Italy
| | - Ernesto Fedele
- Department of Pharmacy and Center of Excellence for Biomedical Research, University of Genoa, Genoa, Italy
| | | |
Collapse
|
16
|
Kuang X, Zhou HJ, Thorne AH, Chen XN, Li LJ, Du JR. Neuroprotective Effect of Ligustilide through Induction of α-Secretase Processing of Both APP and Klotho in a Mouse Model of Alzheimer's Disease. Front Aging Neurosci 2017; 9:353. [PMID: 29163135 PMCID: PMC5673635 DOI: 10.3389/fnagi.2017.00353] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Accepted: 10/17/2017] [Indexed: 12/21/2022] Open
Abstract
Emerging evidence suggests that alpha-processing single transmembrane proteins, amyloid precursor protein (APP) and anti-aging protein Klotho, are likely to be involved in the progression of Alzheimer's disease (AD). The natural phthalide Ligustilide (LIG) has been demonstrated to protect against aging- and amyloid-β (Aβ)-induced brain dysfunction in animal models. The present study is to investigate the effects of LIG on cognitive deficits and metabolism of both APP and Klotho and its underlying mechanism in AD double-transgenic (APP/PS1) mice and cultured human cells. Our results show that treatment with LIG significantly ameliorated memory impairment and Aβ levels and plaques burden. Specifically, LIG might act as a potent enhancer of α-secretase, disintegrin, and metalloprotease 10 (ADAM10), leading to upregulation of alpha-processing of both APP and Klotho and subsequent increases in the levels of both soluble APP fragment (sAPPα) and soluble Klotho (sKL) with inhibition of IGF-1/Akt/mTOR signaling in AD mice and cultured cells. Moreover, the specific ADAM10 inhibitor (G1254023X) effectively reversed LIG-induced alpha-processing of both APP and Klotho in vitro, while Klotho gene knockdown by small interfering RNA significantly blunted LIG-mediated inhibition of IGF-1/Akt/mTOR signaling in vitro. Taken together with the reported neuroprotective effects of both sAPPα and sKL as well as autophagy induction by Akt/mTOR pathway inhibition, our findings suggest that neuroprotection of LIG against AD is associated with induction alpha-processing of APP and Klotho and potential Aβ clearance. Whether LIG might induce Aβ autophagic clearance and the underlying mechanisms need to be further studied.
Collapse
Affiliation(s)
- Xi Kuang
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Hong-Jing Zhou
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Amy H. Thorne
- Ludwig Institute for Cancer Research, University of California, San Diego, La Jolla, CA, United States
| | - Xi-Nan Chen
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Lin-Jiao Li
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Jun-Rong Du
- Key Laboratory of Drug Targeting and Drug Delivery System, Department of Pharmacology, West China School of Pharmacy, Sichuan University, Chengdu, China
- *Correspondence: Jun-Rong Du,
| |
Collapse
|