1
|
Qin F, Zhang M, Wang P, Dai Z, Li X, Li D, Jing L, Qi C, Fan H, Qin M, Li Y, Huang L, Wang T. Transcriptome analysis reveals the anti-Parkinson's activity of Mangiferin in zebrafish. Biomed Pharmacother 2024; 179:117387. [PMID: 39245002 DOI: 10.1016/j.biopha.2024.117387] [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: 05/24/2024] [Revised: 08/24/2024] [Accepted: 08/30/2024] [Indexed: 09/10/2024] Open
Abstract
As the global population ages, the incidence of Parkinson's Disease (PD) continues to rise, imposing significant social and economic burdens. Mangiferin (MGF), a polyphenolic, bioactive compound has been shown to play a role in the prevention and treatment of PD. This study investigates the neuroprotective effects of MGF in an MPTP-induced zebrafish model of PD through transcriptome analysis. Initially, optimal concentrations for modeling were determined using various MPTP and MGF combinations. The zebrafish were then divided into control, MPTP-treated, and MGF co-treated groups. Subsequent evaluations included hatching rates, mortality rates, growth and development conditions, spontaneous motor abilities, as well as measurements of enzymatic activities of SOD, CAT, and levels of GSH. Ultimately, the therapeutic efficacy of MGF on the PD model in zebrafish was assessed through transcriptome sequencing. The results demonstrated that MPTP treatment induced PD-associated symptoms in zebrafish, while MGF treatment significantly improved the motor abilities and survival rates of the PD model zebrafish, effectively reducing oxidative stress and ameliorating PD symptoms. Transcriptome sequencing further revealed that MGF may mitigate mitochondrial-related oxidative stress in PD zebrafish by modulating the expression of critical genes including lrrk2, vps35, atp13a, dnajc6, and uchl1. Differential gene expression analysis indicated that these genes are primarily involved in vital signaling pathways, such as neuroactive ligand-receptor interaction, and the calcium signaling pathway. In summary, our study provides robust scientific evidence supporting MGF as a potential therapeutic candidate for PD by preserving mitochondrial homeostasis and elucidating its mechanisms of action.
Collapse
Affiliation(s)
- Fengqing Qin
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China; College of Light Industry and Food Engineering, Guangxi University, Nanning 530004, China.
| | - Ming Zhang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Pei Wang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Ziru Dai
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Xi Li
- School of Mental Health, Wenzhou Medical University, Wenzhou, China.
| | - Dongliang Li
- Laboratory of Aquaculture Nutrition and Environmental Health (LANEH), School of Life Sciences, East China Normal University, Shanghai 200241, China.
| | - Lijun Jing
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450000, China.
| | - Cen Qi
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Heliang Fan
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Mei Qin
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Ying Li
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Likun Huang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| | - Tianci Wang
- Guangxi College and University Key Laboratory of High-Value Utilization of Seafood and Prepared Food in Beibu Gulf, Qinzhou Key Laboratory of Food Flavor Analysis and Control, Beibu Gulf University, Qinzhou 535011, China.
| |
Collapse
|
2
|
Parate SS, Upadhyay SS, S A, Karthikkeyan G, Pervaje R, Abhinand CS, Modi PK, Prasad TSK. Comparative Metabolomics and Network Pharmacology Analysis Reveal Shared Neuroprotective Mechanisms of Bacopa monnieri (L.) Wettst and Centella asiatica (L.) Urb. Mol Neurobiol 2024:10.1007/s12035-024-04223-3. [PMID: 38814535 DOI: 10.1007/s12035-024-04223-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 05/03/2024] [Indexed: 05/31/2024]
Abstract
Bacopa monnieri (L.) Wettst and Centella asiatica (L.) Urb., two nootropics, are recognized in Indian Ayurvedic texts. Studies have attempted to understand their action as memory enhancers and neuroprotectants, but many molecular aspects remain unknown. We propose that Bacopa monnieri (L.) Wettst and Centella asiatica (L.) Urb. share common neuroprotective mechanisms. Mass spectrometry-based untargeted metabolomics and network pharmacology approach were used to identify potential protein targets for the metabolites from each extract. Phytochemical analyses and cell culture validation studies were also used to assess apoptosis and ROS activity using aqueous extracts prepared from both herbal powders. Further, docking studies were also performed using the LibDock protocol. Untargeted metabolomics and network pharmacology approach unveiled 2751 shared metabolites and 3439 and 2928 non-redundant metabolites from Bacopa monnieri and Centella asiatica extracts, respectively, suggesting a potential common neuroprotective mechanism among these extracts. Protein-target prediction highlighted 92.4% similarity among the proteins interacting with metabolites for these extracts. Among them, kinases mapped to MAPK, mTOR, and PI3K-AKT signaling pathways represented a predominant population. Our results highlight a significant similarity in the metabolome of Bacopa monnieri (L.) Wettst and Centella asiatica (L.) Urb., and their potential protein targets may be attributed to their common neuroprotective functions.
Collapse
Affiliation(s)
- Sakshi Sanjay Parate
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Shubham Sukerndeo Upadhyay
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Amrutha S
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Gayathree Karthikkeyan
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | | | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, 575018, India.
| | | |
Collapse
|
3
|
Gray NE, Hack W, Brandes MS, Zweig JA, Yang L, Marney L, Choi J, Magana AA, Cerruti N, McFerrin J, Koike S, Nguyen T, Raber J, Quinn JF, Maier CS, Soumyanath A. Amelioration of age-related cognitive decline and anxiety in mice by Centella asiatica extract varies by sex, dose and mode of administration. FRONTIERS IN AGING 2024; 5:1357922. [PMID: 38770167 PMCID: PMC11102990 DOI: 10.3389/fragi.2024.1357922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024]
Abstract
Background: A water extract (CAW) of the Ayurvedic plant Centella asiatica administered in drinking water has been shown to improve cognitive deficits in mouse models of aging and neurodegenerative diseases. Here the effects of CAW administered in drinking water or the diet on cognition, measures of anxiety and depression-like behavior in healthy aged mice are compared. Methods: Three- and eighteen-month-old male and female C57BL6 mice were administered rodent AIN-93M diet containing CAW (0, 0.2, 0.5 or 1% w/w) to provide 0, 200 mg/kg/d, 500 mg/kg/d or 1,000 mg/kg/d CAW for a total of 5 weeks. An additional group of eighteen-month-old mice were treated with CAW (10 mg/mL) in their drinking water CAW for a total of 5 weeks to deliver the same exposure of CAW as the highest dietary dose (1,000 mg/kg/d). CAW doses delivered were calculated based on food and water consumption measured in previous experiments. In the fourth and fifth weeks, mice underwent behavioral testing of cognition, anxiety and depression (n = 12 of each sex per treatment group in each test). Results: Aged mice of both sexes showed cognitive deficits relative to young mice while only female aged mice showed increased anxiety compared to the young female mice and no differences in depression were observed between the different ages. CAW (1,000 mg/kg/d) in the drinking water improved deficits in aged mice in learning, executive function and recognition memory in both sexes and attenuated the increased measures of anxiety observed in the aged female mice. However, CAW in the diet only improved executive function in aged mice at the highest dose (1,000 mg/kg/d) in both sexes and did so less robustly than when given in the water. There were no effects of CAW on depression-like behavior in aged animals regardless of whether it was administered in the diet or the water. Conclusions: These results suggest that CAW can ameliorate age-related changes in measures of anxiety and cognition and that the mode of administration is important for the effects of CAW on resilience to these age-related changes.
Collapse
Affiliation(s)
- Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Wyatt Hack
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Mikah S. Brandes
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Jonathan A. Zweig
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Liping Yang
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Luke Marney
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Armando Alcazar Magana
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Natasha Cerruti
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Oregon’s Wild Harvest, Redmond, OR, United States
| | - Janis McFerrin
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Oregon’s Wild Harvest, Redmond, OR, United States
| | - Seiji Koike
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Thuan Nguyen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Jacob Raber
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Joseph F. Quinn
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Healthcare System, Portland, OR, United States
| | - Claudia S. Maier
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
4
|
Maruthiyodan S, Mumbrekar KD, Guruprasad KP. Involvement of mitochondria in Alzheimer's disease pathogenesis and their potential as targets for phytotherapeutics. Mitochondrion 2024; 76:101868. [PMID: 38462158 DOI: 10.1016/j.mito.2024.101868] [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: 08/22/2023] [Revised: 03/01/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Alzheimer's disease (AD) is the leading cause of dementia around the globe. The disease's genesis is multifaceted, and its pathophysiology is complicated. Malfunction of mitochondria has been regarded as one of the intracellular events that are substantially damaged in the onset of AD and are likely a common trait of other neurodegenerative illnesses. Several mitochondrial characteristics begin to diminish with age, eventually reaching a state of significant functional failure concurrent with the beginning of neurodegenerative diseases, however, the exact timing of these processes is unknown. Mitochondrial malfunction has a multitude of negative repercussions, including reduced calcium buffering and secondary excitotoxicity contributing to synaptic dysfunction, also free radical production, and activation of the mitochondrial permeability transition. Hence mitochondria are considered a therapeutic target in neurodegenerative disorders such as Alzheimer's. Traditional medicinal systems practiced in different countries employing various medicinal plants postulated to have potential role in the therapy and management of memory impairment including amnesia, dementia as well as AD. Although, the preclinical and clinical studies using these medicinal plants or plant products have demonstrated the therapeutic efficacy for AD, the precise mechanism of action is still obscure. Therefore, this review discusses the contribution of mitochondria towards AD pathogenesis and considering phytotherapeutics as a potential therapeutic strategy.
Collapse
Affiliation(s)
- Swathi Maruthiyodan
- Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Kamalesh Dattaram Mumbrekar
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India
| | - Kanive Parashiva Guruprasad
- Centre for Ayurvedic Biology, Department of Ageing Research, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka 576104, India.
| |
Collapse
|
5
|
Hack W, Gladen-Kolarsky N, Chatterjee S, Liang Q, Maitra U, Ciesla L, Gray NE. Gardenin A treatment attenuates inflammatory markers, synuclein pathology and deficits in tyrosine hydroxylase expression and improves cognitive and motor function in A53T-α-syn mice. Biomed Pharmacother 2024; 173:116370. [PMID: 38458012 PMCID: PMC11017674 DOI: 10.1016/j.biopha.2024.116370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 02/27/2024] [Accepted: 02/28/2024] [Indexed: 03/10/2024] Open
Abstract
Oxidative stress and neuroinflammation are widespread in the Parkinson's disease (PD) brain and contribute to the synaptic degradation and dopaminergic cell loss that result in cognitive impairment and motor dysfunction. The polymethoxyflavone Gardenin A (GA) has been shown to activate the NRF2-regulated antioxidant pathway and inhibit the NFkB-dependent pro-inflammatory pathway in a Drosophila model of PD. Here, we evaluate the effects of GA on A53T alpha-synuclein overexpressing (A53TSyn) mice. A53TSyn mice were treated orally for 4 weeks with 0, 25, or 100 mg/kg GA. In the fourth week, mice underwent behavioral testing and tissue was harvested for immunohistochemical analysis of tyrosine hydroxylase (TH) and phosphorylated alpha synuclein (pSyn) expression, and quantification of synaptic, antioxidant and inflammatory gene expression. Results were compared to vehicle-treated C57BL6J mice. Treatment with 100 mg/kg GA improved associative memory and decreased abnormalities in mobility and gait in A53TSyn mice. GA treatment also reduced pSyn levels in both the cortex and hippocampus and attenuated the reduction in TH expression in the striatum seen in A53Tsyn mice. Additionally, GA increased cortical expression of NRF2-regulated antioxidant genes and decreased expression of NFkB-dependent pro-inflammatory genes. GA was readily detectable in the brains of treated mice and modulated the lipid profile in the deep gray brain tissue of those animals. While the beneficial effects of GA on cognitive deficits, motor dysfunction and PD pathology are promising, future studies are needed to further fully elucidate the mechanism of action of GA, optimizing dosing and confirm these effects in other PD models.
Collapse
Affiliation(s)
- Wyatt Hack
- Oregon Health & Science University, Neurology, Portland, United States
| | | | | | - Qiaoli Liang
- University of Alabama, Mass spectrometry facility, Chemistry and Biochemistry, Tuscaloosa, United States
| | - Urmila Maitra
- University of Alabama, Biological Sciences, Tuscaloosa, United States
| | - Lukasz Ciesla
- University of Alabama, Biological Sciences, Tuscaloosa, United States.
| | - Nora E Gray
- Oregon Health & Science University, Neurology, Portland, United States.
| |
Collapse
|
6
|
Speers AB, Wright KM, Brandes MS, Kedjejian N, Matthews DG, Caruso M, Harris CJ, Koike S, Nguyen T, Quinn JF, Soumyanath A, Gray NE. Mode of administration influences plasma levels of active Centella asiatica compounds in 5xFAD mice while markers of neuroinflammation remain unaltered. Front Neurosci 2024; 18:1277626. [PMID: 38591068 PMCID: PMC10999680 DOI: 10.3389/fnins.2024.1277626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction A water extract of Centella asiatica (L.) Urban [Apiaceae] (CAW) has demonstrated cognitive-enhancing effects in mouse models of Alzheimer's disease and aging, the magnitude of which is influenced by whether CAW is delivered in the drinking water or the diet. These cognitive benefits are accompanied by improvements in oxidative stress and mitochondrial function in the brain, two pathways related to the neuroinflammatory response. The effect of CAW on neuroinflammation, however, has not been directly studied. Here, we investigated the effect of CAW on neuroinflammation in 5xFAD mice and compared plasma levels of CAW's active compounds following two modes of CAW administration. Methods Eight-to-nine-month-old male and female 5xFAD mice and their wild-type littermates were administered CAW in their diet or drinking water (0 or 1,000 mg/kg/day) for five weeks. Immunohistochemistry was performed for β-amyloid (Aβ), glial fibrillary acidic protein (GFAP), and Griffonia simplicifolia lectin I (GSL I) in the cortex and hippocampus. Gene expression of inflammatory mediators (IL-6, TNFα, IL-1β, TREM2, AIF1, CX3CR1, CX3CL1, CD36, C3AR1, RAGE, CCR6, CD3E) was measured in the deep grey matter. Results CAW decreased cortical Aβ plaque burden in female 5xFAD mice administered CAW in the drinking water but had no effect on Aβ plaques in other treatment groups. CAW did not impact elevated levels of GFAP or GSL I in 5xFAD mice, regardless of sex, brain region, or mode of CAW administration. In the deep grey matter, CAW increased C3AR1 expression in 5xFAD females administered CAW in the drinking water and decreased IL-1β expression in 5xFAD males administered CAW in the diet. CAW had no effect, however, on gene expression levels of any other inflammatory mediator in the deep grey, for either sex or mode of CAW administration. Mice administered CAW in the drinking water versus the diet had significantly higher plasma levels of CAW compounds. Discussion CAW had little impact on the neuroinflammatory markers selected for evaluation in the present study, suggesting that the cognitive benefits of CAW may not be mediated by an anti-inflammatory effect or that additional molecular markers are needed to fully characterize the effect of CAW on neuroinflammation.
Collapse
Affiliation(s)
- Alex B. Speers
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Kirsten M. Wright
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Mikah S. Brandes
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nareg Kedjejian
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Christopher J. Harris
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Seiji Koike
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Thuan Nguyen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Joseph F. Quinn
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
7
|
Gray NE, Hack W, Brandes MS, Zweig JA, Yang L, Marney L, Choi J, Magana AA, Cerruti N, McFerrin J, Koike S, Nguyen T, Raber J, Quinn JF, Maier CS, Soumyanath A. Amelioration of age-related cognitive decline and anxiety in mice by Centella asiatica extract varies by sex, dose and mode of administration. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.01.23.576700. [PMID: 38328129 PMCID: PMC10849617 DOI: 10.1101/2024.01.23.576700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2024]
Abstract
We have previously reported that a water extract (CAW) of the Ayurvedic plant Centella asiatica administered in drinking water can improve cognitive deficits in mouse models of aging and neurodegenerative diseases. Here we compared the effects of CAW administered in drinking water or the diet on cognition, measures of anxiety and depression-like behavior in healthy aged mice. Three- and eighteen-month-old male and female C57BL6 mice were administered rodent AIN-93M diet containing CAW (0, 0.2, 0.5 or 1% w/w) to provide 0, 200 mg/kg/d, 500 mg/kg/d or 1000 mg/kg/d for a total of 5 weeks. An additional group of eighteen-month-old mice were treated with CAW (10 mg/mL) in their drinking water for a total of five weeks to deliver the same exposure of CAW as the highest dietary dose (1000 mg/kg/d). CAW doses delivered were calculated based on food and water consumption measured in previous experiments. In the fourth and fifth weeks, mice underwent behavioral testing of cognition, anxiety and depression (n=12 of each sex per treatment group in each test). Aged mice of both sexes showed cognitive deficits relative to young mice while only female aged mice showed increased anxiety compared to the young female mice and no differences in depression were observed between the different ages. CAW (1000 mg/kg/d) in the drinking water improved deficits in aged mice in learning, executive function and recognition memory in both sexes and attenuated the increased measures of anxiety observed in the aged female mice. However, CAW in the diet only improved executive function in aged mice at the highest dose (1000 mg/kg/d) in both sexes and did so less robustly than when given in the water. There were no effects of CAW on depression-like behavior in aged animals regardless of whether it was administered in the diet or the water. These results suggest that CAW can ameliorate age-related changes in measures of anxiety and cognition and that the mode of administration is important for the effects of CAW on resilience to these age-related changes.
Collapse
|
8
|
Suganuma T, Hatori S, Chen CK, Hori S, Kanuka M, Liu CY, Tatsuzawa C, Yanagisawa M, Hayashi Y. Caffeoylquinic Acid Mitigates Neuronal Loss and Cognitive Decline in 5XFAD Mice Without Reducing the Amyloid-β Plaque Burden. J Alzheimers Dis 2024; 99:1285-1301. [PMID: 38788074 DOI: 10.3233/jad-240033] [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] [Indexed: 05/26/2024]
Abstract
Background Caffeoylquinic acid (CQA), which is abundant in coffee beans and Centella asiatica, reportedly improves cognitive function in Alzheimer's disease (AD) model mice, but its effects on neuroinflammation, neuronal loss, and the amyloid-β (Aβ) plaque burden have remained unclear. Objective To assess the effects of a 16-week treatment with CQA on recognition memory, working memory, Aβ levels, neuronal loss, neuroinflammation, and gene expression in the brains of 5XFAD mice, a commonly used mouse model of familial AD. Methods 5XFAD mice at 7 weeks of age were fed a 0.8% CQA-containing diet for 4 months and then underwent novel object recognition (NOR) and Y-maze tests. The Aβ levels and plaque burden were analyzed by enzyme-linked immunosorbent assay and immunofluorescent staining, respectively. Immunostaining of markers of mature neurons, synapses, and glial cells was analyzed. AmpliSeq transcriptome analysis and quantitative reverse-transcription-polymerase chain reaction were performed to assess the effect of CQA on gene expression levels in the cerebral cortex of the 5XFAD mice. Results CQA treatment for 4 months improved recognition memory and ameliorated the reduction of mature neurons and synaptic function-related gene mRNAs. The Aβ levels, plaque burden, and glial markers of neuroinflammation seemed unaffected. Conclusions These findings suggest that CQA treatment mitigates neuronal loss and improves cognitive function without reducing Aβ levels or neuroinflammation. Thus, CQA is a potential therapeutic compound for AD, improving cognitive function via as-yet unknown mechanisms independent of reductions in Aβ or neuroinflammation.
Collapse
Affiliation(s)
- Takaya Suganuma
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Biological Science Research Laboratories, Kao Corporation, Ichikai, Japan
| | - Sena Hatori
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Chung-Kuan Chen
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Satoshi Hori
- Biological Science Research Laboratories, Kao Corporation, Ichikai, Japan
| | - Mika Kanuka
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Chih-Yao Liu
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Chika Tatsuzawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
| | - Masashi Yanagisawa
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Life Science Center for Survival Dynamics (TARA), University of Tsukuba, Tsukuba, Japan
- R&D Center for Frontiers of Mirai in Policy and Technology (F-MIRAI), University of Tsukuba, Tsukuba, Japan
| | - Yu Hayashi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Japan
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Kundu P, Yasuhara K, Brandes MS, Zweig JA, Neff CJ, Holden S, Kessler K, Matsumoto S, Offner H, Waslo CS, Vandenbark A, Soumyanath A, Sherman LS, Raber J, Gray NE, Spain RI. Centella asiatica promotes antioxidant gene expression and mitochondrial oxidative respiration in experimental autoimmune encephalomyelitis. RESEARCH SQUARE 2023:rs.3.rs-3393042. [PMID: 37886497 PMCID: PMC10602085 DOI: 10.21203/rs.3.rs-3393042/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2023]
Abstract
Centella asiatica (Centella) is a traditional botanical medicine that shows promise in treating dementia based on behavioral alterations seen in animal models of aging and cognitive dysfunction. In order to determine if Centella could similarly improve cognitive function and reduce disease burden in multiple sclerosis (MS), we tested its effects in the neuroinflammatory experimental autoimmune encephalomyelitis (EAE) model of MS. In two independent experiments, C57BL/6J mice were treated following induction of EAE with either a standardized water extract of Centella (CAW) or placebo for 2 weeks. At the dosing schedule and concentrations tested, CAW did not improve behavioral performance, EAE motor disability, or degrees of demyelination. However, CAW-treated mice demonstrated increases in nuclear factor (erythroid-derived 2)-like 2 and other antioxidant response element genes, and increases in mitochondrial respiratory activity. Caw also decreased spinal cord inflammation. Our findings indicate that CAW can increase antioxidant gene expression and mitochondrial respiratory activity in mice with EAE, supporting investigation of the clinical effects of CAW in people with MS.
Collapse
|
10
|
Wei T, Li JD, Wang YJ, Zhao W, Duan F, Wang Y, Xia LL, Jiang ZB, Song X, Zhu YQ, Shao WY, Wang Z, Bi KS, Li H, Zhang XC, Jiao DL. p-Nrf2/HO-1 Pathway Involved in Methamphetamine-induced Executive Dysfunction through Endoplasmic Reticulum Stress and Apoptosis in the Dorsal Striatum. Neurotox Res 2023; 41:446-458. [PMID: 37199892 DOI: 10.1007/s12640-023-00650-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Revised: 04/25/2023] [Accepted: 05/04/2023] [Indexed: 05/19/2023]
Abstract
Methamphetamine (METH) abuse is known to cause executive dysfunction. However, the molecular mechanism underlying METH induced executive dysfunction remains unclear. Go/NoGo experiment was performed in mice to evaluate METH-induced executive dysfunction. Immunoblot analysis of Nuclear factor-E2-related factor 2 (Nrf2), phosphorylated Nrf2 (p-Nrf2), heme-oxygenase-1 (HO-1), Glucose Regulated Protein 78(GRP78), C/EBP homologous protein (CHOP), Bcl-2, Bax and Caspase3 was performed to evaluate the levels of oxidative stress, endoplasmic reticulum (ER) stress and apoptosis in the dorsal striatum (Dstr). Malondialdehyde (MDA) levels and glutathione peroxidase (GSH-Px) activity was conducted to evaluate the level of oxidative stress. TUNEL staining was conducted to detect apoptotic neurons. The animal Go/NoGo testing confirmed that METH abuse impaired the inhibitory control ability of executive function. Meanwhile, METH down-regulated the expression of p-Nrf2, HO-1 and GSH-Px and activated ER stress and apoptosis in the Dstr. Microinjection of Tert-butylhydroxyquinone (TBHQ), an Nrf2 agonist, into the Dstr increased the expression of p-Nrf2, HO-1, and GSH-Px, ameliorated ER stress, apoptosis and executive dysfunction caused by METH. Our results indicated that the p-Nrf2/HO-1 pathway was potentially involved in mediating methamphetamine-induced executive dysfunction by inducing endoplasmic reticulum stress and apoptosis in the dorsal striatum.
Collapse
Affiliation(s)
- Tao Wei
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
- Huainan First People's Hospital, Huainan, 232007, Anhui, China
| | - Jun-Da Li
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yu-Jing Wang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Wei Zhao
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Fan Duan
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yan Wang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Ling-Ling Xia
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Zhao-Bin Jiang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Xun Song
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yu-Qiong Zhu
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Wen-Yi Shao
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Ze Wang
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Kang-Sheng Bi
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Hui Li
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Xiao-Chu Zhang
- CAS Key Laboratory of Brain Function and Disease and School of Life Sciences, University of Science and Technology of China, Hefei, 230027, Anhui, China.
| | - Dong-Liang Jiao
- School of Mental Health, Bengbu Medical College, Bengbu, 233030, Anhui, China.
| |
Collapse
|
11
|
Qiao J, Wang C, Chen Y, Yu S, Liu Y, Yu S, Jiang L, Jin C, Wang X, Zhang P, Zhao D, Wang J, Liu M. Herbal/Natural Compounds Resist Hallmarks of Brain Aging: From Molecular Mechanisms to Therapeutic Strategies. Antioxidants (Basel) 2023; 12:antiox12040920. [PMID: 37107295 PMCID: PMC10136184 DOI: 10.3390/antiox12040920] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 03/30/2023] [Accepted: 04/10/2023] [Indexed: 04/29/2023] Open
Abstract
Aging is a complex process of impaired physiological integrity and function, and is associated with increased risk of cardiovascular disease, diabetes, neurodegeneration, and cancer. The cellular environment of the aging brain exhibits perturbed bioenergetics, impaired adaptive neuroplasticity and flexibility, abnormal neuronal network activity, dysregulated neuronal Ca2+ homeostasis, accumulation of oxidatively modified molecules and organelles, and clear signs of inflammation. These changes make the aging brain susceptible to age-related diseases, such as Alzheimer's and Parkinson's diseases. In recent years, unprecedented advances have been made in the study of aging, especially the effects of herbal/natural compounds on evolutionarily conserved genetic pathways and biological processes. Here, we provide a comprehensive review of the aging process and age-related diseases, and we discuss the molecular mechanisms underlying the therapeutic properties of herbal/natural compounds against the hallmarks of brain aging.
Collapse
Affiliation(s)
- Juhui Qiao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chenxi Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Yu Chen
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shuang Yu
- School of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Ying Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shiting Yu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Leilei Jiang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Chenrong Jin
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Xinran Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Peiguang Zhang
- Changchun Institute of Optics, Fine Mechanics and Physics, Chinese Academy of Sciences, Changchun 130033, China
| | - Daqing Zhao
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiawen Wang
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
- Division of Cardiovascular Medicine, Department of Medicine, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Meichen Liu
- Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| |
Collapse
|
12
|
Quinn JF, Kelly MJ, Harris CJ, Hack W, Gray NE, Kulik V, Bostick Z, Brumbach BH, Copenhaver PF. The novel estrogen receptor modulator STX attenuates Amyloid-β neurotoxicity in the 5XFAD mouse model of Alzheimer's disease. Neurobiol Dis 2022; 174:105888. [PMID: 36209948 PMCID: PMC10108899 DOI: 10.1016/j.nbd.2022.105888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 09/09/2022] [Accepted: 10/05/2022] [Indexed: 11/21/2022] Open
Abstract
Based on previous evidence that the non-steroidal estrogen receptor modulator STX mitigates the effects of neurotoxic Amyloid-β (Aβ) in vitro, we have evaluated its neuroprotective benefits in a mouse model of Alzheimer's disease. Cohorts of 5XFAD mice, which begin to accumulate cerebral Aβ at two months of age, were treated with orally-administered STX starting at 6 months of age for two months. After behavioral testing to evaluate cognitive function, biochemical and immunohistochemical assays were used to analyze key markers of mitochondrial function and synaptic integrity. Oral STX treatment attenuated Aβ-associated mitochondrial toxicity and synaptic toxicity in the brain, as previously documented in cultured neurons. STX also moderately improved spatial memory in 5XFAD mice. In addition, STX reduced markers for reactive astrocytosis and microgliosis surrounding amyloid plaques, and also unexpectedly reduced overall levels of cerebral Aβ in the brain. The neuroprotective effects of STX were more robust in females than in males. These results suggest that STX may have therapeutic potential in Alzheimer's Disease.
Collapse
Affiliation(s)
- Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States of America; Parkinson's Disease Research, Education, and Clinical Center, Portland Veterans Affairs Medical Center, Portland, OR, United States of America.
| | - Martin J Kelly
- Department of Chemical Physiology and Biochemistry, OHSU, Portland, OR, United States of America
| | - Christopher J Harris
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States of America
| | - Wyatt Hack
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States of America
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States of America
| | - Veronika Kulik
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States of America
| | - Zoe Bostick
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR, United States of America
| | - Barbara H Brumbach
- Biostatistics and Design Program, OHSU-PSU School of Public Health, Portland, OR, United States of America
| | - Philip F Copenhaver
- Department of Cell, Developmental and Cancer Biology, OHSU, Portland, OR, United States of America
| |
Collapse
|
13
|
The Role of the NRF2 Pathway in Maintaining and Improving Cognitive Function. Biomedicines 2022; 10:biomedicines10082043. [PMID: 36009590 PMCID: PMC9405981 DOI: 10.3390/biomedicines10082043] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/10/2022] [Accepted: 08/17/2022] [Indexed: 11/24/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a redox-sensitive transcription factor that binds to the antioxidant response element consensus sequence, decreasing reactive oxygen species and regulating the transcription of a wide array of genes, including antioxidant and detoxifying enzymes, regulating genes involved in mitochondrial function and biogenesis. Moreover, NRF2 has been shown to directly regulate the expression of anti-inflammatory mediators reducing the expression of pro-inflammatory cytokines. In recent years, attention has turned to the role NRF2 plays in the brain in different diseases such Alzheimer’s disease, Parkinson’s disease, Huntington’s disease and others. This review focused on the evidence, derived in vitro, in vivo and from clinical trials, supporting a role for NRF2 activation in maintaining and improving cognitive function and how its activation can be used to elicit neuroprotection and lead to cognitive enhancement. The review also brings a critical discussion concerning the possible prophylactic and/or therapeutic use of NRF2 activators in treating cognitive impairment-related conditions.
Collapse
|
14
|
Therapeutic Potential of Different Natural Products for the Treatment of Alzheimer’s Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:6873874. [PMID: 35910833 PMCID: PMC9337964 DOI: 10.1155/2022/6873874] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 07/09/2022] [Indexed: 12/03/2022]
Abstract
A high incidence of dementia (60–80%) and a high rate of memory loss are two of the most common symptoms of Alzheimer's disease (AD), which affects the elderly. Researchers have recommended that traditional Chinese medicine (TCM) and Indian medicines can be used to prevent and cure AD. Several studies have linked neuroinflammation linked to amyloid-β (Aβ) deposition in the brain to the pathophysiology of neurodegenerative disorders. As a result, more research is needed to determine the role of inflammation in neurodegeneration. Increased microglial activation, cytokine production, reactive oxygen species (ROS), and nuclear factor kappa B (NF-κB) all play a role in the inflammatory process of AD. This review focuses on the role of neuroinflammation in neuroprotection and the molecular processes used by diverse natural substances, phytochemicals, and herbal formulations in distinct signaling pathways. Currently, researchers are focusing on pharmacologically active natural compounds with the anti-neuroinflammatory potential, making them a possible contender for treating AD. Furthermore, the researchers investigated the limits of past studies on TCM, Indian Ayurveda, and AD. Numerous studies have been carried out to examine the effects of medicinal whole-plant extracts on AD. Clinical investigations have shown that lignans, flavonoids, tannins, polyphenols, triterpenoids, sterols, and alkaloids have anti-inflammatory, antiamyloidogenic, anticholinesterase, and antioxidant properties. This review summarizes information about numerous medicinal plants and isolated compounds used in the treatment of AD and a list of further references.
Collapse
|
15
|
Wright KM, Bollen M, David J, Speers AB, Brandes MS, Gray NE, Alcázar Magaña A, McClure C, Stevens JF, Maier CS, Quinn JF, Soumyanath A. Pharmacokinetics and Pharmacodynamics of Key Components of a Standardized Centella asiatica Product in Cognitively Impaired Older Adults: A Phase 1, Double-Blind, Randomized Clinical Trial. Antioxidants (Basel) 2022; 11:215. [PMID: 35204098 PMCID: PMC8868383 DOI: 10.3390/antiox11020215] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/24/2022] Open
Abstract
Centella asiatica is reputed in Eastern medicine to improve cognitive function in humans. Preclinical studies have demonstrated that aqueous extracts of C. asiatica improve cognition in mouse models of aging and Alzheimer's disease (AD) through the modulation of mitochondrial biogenesis and nuclear factor-erythroid-2-related factor 2 (Nrf2)-dependent antioxidant response genes. This randomized, double-blind, crossover Phase I trial explored the oral bioavailability and pharmacokinetics of key compounds from two doses (2 g and 4 g) of a standardized C. asiatica aqueous extract product (CAP), over 10 h, in four mildly demented older adults on cholinesterase inhibitor therapy. The analysis focused on triterpenes (TTs) and caffeoylquinic acids (CQAs), which are known to contribute to C. asiatica's neurological activity. The acute safety of CAP and the effects on NRF2 gene expression in peripheral blood mononuclear cells were evaluated. Single administration of 2 g or 4 g of CAP was safe and well-tolerated. The TT aglycones, asiatic acid and madecassic acid, were identified in plasma and urine, while the parent glycosides, asiaticoside and madecassoside, although abundant in CAP, were absent in plasma and had limited renal excretion. Similarly, mono- and di-CQAs showed delayed absorption and limited presence in plasma or urine, while the putative metabolites of these compounds showed detectable plasma pharmacokinetic profiles and urinary excretion. CAP elicited a temporal change in NRF2 gene expression, mirroring the TT aglycone's pharmacokinetic curve in a paradoxical dose-dependent manner. The oral bioavailability of active compounds or their metabolites, NRF2 target engagement, and the acute safety and tolerability of CAP support the validity of using CAP in future clinical studies.
Collapse
Affiliation(s)
- Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Melissa Bollen
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Jason David
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Alex B. Speers
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Mikah S. Brandes
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA;
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Christine McClure
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Jan F. Stevens
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA;
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
- Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| |
Collapse
|
16
|
Wright KM, McFerrin J, Alcázar Magaña A, Roberts J, Caruso M, Kretzschmar D, Stevens JF, Maier CS, Quinn JF, Soumyanath A. Developing a Rational, Optimized Product of Centella asiatica for Examination in Clinical Trials: Real World Challenges. Front Nutr 2022; 8:799137. [PMID: 35096945 PMCID: PMC8797052 DOI: 10.3389/fnut.2021.799137] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/21/2021] [Indexed: 11/13/2022] Open
Abstract
Botanical products are frequently sold as dietary supplements and their use by the public is increasing in popularity. However, scientific evaluation of their medicinal benefits presents unique challenges due to their chemical complexity, inherent variability, and the involvement of multiple active components and biological targets. Translation away from preclinical models, and developing an optimized, reproducible botanical product for use in clinical trials, presents particular challenges for phytotherapeutic agents compared to single chemical entities. Common deficiencies noted in clinical trials of botanical products include limited characterization of the product tested, inadequate placebo control, and lack of rationale for the type of product tested, dose used, outcome measures or even the study population. Our group has focused on the botanical Centella asiatica due to its reputation for enhancing cognition in Eastern traditional medicine systems. Our preclinical studies on a Centella asiatica water extract (CAW) and its bioactive components strongly support its potential as a phytotherapeutic agent for cognitive decline in aging and Alzheimer's disease through influences on antioxidant response, mitochondrial activity, and synaptic density. Here we describe our robust, scientific approach toward developing a rational phytotherapeutic product based on Centella asiatica for human investigation, addressing multiple factors to optimize its valid clinical evaluation. Specific aspects covered include approaches to identifying an optimal dose range for clinical assessment, design and composition of a dosage form and matching placebo, sourcing appropriate botanical raw material for product manufacture (including the evaluation of active compounds and contaminants), and up-scaling of laboratory extraction methods to available current Good Manufacturing Practice (cGMP) certified industrial facilities. We also address the process of obtaining regulatory approvals to proceed with clinical trials. Our study highlights the complexity of translational research on botanicals and the importance of identifying active compounds and developing sound analytical and bioanalytical methods for their determination in botanical materials and biological samples. Recent Phase I pharmacokinetic studies of our Centella asiatica product in humans (NCT03929250, NCT03937908) have highlighted additional challenges associated with designing botanical bioavailability studies, including specific dietary considerations that need to be considered.
Collapse
Affiliation(s)
- Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | | | - Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | | | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Doris Kretzschmar
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR, United States
| | - Jan F. Stevens
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, OR, United States
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
17
|
NRF2 Activation Ameliorates Oxidative Stress and Improves Mitochondrial Function and Synaptic Plasticity, and in A53T α-Synuclein Hippocampal Neurons. Antioxidants (Basel) 2021; 11:antiox11010026. [PMID: 35052530 PMCID: PMC8772776 DOI: 10.3390/antiox11010026] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 12/20/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
In Parkinson's disease (PD), brain oxidative stress and mitochondrial dysfunction contribute to neuronal loss as well as motor and cognitive deficits. The transcription factor NRF2 has emerged as a promising therapeutic target in PD because it sits at the intersection of antioxidant and mitochondrial pathways. Here, we investigate the effects of modulating NRF2 activity in neurons isolated from a A53T α-synuclein (A53TSyn) mouse model of synucleinopathy. Embryonic hippocampal neurons were isolated from A53TSyn mice and their wild type (WT) littermates. Neurons were treated with either the NRF2 activator dimethyl fumarate (DMF) or the NRF2 inhibitor ML385. Reactive oxygen species (ROS), dendritic arborization and dendritic spine density were quantified. Mitochondrial bioenergetics were also profiled in these neurons. A53TSyn neurons had increased ROS and reduced basal and maximal mitochondrial respiration relative to WT neurons. A53TSyn neurons also displayed decreased dendritic arborization and reduced spine density. Treatment with DMF reduced ROS levels and improved both mitochondrial function and arborization, while inhibition of NRF2 with ML385 exacerbated these endpoints. Modulation of NRF2 activity had a significant effect on mitochondrial function, oxidative stress, and synaptic plasticity in A53TSyn neurons. These data suggest that NRF2 may be a viable target for therapeutic interventions in PD.
Collapse
|
18
|
Khorani M, Bobe G, Matthews DG, Magana AA, Caruso M, Gray NE, Quinn JF, Stevens JF, Soumyanath A, Maier CS. The Impact of the hAPP695SW Transgene and Associated Amyloid-β Accumulation on Murine Hippocampal Biochemical Pathways. J Alzheimers Dis 2021; 85:1601-1619. [DOI: 10.3233/jad-215084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Background: Alzheimer’s disease (AD) is a neurodegenerative disease characterized by the accumulation of amyloid-β (Aβ) peptide in the brain. Objective: Gain a better insight into alterations in major biochemical pathways underlying AD. Methods: We compared metabolomic profiles of hippocampal tissue of 20-month-old female Tg2576 mice expressing the familial AD-associated hAPP695SW transgene with their 20-month-old wild type female littermates. Results: The hAPP695SW transgene causes overproduction and accumulation of Aβ in the brain. Out of 180 annotated metabolites, 54 metabolites differed (30 higher and 24 lower in Tg2576 versus wild-type hippocampal tissue) and were linked to the amino acid, nucleic acid, glycerophospholipid, ceramide, and fatty acid metabolism. Our results point to 1) heightened metabolic activity as indicated by higher levels of urea, enhanced fatty acid β-oxidation, and lower fatty acid levels; 2) enhanced redox regulation; and 3) an imbalance of neuro-excitatory and neuro-inhibitory metabolites in hippocampal tissue of aged hAPP695SW transgenic mice. Conclusion: Taken together, our results suggest that dysregulation of multiple metabolic pathways associated with a concomitant shift to an excitatory-inhibitory imbalance are contributing mechanisms of AD-related pathology in the Tg2576 mouse.
Collapse
Affiliation(s)
- Mona Khorani
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Gerd Bobe
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Armando Alcazar Magana
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
| |
Collapse
|
19
|
Speers AB, García-Jaramillo M, Feryn A, Matthews DG, Lichtenberg T, Caruso M, Wright KM, Quinn JF, Stevens JF, Maier CS, Soumyanath A, Gray NE. Centella asiatica Alters Metabolic Pathways Associated With Alzheimer's Disease in the 5xFAD Mouse Model of ß-Amyloid Accumulation. Front Pharmacol 2021; 12:788312. [PMID: 34975484 PMCID: PMC8717922 DOI: 10.3389/fphar.2021.788312] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Centella asiatica is an herb used in Ayurvedic and traditional Chinese medicine for its beneficial effects on brain health and cognition. Our group has previously shown that a water extract of Centella asiatica (CAW) elicits cognitive-enhancing effects in animal models of aging and Alzheimer's disease, including a dose-related effect of CAW on memory in the 5xFAD mouse model of ß-amyloid accumulation. Here, we endeavor to elucidate the mechanisms underlying the effects of CAW in the brain by conducting a metabolomic analysis of cortical tissue from 5xFAD mice treated with increasing concentrations of CAW. Tissue was collected from 8-month-old male and female 5xFAD mice and their wild-type littermates treated with CAW (0, 200, 500, or 1,000 mg/kg/d) dissolved in their drinking water for 5 weeks. High-performance liquid chromatography coupled to high-resolution mass spectrometry analysis was performed and relative levels of 120 annotated metabolites were assessed in the treatment groups. Metabolomic analysis revealed sex differences in the effect of the 5xFAD genotype on metabolite levels compared to wild-type mice, and variations in the metabolomic response to CAW depending on sex, genotype, and CAW dose. In at least three of the four treated groups (5xFAD or wild-type, male or female), CAW (500 mg/kg/d) significantly altered metabolic pathways related to purine metabolism, nicotinate and nicotinamide metabolism, and glycerophospholipid metabolism. The results are in line with some of our previous findings regarding specific mechanisms of action of CAW (e.g., improving mitochondrial function, reducing oxidative stress, and increasing synaptic density). Furthermore, these findings provide new information about additional, potential mechanisms for the cognitive-enhancing effect of CAW, including upregulation of nicotinamide adenine dinucleotide in the brain and modulation of brain-derived neurotrophic factor. These metabolic pathways have been implicated in the pathophysiology of Alzheimer's disease, highlighting the therapeutic potential of CAW in this neurodegenerative disease.
Collapse
Affiliation(s)
- Alex B. Speers
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Manuel García-Jaramillo
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Alicia Feryn
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland, OR, United States
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Talia Lichtenberg
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, United States
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Claudia S. Maier
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
20
|
Buccellato FR, D’Anca M, Fenoglio C, Scarpini E, Galimberti D. Role of Oxidative Damage in Alzheimer's Disease and Neurodegeneration: From Pathogenic Mechanisms to Biomarker Discovery. Antioxidants (Basel) 2021; 10:antiox10091353. [PMID: 34572985 PMCID: PMC8471953 DOI: 10.3390/antiox10091353] [Citation(s) in RCA: 51] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 08/11/2021] [Accepted: 08/17/2021] [Indexed: 12/16/2022] Open
Abstract
Alzheimer’s disease (AD) is a neurodegenerative disorder accounting for over 50% of all dementia patients and representing a leading cause of death worldwide for the global ageing population. The lack of effective treatments for overt AD urges the discovery of biomarkers for early diagnosis, i.e., in subjects with mild cognitive impairment (MCI) or prodromal AD. The brain is exposed to oxidative stress as levels of reactive oxygen species (ROS) are increased, whereas cellular antioxidant defenses are decreased. Increased ROS levels can damage cellular structures or molecules, leading to protein, lipid, DNA, or RNA oxidation. Oxidative damage is involved in the molecular mechanisms which link the accumulation of amyloid-β and neurofibrillary tangles, containing hyperphosphorylated tau, to microglia response. In this scenario, microglia are thought to play a crucial role not only in the early events of AD pathogenesis but also in the progression of the disease. This review will focus on oxidative damage products as possible peripheral biomarkers in AD and in the preclinical phases of the disease. Particular attention will be paid to biological fluids such as blood, CSF, urine, and saliva, and potential future use of molecules contained in such body fluids for early differential diagnosis and monitoring the disease course. We will also review the role of oxidative damage and microglia in the pathogenesis of AD and, more broadly, in neurodegeneration.
Collapse
Affiliation(s)
- Francesca Romana Buccellato
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (E.S.); (D.G.)
- Correspondence: ; Tel.: +39-02 55033814
| | - Marianna D’Anca
- Fondazione IRCSS ca’ Granda, Ospedale Policlinico, 20122 Milano, Italy;
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, University of Milan, 20122 Milan, Italy;
| | - Elio Scarpini
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (E.S.); (D.G.)
- Fondazione IRCSS ca’ Granda, Ospedale Policlinico, 20122 Milano, Italy;
| | - Daniela Galimberti
- Department of Biomedical, Surgical and Dental Sciences, University of Milan, 20122 Milan, Italy; (E.S.); (D.G.)
- Fondazione IRCSS ca’ Granda, Ospedale Policlinico, 20122 Milano, Italy;
| |
Collapse
|
21
|
Experimental evidence and mechanism of action of some popular neuro-nutraceutical herbs. Neurochem Int 2021; 149:105124. [PMID: 34245808 DOI: 10.1016/j.neuint.2021.105124] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Revised: 06/28/2021] [Accepted: 06/30/2021] [Indexed: 12/12/2022]
Abstract
Brain and neuronal circuits constitute the most complex organ networks in human body. They not only control and coordinate functions of all other organs, but also represent one of the most-affected systems with stress, lifestyle and age. With global increase in aging populations, these neuropathologies have emerged as major concern for maintaining quality of life. Recent era has witnessed a surge in nutritional remediation of brain dysfunctions primarily by "nutraceuticals" that refer to functional foods and supplements with pharmacological potential. Specific dietary patterns with a balanced intake of carbohydrates, fatty acids, vitamins and micronutrients have also been ascertained to promote brain health. Dietary herbs and their phytochemicals with wide range of biological and pharmacological activities and minimal adverse effects have gained remarkable attention as neuro-nutraceuticals. Neuro-nutraceutical potentials of herbs are often expressed as effects on cognitive response, circadian rhythm, neuromodulatory, antioxidant and anti-inflammatory activities that are mediated by effects on gene expression, epigenetics, protein synthesis along with their turnover and metabolic pathways. Epidemiological and experimental evidence have implicated enormous applications of herbal supplementation in neurodegenerative and psychiatric disorders. The present review highlights the identification, experimental evidence and applications of some herbs including Bacopa monniera, Withania somnifera, Curcuma longa, Helicteres angustifolia, Undaria pinnatifida, Haematococcus pluvialis, and Vitis vinifera, as neuro-nutraceuticals.
Collapse
|
22
|
Buranasudja V, Rani D, Malla A, Kobtrakul K, Vimolmangkang S. Insights into antioxidant activities and anti-skin-aging potential of callus extract from Centella asiatica (L.). Sci Rep 2021; 11:13459. [PMID: 34188145 PMCID: PMC8241881 DOI: 10.1038/s41598-021-92958-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/18/2021] [Indexed: 12/17/2022] Open
Abstract
Formation of oxidative stress in dermal fibroblasts plays crucial roles in aging processes of skin. The use of phytochemicals that can promote capacity of fibroblasts to combat oxidative stress is an attractive strategy to prevent skin aging and promote skin beauty. Centella asiatica has been used to treat multitude of diseases for centuries. Previous investigations demonstrated that extracts from C. asiatica have a broad range of beneficial activities through their antioxidant activity. Hence, the extract from this medicinal plant could be a great candidate for anti-skin-aging agent. Callus culture offers a powerful platform for sustainable, rapid and large-scale production of phytochemicals to serve extensive demands of pharmaceutical and cosmeceutical industries. Here, we demonstrated the application of callus culture of Centella asiatica to produce bioactive metabolites. The 50% ethanolic extract of callus culture has distinctive features of chemical compositions and biological profiles. Information from HPTLC-DPPH and HPLC analysis suggested that the callus extract comprises distinctive antioxidant compounds, compared with those isolated from authentic plant. Moreover, results from cell culture experiment demonstrated that callus extract possesses promising antioxidant and anti-skin-aging activities. Pre-treatment with callus extract attenuated H2O2-induced-cytotoxicity on human dermal fibroblasts. The results from RT-qPCR clearly suggested that the upregulation of cellular antioxidant enzymes appeared to be major contributor for the protective effects of callus extract against oxidative stress. Moreover, supplementation with callus extract inhibited induction of matrix metalloprotease-9 following H2O2 exposure, suggesting its potential anti-skin-aging activity. Our results demonstrate the potential utility of C. asiatica callus extract as anti-skin-aging agent in cosmeceutical preparations.
Collapse
Affiliation(s)
- Visarut Buranasudja
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Dolly Rani
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Patumwan, Bangkok, 10330, Thailand
| | - Ashwini Malla
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Patumwan, Bangkok, 10330, Thailand.,Research Unit for Plant-Produced Pharmaceuticals, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Khwanlada Kobtrakul
- Graduate Program in Pharmaceutical Science and Technology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand
| | - Sornkanok Vimolmangkang
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Chulalongkorn University, 254 Phayathai Road, Patumwan, Bangkok, 10330, Thailand. .,Research Unit for Natural Product Biotechnology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, 10330, Thailand.
| |
Collapse
|
23
|
Wong JH, Barron AM, Abdullah JM. Mitoprotective Effects of Centella asiatica (L.) Urb.: Anti-Inflammatory and Neuroprotective Opportunities in Neurodegenerative Disease. Front Pharmacol 2021; 12:687935. [PMID: 34267660 PMCID: PMC8275827 DOI: 10.3389/fphar.2021.687935] [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: 03/30/2021] [Accepted: 06/17/2021] [Indexed: 11/13/2022] Open
Abstract
Natural products remain a crucial source of drug discovery for accessible and affordable solutions for healthy aging. Centella asiatica (L.) Urb. (CA) is an important medicinal plant with a wide range of ethnomedicinal uses. Past in vivo and in vitro studies have shown that the plant extract and its key components, such as asiatic acid, asiaticoside, madecassic acid and madecassoside, exhibit a range of anti-inflammatory, neuroprotective, and cognitive benefits mechanistically linked to mitoprotective and antioxidant properties of the plant. Mitochondrial dysfunction and oxidative stress are key drivers of aging and neurodegenerative disease, including Alzheimer’s disease and Parkinson’s disease. Here we appraise the growing body of evidence that the mitoprotective and antioxidative effects of CA may potentially be harnessed for the treatment of brain aging and neurodegenerative disease.
Collapse
Affiliation(s)
- Jia Hui Wong
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Anna M Barron
- Neurobiology of Aging and Disease Laboratory, Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore
| | - Jafri Malin Abdullah
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia.,Brain & Behaviour Cluster and Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
24
|
Lu J, Chen C, Gai R, Qiu H, Wu Y, He Q, Yang X. Protective effects and possible mechanisms of Centella asiatica (L.) urban extract against acute and chronic liver injury: Evidence from in vivo and in vitro studies. Phytother Res 2021; 35:2785-2796. [PMID: 33462870 DOI: 10.1002/ptr.7024] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/10/2020] [Accepted: 01/04/2021] [Indexed: 11/10/2022]
Abstract
Drug-induced liver injury (DILI) has become a significant health care problem worldwide. Centella asiatica (L.) urban was traditionally used to prevent or treat various diseases, yet whether it works on hepatic injury remains unclear. In this study, multiple experimental models with different damage degrees and types of liver injury have been established to evaluate the hepatoprotective effects of an n-butanol extract of Centella asiatica (CA-BU). Our results revealed that CA-BU improved hepatocyte L02 cells survival from H2 O2 -induced oxidative damage in a concentration-dependent manner. We further verified the hepatoprotective effects of CA-BU in mice models of acetaminophen-induced acute liver injury (one of the most common DILIs clinically) and CCl4 -induced acute chemical liver injury, and a rat model of chronic alcoholic steatohepatitis. Furthermore, network pharmacology approaches were performed to explore the underlying mechanisms, and we predicted AKT1, EGFR, VEGFA, and STAT3 as the potential therapeutic targets. In follow-up studies, we will focus on targets verification and provide a deeper insight into the mechanisms of CA-BU against liver damage. Finally, we hope that these findings will provide new ideas and insights for the treatment of acute or chronic liver injury in the clinic.
Collapse
Affiliation(s)
- Jiabin Lu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Chao Chen
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Renhua Gai
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Huacheng Qiu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Yue Wu
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Qiaojun He
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Xiaochun Yang
- Center for Drug Safety Evaluation and Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| |
Collapse
|
25
|
Zweig JA, Brandes MS, Brumbach BH, Caruso M, Wright KM, Quinn JF, Soumyanath A, Gray NE. Loss of NRF2 accelerates cognitive decline, exacerbates mitochondrial dysfunction, and is required for the cognitive enhancing effects of Centella asiatica during aging. Neurobiol Aging 2021; 100:48-58. [PMID: 33486357 PMCID: PMC7920997 DOI: 10.1016/j.neurobiolaging.2020.11.019] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Revised: 11/30/2020] [Accepted: 11/30/2020] [Indexed: 02/07/2023]
Abstract
The water extract of Centella asiatica (CAW) improves cognitive and mitochondrial function and activates the nuclear factor erythroid 2-related factor 2 (NRF2) regulated antioxidant response pathway in aged mice. Here we investigate whether NRF2 activation is required for the cognitive and mitochondrial effects of prolonged CAW exposure during aging. Five-month-old NRF2 knockout (NRF2KO) and wild-type mice were treated with CAW for 1, 7, or 13 months. Each cohort underwent cognitive testing and hippocampal mitochondrial analyses. Age-related cognitive decline was accelerated in NRF2KO mice and while CAW treatment improved cognitive performance in wild-type mice, it had no effect on NRF2KO animals. Hippocampal mitochondrial function also declined further with age in NRF2KO mice and greater hippocampal mitochondrial dysfunction was associated with poorer cognitive performance in both genotypes. Long-term CAW treatment did not affect mitochondrial endpoints in animals of either genotype. These data indicate that loss of NRF2 results in accelerated age-related cognitive decline and worsened mitochondrial deficits. NRF2 also appears to be required for the cognitive enhancing effects of CAW during aging.
Collapse
Affiliation(s)
- Jonathan A Zweig
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Mikah S Brandes
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Barbara H Brumbach
- Biostatistics & Design Program, Oregon Health & Science University, Portland, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Kirsten M Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA; Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
26
|
Hannan MA, Haque MN, Munni YA, Oktaviani DF, Timalsina B, Dash R, Afrin T, Moon IS. Centella asiatica promotes early differentiation, axodendritic maturation and synaptic formation in primary hippocampal neurons. Neurochem Int 2021; 144:104957. [PMID: 33444677 DOI: 10.1016/j.neuint.2021.104957] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 12/21/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Centella asiatica is a 'medhya-rasayana (nootrophic or memory booster)' herb that has been indicated in Ayurveda for improving memory function and treating dementia disorders. Although the neuroprotective effects of C. asiatica have been reported in earlier studies, the information on whether this nootropic herb could promote early differentiation and development of axon and dendrites in primary hippocampal neurons is currently limited. THE AIM OF THE STUDY To investigate the effects of C. asiatica and asiatic acid, one of the principal active constituents of C. asiatica, on the various stages of neuronal polarity, including early neuronal differentiation, axonal outgrowth, dendritic arborization, axonal maturation, and synaptic formation. MATERIALS AND METHODS Embryonic rat hippocampal neurons were incubated with C. asiatica leaf extract (CAE) or asiatic acid. After an indicated time, neurons were fixed and immunolabeled to visualize the neuronal morphology. Morphometric analyses for early neuronal differentiation, axonal and dendritic maturation and synaptogenesis were performed using Image J software. Neuronal viability was determined using trypan blue exclusion assay. RESULTS CAE at varying concentrations ranging from 3.75 to 15 μg/mL enhanced neurite outgrowth with the highest optimal concentration of 7.5 μg/mL. The effects of CAE commenced immediately after cell seeding, as indicated by its accelerating effect on neuronal differentiation. Subsequently, CAE significantly elaborated dendritic and axonal morphology and facilitated synapse formation. Asiatic acid also facilitated neurite outgrowth, but to a lesser extent than CAE. CONCLUSION These findings revealed that CAE exerted its modulatory effects in every stage of neuronal development, supporting its previously claimed neurotrophic function and suggest that this natural nootropic and its active component asiatic acid can be further investigated to explore a promising solution for degenerative brain disorders and injuries.
Collapse
Affiliation(s)
- Md Abdul Hannan
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea; Department of Biochemistry and Molecular Biology, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Md Nazmul Haque
- Department of Fisheries Biology and Genetics, Patuakhali Science and Technology University, Patuakhali, 8602, Bangladesh
| | - Yeasmin Akter Munni
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Diyah Fatimah Oktaviani
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Binod Timalsina
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Raju Dash
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea
| | - Tania Afrin
- Interdisciplinary Institute for Food Security, Bangladesh Agricultural University, Mymensingh, 2202, Bangladesh
| | - Il Soo Moon
- Department of Anatomy, Dongguk University College of Medicine, Gyeongju, 38066, Republic of Korea.
| |
Collapse
|
27
|
Torbati FA, Ramezani M, Dehghan R, Amiri MS, Moghadam AT, Shakour N, Elyasi S, Sahebkar A, Emami SA. Ethnobotany, Phytochemistry and Pharmacological Features of Centella asiatica: A Comprehensive Review. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1308:451-499. [PMID: 33861456 DOI: 10.1007/978-3-030-64872-5_25] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Centella asiatica (CA) or Gotu cola is an herbal plant from the Apiaceae family with a long history of usage in different traditional medicines. It has long been used for the treatment of various ailments such as central nervous system (CNS), skin and gastrointestinal disorders especially in the Southeast Asia. This chapter focused on the phytochemical constituent and pharmacological activities of CA based on preclinical and clinical studies. Additionally, botanical description and distribution, traditional uses, interactions, and safety issues are reviewed. Electronic databases of Google Scholar, Scopus, PubMed, and Web of Science were searched to obtain relevant studies on the pharmacological activities of CA. Approximately, 124 chemical compounds including triterpenoids, polyphenolic compounds, and essential oils have been isolated and identified from CA. Ethnomedicinal applications of CA mostly include treatment of gastrointestinal diseases, wounds, nervous system disorders, circulatory diseases, skin problems, respiratory ailments, diabetes and sleep disorders in various ethnobotanical practices. Pharmacological studies revealed a wide range of beneficial effects of CA on CNS, cardiovascular, lung, liver, kidney, gastrointestinal, skin, and endocrine system. Among them, neuroprotective activity, wound healing and treatment of venous insufficiency, as well as antidiabetic activity seem to be more frequently reported. At the moment, considering various health benefits of CA, it is marketed as an oral supplement as well as a topical ingredient in some cosmetic products. Additional preclinical studies and particularly randomized controlled trials are needed to clarify the therapeutic roles of CA.
Collapse
Affiliation(s)
- Farshad Abedi Torbati
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mahin Ramezani
- Nanotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Reza Dehghan
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ali Tafazoli Moghadam
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Department of Clinical Pharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Neda Shakour
- Department of Medicinal Chemistry, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sepideh Elyasi
- Department of Clinical Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.,School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.,Polish Mother's Memorial Hospital Research Institute (PMMHRI), Lodz, Poland
| | - Seyed Ahmad Emami
- Department of Traditional Pharmacy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
28
|
Zweig JA, Brandes MS, Brumbach BH, Caruso M, Wright KM, Quinn JF, Soumyanath A, Gray NE. Prolonged Treatment with Centella asiatica Improves Memory, Reduces Amyloid-β Pathology, and Activates NRF2-Regulated Antioxidant Response Pathway in 5xFAD Mice. J Alzheimers Dis 2021; 81:1453-1468. [PMID: 33935097 PMCID: PMC10878128 DOI: 10.3233/jad-210271] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
BACKGROUND The medicinal herb Centella asiatica has been long been used for its neuroprotective and cognitive enhancing effects. We have previously shown that two weeks of treatment with a water extract of Centella asiatica (CAW) improves cognition and activates the endogenous antioxidant response pathway without altering amyloid-β (Aβ) plaque burden. OBJECTIVE Here, we assess the effect of long-term treatment of CAW in the 5xFAD mouse model of Aβ accumulation. METHODS Four-month-old 5xFAD mice were treated with CAW in their drinking water (2 g/L) for three months at which point they underwent cognitive testing as well as analysis of Aβ plaque levels and antioxidant and synaptic gene expression. In order to confirm the involvement of the antioxidant regulatory transcription factor NRF2 on the effects of CAW on synaptic plasticity, neurons isolated from 5xFAD mice were also treated with CAW and the targeted inhibitor ML385. RESULTS Three months of treatment with CAW improved spatial and contextual memory as well as executive function in 5xFAD mice. This improvement was accompanied by increased antioxidant gene expression and a decrease in Aβ plaque burden relative to untreated 5xFAD animals. In isolated neurons, treatment with ML385 blocked the effects of CAW on dendritic arborization and synaptic gene expression. CONCLUSION These results suggest that prolonged CAW exposure could be beneficial in Alzheimer's disease and that these effects likely involve NRF2 activation. Moreover, these findings suggest that targeting NRF2 itself may be a relevant therapeutic strategy for improving synaptic plasticity and cognitive function in Alzheimer's disease.
Collapse
Affiliation(s)
- Jonathan A. Zweig
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Mikah S. Brandes
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Barbara H. Brumbach
- Biostatistics & Design Program Core, Oregon Health & Science University, Portland, OR, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- Department of Neurology and Parkinson’s Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
29
|
Parodi-Rullán R, Sone JY, Fossati S. Endothelial Mitochondrial Dysfunction in Cerebral Amyloid Angiopathy and Alzheimer's Disease. J Alzheimers Dis 2020; 72:1019-1039. [PMID: 31306129 DOI: 10.3233/jad-190357] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is the most prevalent form of dementia. Cerebrovascular dysfunction is one of the earliest events in the pathogenesis of AD, as well as in vascular and mixed dementias. Cerebral amyloid angiopathy (CAA), the deposition of amyloid around cerebral vessels, is observed in up to 90% of AD patients and in approximately 50% of elderly individuals over 80 years of age. CAA is a strong contributor to vascular dysfunction in AD. CAA-laden brain vessels are characterized by dysfunctional hemodynamics and leaky blood-brain barrier (BBB), contributing to clearance failure and further accumulation of amyloid-β (Aβ) in the cerebrovasculature and brain parenchyma. Mitochondrial dysfunction is increasingly recognized as an important early initiator of the pathogenesis of AD and CAA. The objective of this review is to discuss the effects of Aβ on cerebral microvascular cell function, focusing on its impact on endothelial mitochondria. After introducing CAA and its etiology and genetic risk factors, we describe the pathological relationship between cerebrovascular amyloidosis and brain microvascular endothelial cell dysfunction, critically analyzing its roles in disease progression, hypoperfusion, and BBB integrity. Then, we focus on discussing the effect of Aβ challenge on endothelial mitochondrial dysfunction pathways, and their contribution to the progression of neurovascular dysfunction in AD and dementia. Finally, we report potential pharmacological and non-pharmacological mitochondria-targeted therapeutic strategies which may help prevent or delay cerebrovascular failure.
Collapse
Affiliation(s)
- Rebecca Parodi-Rullán
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Je Yeong Sone
- Department of Psychiatry, Center for Brain Health, NYU School of Medicine, New York, NY, USA
| | - Silvia Fossati
- Alzheimer's Center at Temple, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| |
Collapse
|
30
|
Matthews DG, Caruso M, Alcazar Magana A, Wright KM, Maier CS, Stevens JF, Gray NE, Quinn JF, Soumyanath A. Caffeoylquinic Acids in Centella asiatica Reverse Cognitive Deficits in Male 5XFAD Alzheimer's Disease Model Mice. Nutrients 2020; 12:E3488. [PMID: 33202902 PMCID: PMC7698091 DOI: 10.3390/nu12113488] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 11/04/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Centella asiatica (CA) is an edible plant and a popular botanical dietary supplement. It is reputed, in Ayurveda, to mitigate age-related cognitive decline. There is a considerable body of preclinical literature supporting CA's ability to improve learning and memory. This study evaluated the contribution of CA's triterpenes (TT), widely considered its active compounds, and caffeoylquinic acids (CQA) to the cognitive effects of CA water extract (CAW) in 5XFAD mice, a model of Alzheimer's disease. 5XFAD mice were fed a control diet alone, or one containing 1% CAW or compound groups (TT, CQA, or TT + CQA) equivalent to their content in 1% CAW. Wild-type (WT) littermates received the control diet. Conditioned fear response (CFR) was evaluated after 4.5 weeks. Female 5XFAD controls showed no deficit in CFR compared to WT females, nor any effects from treatment. In males, CFR of 5XFAD controls was attenuated compared to WT littermates (p = 0.005). 5XFAD males receiving CQA or TT + CQA had significantly improved CFR (p < 0.05) compared to 5XFAD male controls. CFR did not differ between 5XFAD males receiving treatment diets and WT males. These data confirm a role for CQA in CAW's cognitive effects.
Collapse
Affiliation(s)
- Donald G. Matthews
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Maya Caruso
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Armando Alcazar Magana
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA;
| | - Kirsten M. Wright
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA;
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA;
- Department of Pharmaceutical Sciences, College of Pharmacy, Oregon State University, Corvallis, OR 97331, USA
| | - Nora E. Gray
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| | - Joseph F. Quinn
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, School of Medicine, Oregon Health & Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (K.M.W.); (N.E.G.); (J.F.Q.)
| |
Collapse
|
31
|
Wright KM, Magana AA, Laethem RM, Moseley CL, Banks TT, Maier CS, Stevens JF, Quinn JF, Soumyanath A. Centella asiatica Water Extract Shows Low Potential for Cytochrome P450-Mediated Drug Interactions. Drug Metab Dispos 2020; 48:1053-1063. [PMID: 32581050 PMCID: PMC7543484 DOI: 10.1124/dmd.120.090860] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Centella asiatica (CA) shows considerable promise for development as a botanical drug for cognitive decline. Its primary bioactive components include triterpene glycosides asiaticoside and madecassoside and their corresponding aglycones asiatic acid and madecassic acid. Exploration of the bioactivity of CA's caffeoylquinic acids is ongoing. In this study, an aqueous extract of CA (CAW-R61J) was evaluated for drug interaction potential through inhibition or induction of P450 enzymes, as required by the US Food and Drug Administration. CAW-R61J was assessed for induction potential of CYP1A2, CYP2B6, and CYP3A4 using transporter-certified cryopreserved human hepatocytes in sandwich culture. Gene expression of these target P450s was quantified, and enzyme activities were determined to confirm gene expression results. No induction was observed up to 16.7 µg/ml CAW-R61J (equivalent to 1.1 µM asiaticoside, 0.8 µM madecassoside, 0.09 µM asiatic acid, and 0.12 µM madecassic acid). Reversible and time-dependent inhibitory effects of CAW-R61J on CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4/5 were evaluated using human liver microsomes. CAW-R61J showed weak reversible inhibition of most of the P450 forms tested, with the strongest being CYP2C9 (IC50 of 330 µg/ml). CAW-R61J (≤1000 µg/ml) was not a time-dependent inhibitor of any of these P450 enzymes. In summary, CAW-R61J had no, or only a weak impact, on P450 induction and inhibition in vitro. The clinical relevance of these results will depend on the in vivo concentration of CAW-R61J components achieved in humans. Plasma triterpene concentrations measured in our recent clinical studies suggest minimal risk of P450-mediated drug interactions by these components. SIGNIFICANCE STATEMENT: A preparation of Centella asiatica is currently under clinical development for the prevention or treatment of cognitive decline. The US Food and Drug Administration required an evaluation of its potential for drug interactions mediated through drug-metabolizing enzymes. This in vitro study revealed minimal induction or inhibition of a range of P450 enzymes, including CYP3A4, by the C. asiatica extract, suggesting a low potential for drug interactions modulated by P450 metabolism.
Collapse
Affiliation(s)
- Kirsten M Wright
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Armando Alcazar Magana
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Ronald M Laethem
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Caroline L Moseley
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Troy T Banks
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Claudia S Maier
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Jan F Stevens
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| |
Collapse
|
32
|
Sun B, Wu L, Wu Y, Zhang C, Qin L, Hayashi M, Kudo M, Gao M, Liu T. Therapeutic Potential of Centella asiatica and Its Triterpenes: A Review. Front Pharmacol 2020; 11:568032. [PMID: 33013406 PMCID: PMC7498642 DOI: 10.3389/fphar.2020.568032] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Accepted: 08/14/2020] [Indexed: 12/12/2022] Open
Abstract
Centella asiatica (also known as Centella asiatica (L.) Urb. or Gotu kola) is a traditional Chinese medicine with extensive medicinal value, which is commonly used in Southeast Asian countries. This study aimed to summarize the effects of C. asiatica and its main components on neurological diseases, endocrine diseases, skin diseases, cardiovascular diseases, gastrointestinal diseases, immune diseases, and gynecological diseases, as well as potential molecular mechanisms, to study the pathological mechanism of these diseases based on the changes at the molecular level. The results showed that C. asiatica and its triterpenoids had extensive beneficial effects on neurological and skin diseases, which were confirmed through clinical studies. They exhibited anti-inflammatory, anti-oxidative stress, anti-apoptotic effects, and improvement in mitochondrial function. However, further clinical studies are urgently required due to the low level of evidence and lack of patients.
Collapse
Affiliation(s)
- Boju Sun
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Lili Wu
- Key Laboratory of Health Cultivation of the Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - You Wu
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Chengfei Zhang
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Lingling Qin
- Technology Department, Beijing University of Chinese Medicine, Beijing, China
| | - Misa Hayashi
- School of Pharmaceutical Sciences, Mukogawa Women’s University, Hyogo, Japan
| | - Maya Kudo
- School of Pharmaceutical Sciences, Mukogawa Women’s University, Hyogo, Japan
| | - Ming Gao
- School of Pharmaceutical Sciences, Mukogawa Women’s University, Hyogo, Japan
| | - Tonghua Liu
- Second Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
33
|
Network Pharmacology Study of Heat-Clearing and Detoxifying Traditional Chinese Medicine for Alzheimer's Disease. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2020; 2020:7831675. [PMID: 32382304 PMCID: PMC7196989 DOI: 10.1155/2020/7831675] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 02/15/2020] [Accepted: 03/25/2020] [Indexed: 01/13/2023]
Abstract
This study aims to explore the possible homologous mechanism of 7 frequently-used herbs for heat-clearing and detoxification in traditional Chinese medicine (HDTCM) for treating Alzheimer's disease (AD), one of the most common types of dementia, based on network pharmacology. Herbs that satisfied the criteria of containing chlorogenic acid, relating to AD and aligning with HDTCM, were simultaneously collected to determine whether they have anti-AD effect based on a survey of the literature. Herb-ingredient-target-disease networks were constructed by collecting information from the TCMSP and GeneCards public databases. The common targets of the herbs and AD were identified for conducting a Gene Ontology (GO) analyses and a Reactome pathway enrichment analysis. The results showed that PTGS1, IL-6, CASP3, and VEGFA were the predicted key gene targets. The IL-4 and IL-13 signaling pathway, the ESR-mediated signaling pathway, and the extranuclear estrogen signaling pathway were the significant pathways associated with the 7 herbs. This study revealed that the analogous anti-AD mechanism of the 7 herbs of HDTCM may be associated with anti-inflammation, which is a common effect of the chlorogenic acid and quercetin components.
Collapse
|
34
|
Doulah A, Mahmoodi G, Pourmahdi Borujeni M. Evaluation of the pre-treatment effect of Centella asiatica medicinal plants on long-term potentiation (LTP) in rat model of Alzheimer's disease. Neurosci Lett 2020; 729:135026. [PMID: 32387717 DOI: 10.1016/j.neulet.2020.135026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2019] [Revised: 04/27/2020] [Accepted: 04/29/2020] [Indexed: 01/10/2023]
Abstract
The present study was aimed to investigate the pre-treatment effect of Centella asiatica (CeA) extract on long-term potentiation (LTP) in a rat model of Alzheimer's disease (AD). A total of 32 male Wistar rats weighing 380 ± 30 g were randomly divided into four groups (n = 8). Group 1 (C: Control): the control group. Group 2 (L: Lesion): The nucleus basalis of Meynert (NBM) of rats' brain was bilaterally destroyed by injection of Ibotenic acid. Group 3 (CeA): Animals in this group received the CeA leaf extract for only a period of six weeks. Group 4 (CeA + L): The NBM of rats was destroyed by Ibotenic acid after six weeks of a diet containing the CeA leaf extract. In all groups, LTP was recorded using the electrophysiological technique and fEPSP after high frequency stimulation (HFS). The results showed that the slope and amplitude of PS as well as the sub-curve level significantly increased in the CeA + L group compared with the L and CeA groups. The CeA extract improved and strengthened the slope, amplitude and sub-curve surface of cumulative waves in animals with NBM lesion. The results showed that administration CeA extract for six weeks before induction of NBM lesion and induction of Alzheimer could enhance memory. In other words, the CeA extract had a preventive or protective role. The present study showed that CeA had a protective role for neurons among rats with NBM lesion.
Collapse
Affiliation(s)
| | - Gelavij Mahmoodi
- Experimental Science Department, Kermanshah Branch, Islamic Azad University, Kermanshah, Iran
| | - Mahdi Pourmahdi Borujeni
- Department of Food Hygiene, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| |
Collapse
|
35
|
Han B, Jiang W, Liu H, Wang J, Zheng K, Cui P, Feng Y, Dang C, Bu Y, Wang QM, Ju Z, Hao J. Upregulation of neuronal PGC-1α ameliorates cognitive impairment induced by chronic cerebral hypoperfusion. Am J Cancer Res 2020; 10:2832-2848. [PMID: 32194838 PMCID: PMC7052889 DOI: 10.7150/thno.37119] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
Rationale: Mitochondrial dysfunction and oxidative stress occur in vascular dementia (VaD), but the specific molecular mechanism regulating these events remains unclear. Peroxisome proliferator-activated receptor-γ co-activator-1α (PGC-1α) is a master regulator for mitochondrial function. This study aims to investigate whether PGC-1α is involved in the pathophysiology of VaD. Methods: We firstly generated PGC-1α f/f Eno2-Cre mice to induce neuron-specific overexpression of PGC-1α by crossbreeding PGC-1α f/f mice with Eno2-cre mice. Then, the mice were subjected to bilateral common carotid artery stenosis to induce chronic cerebral hypoperfusion. Neurological function and hippocampal PGC-1α expression was evaluated. Next, RNA-Seq analysis and Seahorse assay were performed on the hippocampal neurons. In addition, mitochondrial antioxidants, uncoupling proteins, ROS production and the activation of glial cells were also measured. Results: Our results showed that hippocampal PGC-1α expression is down-regulated in the mouse VaD model induced by chronic cerebral hypoperfusion. In contrast, neuronal PGC-1α overexpression significantly ameliorated cognitive deficits. RNA-Seq analysis indicated that PGC-1α improved energy metabolism of neurons under hypoxic condition, and Seahorse assay confirmed that PGC-1α increases the metabolic activity of neurons. Further study demonstrated that PGC-1α boosted the expressions of mitochondrial antioxidants and uncoupling proteins (UCPs), including SOD2, Prx3, GPx1, UCP2, UCP4 and UCP5, which in turn reduced reactive oxygen species (ROS) production. Moreover, the activation of microglia and astrocytes was also found to decrease in the hippocampus. All of these changes greatly contributed to protect hippocampal neurons against ischemic insults. Conclusions: PGC-1α could suppress the excessive ROS and neuroinflammation in the hippocampus, opening up a potential therapeutic target for cognitive impairment.
Collapse
|
36
|
Meng SX, Wang B, Li WT. Intermittent hypoxia improves cognition and reduces anxiety-related behavior in APP/PS1 mice. Brain Behav 2020; 10:e01513. [PMID: 31877583 PMCID: PMC7010588 DOI: 10.1002/brb3.1513] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2019] [Revised: 09/20/2019] [Accepted: 11/08/2019] [Indexed: 01/09/2023] Open
Abstract
INTRODUCTION Although hypoxia can exacerbate symptoms of various neurological disorders, accumulating evidence has indicated that intermittent hypoxia (IH) may exert protective effects against brain diseases. In the present study, we aimed to determine whether exposure to IH exerts beneficial effects in a transgenic murine model of Alzheimer's disease (AD). Because comorbid anxiety is prevalent among patients with AD, we explored the effects of IH on anxiety-like behaviors and associated factors in APP/PS1 mice. METHODS APP/PS1 mice were subjected to IH for two weeks. We assessed cognitive performance and anxiety-related behavior using standard behavioral assessments. Amyloid beta (Aβ) levels in the hippocampus were assessed using immunofluorescence and enzyme-linked immunosorbent assays (ELISA). We also assessed cell morphology and brain-derived neurotrophic factor (BDNF) expression in the hippocampus. RESULTS Exposure to IH significantly increased cognitive performance and decreased anxiety-related behaviors in APP/PS1 mice. Immunofluorescence and ELISA results revealed that IH pretreatment significantly lowered Aβ levels in the cortex and hippocampus. Morphological studies validated the neuroprotective effect of IH exposure on hippocampal neurogenesis. Molecular studies revealed IH-enhanced BDNF expression and inhibition of apoptosis-related protein expression in the hippocampus of APP/PS1 mice. CONCLUSIONS Our study demonstrates that IH improves cognition and reduces anxiety in a murine model of AD. Thus, further studies are required to determine whether IH can be used as a preventive/adjuvant therapy in patients with AD.
Collapse
Affiliation(s)
- Sheng-Xi Meng
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Bing Wang
- Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Wen-Tao Li
- Department of Vasculocardiology, Municipal Hospital of Traditional Chinese Medicine Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
37
|
Sbrini G, Brivio P, Fumagalli M, Giavarini F, Caruso D, Racagni G, Dell’Agli M, Sangiovanni E, Calabrese F. Centella asiatica L. Phytosome Improves Cognitive Performance by Promoting Bdnf Expression in Rat Prefrontal Cortex. Nutrients 2020; 12:nu12020355. [PMID: 32013132 PMCID: PMC7071263 DOI: 10.3390/nu12020355] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2019] [Revised: 01/23/2020] [Accepted: 01/27/2020] [Indexed: 01/15/2023] Open
Abstract
A wide range of people in the world use natural remedies as primary approaches against illnesses. Accordingly, understanding the mechanisms of action of phytochemicals has become of great interest. In this context, Centella asiatica L. is extensively used, not only as anti-inflammatory or antioxidant agent but also as brain tonic. On this basis, the purpose of this study was to evaluate whether the chronic administration of C. asiatica L. to adult male rats was able to improve the expression of Bdnf, one of the main mediators of brain plasticity. Moreover, we assessed whether the treatment could affect the cognitive performance in the novel object recognition (NOR) test. We confirmed the presence of the main compounds in the plasma. Furthermore, C. asiatica L. administration induced an increase of Bdnf in the prefrontal cortex, and the administration of the higher dose of the extract was able to improve cognitive performance. Finally, the increase in the preference index in the NOR test was paralleled by a further increase in Bdnf expression. Overall, we highlight the ability of C. asiatica L. to affect brain functions by increasing Bdnf expression and by enhancing the cognitive performance.
Collapse
|
38
|
Sotolongo K, Ghiso J, Rostagno A. Nrf2 activation through the PI3K/GSK-3 axis protects neuronal cells from Aβ-mediated oxidative and metabolic damage. ALZHEIMERS RESEARCH & THERAPY 2020; 12:13. [PMID: 31931869 PMCID: PMC6958642 DOI: 10.1186/s13195-019-0578-9] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/30/2019] [Indexed: 01/08/2023]
Abstract
Background Mounting evidence points to a crucial role of amyloid-β (Aβ) in the pathophysiology of Alzheimer’s disease (AD), a disorder in which brain glucose hypometabolism, downregulation of central elements of phosphorylation pathways, reduced ATP levels, and enhanced oxidative damage coexist, and sometimes precede, synaptic alterations and clinical manifestations. Since the brain has limited energy storage capacity, mitochondria play essential roles in maintaining the high levels of energy demand, but, as major consumers of oxygen, these organelles are also the most important generators of reactive oxygen species (ROS). Thus, it is not surprising that mitochondrial dysfunction is tightly linked to synaptic loss and AD pathophysiology. In spite of their relevance, the mechanistic links among ROS homeostasis, metabolic alterations, and cell bioenergetics, particularly in relation to Aβ, still remain elusive. Methods We have used classic biochemical and immunocytochemical approaches together with the evaluation of real-time changes in global energy metabolism in a Seahorse Metabolic Analyzer to provide insights into the detrimental role of oligAβ in SH-SY5Y and primary neurons testing their pharmacologic protection by small molecules. Results Our findings indicate that oligomeric Aβ induces a dramatic increase in ROS production and severely affects neuronal metabolism and bioenergetics. Assessment of global energy metabolism in real time demonstrated Aβ-mediated reduction in oxygen consumption affecting basal and maximal respiration and causing decreased ATP production. Pharmacologic targeting of Aβ-challenged neurons with a set of small molecules of known antioxidant and cytoprotective activity prevented the metabolic/bioenergetic changes induced by the peptide, fully restoring mitochondrial function while inducing an antioxidant response that counterbalanced the ROS production. Search for a mechanistic link among the protective small molecules tested identified the transcription factor Nrf2—compromised by age and downregulated in AD and transgenic models—as their main target and the PI3K/GSK-3 axis as the central pathway through which the compounds elicit their Aβ protective action. Conclusions Our study provides insights into the complex molecular mechanisms triggered by oligAβ which profoundly affect mitochondrial performance and argues for the inclusion of small molecules targeting the PI3K/GSK-3 axis and Nrf2-mediated pathways as part of the current or future combinatorial therapies.
Collapse
Affiliation(s)
- Krystal Sotolongo
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA
| | - Jorge Ghiso
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA. .,Department of Psychiatry, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA.
| | - Agueda Rostagno
- Department of Pathology, New York University School of Medicine, 550 First Avenue, New York, NY, 10016, USA.
| |
Collapse
|
39
|
Polyphenols from Food and Natural Products: Neuroprotection and Safety. Antioxidants (Basel) 2020; 9:antiox9010061. [PMID: 31936711 PMCID: PMC7022568 DOI: 10.3390/antiox9010061] [Citation(s) in RCA: 129] [Impact Index Per Article: 32.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 12/31/2019] [Accepted: 01/07/2020] [Indexed: 02/07/2023] Open
Abstract
Polyphenols are naturally occurring micronutrients that are present in many food sources. Besides being potent antioxidants, these molecules may also possess anti-inflammatory properties. Many studies have highlighted their potential role in the prevention and treatment of various pathological conditions connected to oxidative stress and inflammation (e.g., cancer, and cardiovascular and neurodegenerative disorders). Neurodegenerative diseases are globally one of the main causes of death and represent an enormous burden in terms of human suffering, social distress, and economic costs. Recent data expanded on the initial antioxidant-based mechanism of polyphenols’ action by showing that they are also able to modulate several cell-signaling pathways and mediators. The proposed benefits of polyphenols, either as protective/prophylactic substances or as therapeutic molecules, may be achieved by the consumption of a natural polyphenol-enriched diet, by their use as food supplements, or with formulations as pharmaceutical drugs/nutraceuticals. It has also been proved that the health effects of polyphenols depend on the consumed amount and their bioavailability. However, their overconsumption may raise safety concerns due to the accumulation of high levels of these molecules in the organism, particularly if we consider the loose regulatory legislation regarding the commercialization and use of food supplements. This review addresses the main beneficial effects of food polyphenols, and focuses on neuroprotection and the safety issues related to overconsumption.
Collapse
|
40
|
Brandes MS, Gray NE. NRF2 as a Therapeutic Target in Neurodegenerative Diseases. ASN Neuro 2020; 12:1759091419899782. [PMID: 31964153 PMCID: PMC6977098 DOI: 10.1177/1759091419899782] [Citation(s) in RCA: 159] [Impact Index Per Article: 39.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2019] [Revised: 11/26/2019] [Accepted: 12/03/2019] [Indexed: 12/13/2022] Open
Abstract
Increased reactive oxygen species production and oxidative stress have been implicated in the pathogenesis of numerous neurodegenerative conditions including among others Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, Friedrich’s ataxia, multiple sclerosis, and stroke. The endogenous antioxidant response pathway protects cells from oxidative stress by increasing the expression of cytoprotective enzymes and is regulated by the transcription factor nuclear factor erythroid 2-related factor 2 (NRF2). In addition to regulating the expression of antioxidant genes, NRF2 has also been shown to exert anti-inflammatory effects and modulate both mitochondrial function and biogenesis. This is because mitochondrial dysfunction and neuroinflammation are features of many neurodegenerative diseases as well NRF2 has emerged as a promising therapeutic target. Here, we review evidence for a beneficial role of NRF2 in neurodegenerative conditions and the potential of specific NRF2 activators as therapeutic agents.
Collapse
Affiliation(s)
- Mikah S. Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
41
|
Wang L, Pu Z, Li M, Wang K, Deng L, Chen W. Antioxidative and antiapoptosis: Neuroprotective effects of dauricine in Alzheimer's disease models. Life Sci 2019; 243:117237. [PMID: 31887302 DOI: 10.1016/j.lfs.2019.117237] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 12/16/2019] [Accepted: 12/24/2019] [Indexed: 12/14/2022]
Abstract
AIMS Dauricine has been found that has significant neuroprotective effect on Alzheimer's disease (AD), but the mechanism is unclear, so we further investigated the possible mechanism of dauricine on AD. MAIN METHODS Cell counting kit-8 (CCK8) was applied to measure the cytotoxicity of dauricine on SH-SY5Y cells that overexpress the Swedish mutant form of human β-amyloid precursor protein (APPsw) and control cells (Neo). We used the Cu2+ to induce oxidative damage on APPsw cells, then tested the effect of dauricine on the damage and relative factors including reactive oxygen species (ROS), mitochondrial membrane potential (MMP) and superoxide dismutase (SOD) activity. The secretion level of amyloid beta 1-42(Aβ1-42), protein expression of apoptosis-related factors and the components of nuclear factor erythroid 2-related factor 2 (Nrf2) pathway were determined by western blotting. Aβ1-42-transgenic Caenorhabditis elegans GMC101, a model of AD, was applied to evaluate the neuroprotective effect of dauricine through the behavioral experiment and relative anti-oxidative tests. KEY FINDINGS In vitro, dauricine decreased the secretion level of Aβ1-42, significantly reduced the level of Cu2+-induced ROS, and restored MMP and SOD activity in APPsw cells. Meanwhile, dauricine could suppress the activation of caspase-3 and to upregulate the expression of Bcl-2. Dauricine also regulated the proteins levels of Nrf2, and Kelch-like ECH-associated protein 1 (Keap1) that is necessary for the activation of Nrf2 in APPsw cell. As oxidative stress induced by Aβ or paraquat (PQ), dauricine showed protective effects in the survival experiment of GMC101 worms. SIGNIFICANCE Those data revealed that dauricine has the pharmacological activity of anti-oxidative and anti-apoptosis, and shows the potential therapeutic value for AD.
Collapse
Affiliation(s)
- Lingfeng Wang
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Zhijun Pu
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China; Department of Neurology, Drum Tower Hospital, Medical School and The State Key Laboratory of Pharmaceutical Biotechnology, Institute of Brain Science, Nanjing University, Nanjing, China
| | - Mingxin Li
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Kaixuan Wang
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Lijuan Deng
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China
| | - Wei Chen
- Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, China.
| |
Collapse
|
42
|
Zweig JA, Caruso M, Brandes MS, Gray NE. Loss of NRF2 leads to impaired mitochondrial function, decreased synaptic density and exacerbated age-related cognitive deficits. Exp Gerontol 2019; 131:110767. [PMID: 31843395 DOI: 10.1016/j.exger.2019.110767] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Revised: 10/15/2019] [Accepted: 10/29/2019] [Indexed: 12/14/2022]
Abstract
Activation of the antioxidant regulatory transcription factor NRF2 (Nuclear factor erythroid-derived 2) regulates cellular bioenergetics and improves neuronal health in aging. Yet how NRF2 participates in maintaining synaptic, mitochondrial and cognitive function has not been fully elucidated. This study investigates how loss of NRF2 affects neuronal metabolism, synaptic density and cognitive performance in aged mice. Dendritic arborization as well as synaptic and mitochondrial gene expression was evaluated in hippocampal neurons isolated from mice lacking NRF2 (NRF2KO) and from wild-type (WT) C57BL6 mice. Mitochondrial function of these neurons was evaluated using the Seahorse XF platform. Additionally learning, memory and executive function were assessed in 20 month old NRF2KO and age-matched WT mice using conditioned fear response (CFR) and odor discrimination reversal learning (ODRL) tests. Hippocampal bioenergetics was profiled using mitochondria isolated from these animals and tissue was harvested for assessment of mitochondrial and synaptic genes. NRF2KO neurons had reduced dendritic complexity and diminished synaptic gene expression. This was accompanied by impaired mitochondrial function and decreased mitochondrial gene expression. Similar mitochondrial deficits were observed in the brains of aged NRF2KO mice. These animals also had significantly impaired cognitive performance and reduced synaptic gene expression as well. These data point to a role for NRF2 in maintaining mitochondrial and cognitive function during aging and suggest that the transcription factor may be a viable target for cognitive enhancing interventions. Because mitochondrial dysfunction and cognitive impairment also occur together in many neurodegenerative conditions there may be broad therapeutic potential of NRF2 activating agents.
Collapse
Affiliation(s)
- Jonathan A Zweig
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Mikah S Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA.
| |
Collapse
|
43
|
Matthews DG, Caruso M, Murchison CF, Zhu JY, Wright KM, Harris CJ, Gray NE, Quinn JF, Soumyanath A. Centella Asiatica Improves Memory and Promotes Antioxidative Signaling in 5XFAD Mice. Antioxidants (Basel) 2019; 8:antiox8120630. [PMID: 31817977 PMCID: PMC6943631 DOI: 10.3390/antiox8120630] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/03/2019] [Accepted: 12/06/2019] [Indexed: 12/11/2022] Open
Abstract
Centella asiatica (CA) herb is a traditional medicine, long reputed to provide cognitive benefits. We have reported that CA water extract (CAW) treatment improves cognitive function of aged Alzheimer’s disease (AD) model Tg2576 and wild-type (WT) mice, and induces an NRF2-regulated antioxidant response in aged WT mice. Here, CAW was administered to AD model 5XFAD female and male mice and WT littermates (age: 7.6 +/− 0.6 months), and object recall and contextual fear memory were tested after three weeks treatment. CAW’s impact on amyloid-β plaque burden, and markers of neuronal oxidative stress and synaptic density, was assessed after five weeks treatment. CAW antioxidant activity was evaluated via nuclear transcription factor (erythroid-derived 2)-like 2 (NRF2) and NRF2-regulated antioxidant response element gene expression. Memory improvement in both genders and genotypes was associated with dose-dependent CAW treatment without affecting plaque burden, and marginally increased synaptic density markers in the hippocampus and prefrontal cortex. CAW treatment increased Nrf2 in hippocampus and other NRF2 targets (heme oxygenase-1, NAD(P)H quinone dehydrogenase 1, glutamate-cysteine ligase catalytic subunit). Reduced plaque-associated SOD1, an indicator of oxidative stress, was observed in the hippocampi and cortices of CAW-treated 5XFAD mice. We postulate that CAW treatment leads to reduced oxidative stress, contributing to improved neuronal health and cognition.
Collapse
Affiliation(s)
- Donald G Matthews
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
| | - Charles F Murchison
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
- Department of Biostatistics, University of Alabama at Birmingham, Birmingham, AL 35294, USA
| | - Jennifer Y Zhu
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
| | - Kirsten M Wright
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
| | - Christopher J Harris
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR 97239, USA
| | - Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (D.G.M.); (M.C.); (C.F.M.); (J.Y.Z.); (K.M.W.); (C.J.H.); (N.E.G.); (J.F.Q.)
- Correspondence: ; Tel.: +1-503-494-6878
| |
Collapse
|
44
|
Li J, Long X, Hu J, Bi J, Zhou T, Guo X, Han C, Huang J, Wang T, Xiong N, Lin Z. Multiple pathways for natural product treatment of Parkinson's disease: A mini review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 60:152954. [PMID: 31130327 DOI: 10.1016/j.phymed.2019.152954] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/07/2019] [Accepted: 05/08/2019] [Indexed: 06/09/2023]
Abstract
BACKGROUND It is established that natural medicines for Parkinson's disease (PD) provide an antioxidant activity in preventing dopaminergic neurons from degeneration. However, the underlying and related molecular details remain poorly understood. METHODS AND AIM We review published in vitro and rodent studies of natural products in PD models with the aim to identify common molecular pathways contributing to the treatment efficacy. Commonly regulated genes were identified through the systemic literature search and further analyzed from a network perspective. FINDINGS Approximately thirty different types of natural products have been investigated for their ability to regulate protein density and gene activity in various experimental systems. Most were found to attenuate neurotoxin-induced regulations. Three common PD pathways are involved. The most studied pathway was neuronal development/anti-apoptosis consisting of Bax/Bcl-2, caspases 3/9, and MAPK signaling. Another well studied was anti-inflammation comprising iNOS, nNOS, Nrf2/ARE, cytokines, TNFα, COX2 and MAPK signaling. The third pathway referred to dopamine transmission modulation with upregulated VMAT2, DAT, NURR1 and GDNF levels. To date, HIPK2, a conserved serine/threonine kinase and transcriptional target of Nrf2 in an anti-apoptosis signaling pathway, is the first protein identified as the direct binding target of a natural product (ZMHC). IMPLICATIONS Natural products may utilize multiple and intercellular pathways at various steps to prevent DA neurons from degeneration. Molecular delineation of the mechanisms of actions is revealing new, perhaps combinational therapeutic approaches to stop the progression of DA degeneration.
Collapse
Affiliation(s)
- Jingwen Li
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Xi Long
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Jichuan Hu
- Department of Neurology, People's Hospital of Dongxihu District, Wuhan, Hubei 430040, China
| | - Juan Bi
- Department of Neurology, People's Hospital of Dongxihu District, Wuhan, Hubei 430040, China
| | - Ting Zhou
- Department of Neurology, People's Hospital of Dongxihu District, Wuhan, Hubei 430040, China
| | - Xingfang Guo
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Chao Han
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Department of Neurology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230001, China
| | - Jinsha Huang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Tao Wang
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China
| | - Nian Xiong
- Department of Neurology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, China; Department of Neurology, People's Hospital of Dongxihu District, Wuhan, Hubei 430040, China.
| | - Zhicheng Lin
- Laboratory of Psychiatric Neurogenomics, McLean Hospital, Harvard Medical School, Belmont, MA 02478, United States.
| |
Collapse
|
45
|
Chiroma SM, Baharuldin MTH, Mat Taib CN, Amom Z, Jagadeesan S, Ilham Adenan M, Mahdi O, Moklas MAM. Centella asiatica Protects d-Galactose/AlCl 3 Mediated Alzheimer's Disease-Like Rats via PP2A/GSK-3β Signaling Pathway in Their Hippocampus. Int J Mol Sci 2019; 20:E1871. [PMID: 31014012 PMCID: PMC6514756 DOI: 10.3390/ijms20081871] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Revised: 03/02/2019] [Accepted: 03/05/2019] [Indexed: 02/07/2023] Open
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder more prevalent among the elderly population. AD is characterised clinically by a progressive decline in cognitive functions and pathologically by the presence of neurofibrillary tangles (NFTs), deposition of beta-amyloid (Aβ) plaque and synaptic dysfunction in the brain. Centella asiatica (CA) is a valuable herb being used widely in African, Ayurvedic, and Chinese traditional medicine to reverse cognitive impairment and to enhance cognitive functions. This study aimed to evaluate the effectiveness of CA in preventing d-galactose/aluminium chloride (d-gal/AlCl3) induced AD-like pathologies and the underlying mechanisms of action were further investigated for the first time. Results showed that co-administration of CA to d-gal/AlCl3 induced AD-like rat models significantly increased the levels of protein phosphatase 2 (PP2A) and decreased the levels of glycogen synthase kinase-3 beta (GSK-3β). It was further observed that, CA increased the expression of mRNA of Bcl-2, while there was minimal effect on the expression of caspase 3 mRNA. The results also showed that, CA prevented morphological aberrations in the connus ammonis 3 (CA 3) sub-region of the rat's hippocampus. The results clearly demonstrated for the first time that CA could alleviate d-gal/AlCl3 induced AD-like pathologies in rats via inhibition of hyperphosphorylated tau (P-tau) bio-synthetic proteins, anti-apoptosis and maintenance of cytoarchitecture.
Collapse
Affiliation(s)
- Samaila Musa Chiroma
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Department of Human Anatomy, Faculty of Basic Medical Sciences, University of Maiduguri, Maiduguri 600230, Borno State, Nigeria.
| | | | - Che Norma Mat Taib
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| | - Zulkhairi Amom
- Faculty of Health Sciences, Universiti Teknologi Mara (UiTM) Kampus Puncak Alam, Bandar Puncak Alam 42300, Selangor, Malaysia.
| | - Saravanan Jagadeesan
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Department of Human Anatomy, Universiti Tunku Abdul Rahman (UTAR), Bandar Sungai Long Cheras 43000, Selangor, Malaysia.
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi Mara (UiTM) Kampus Puncak Alam, Bandar Puncak Alam 42300, Selangor, Malaysia.
| | - Onesimus Mahdi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
- Department of Human Anatomy, College of Medical Sciences, Gombe State University, Gombe 760211, Gombe State, Nigeria.
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang 43400, Selangor, Malaysia.
| |
Collapse
|
46
|
Chiroma SM, Baharuldin MTH, Mat Taib CN, Amom Z, Jagadeesan S, Ilham Adenan M, Mahdi O, Moklas MAM. Protective Effects of Centella asiatica on Cognitive Deficits Induced by D-gal/AlCl₃ via Inhibition of Oxidative Stress and Attenuation of Acetylcholinesterase Level. TOXICS 2019; 7:toxics7020019. [PMID: 30935005 PMCID: PMC6630231 DOI: 10.3390/toxics7020019] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/19/2019] [Revised: 03/06/2019] [Accepted: 03/08/2019] [Indexed: 12/18/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder with cholinergic dysfunctions and impaired redox homeostasis. The plant Centella asiatica (CA) is renowned for its nutritional benefits and herbal formulas for promoting health, enhancing cognition, and its neuroprotective effects. The present study aims to investigate the protective role of CA on D-gal/AlCl3-induced cognitive deficits in rats. The rats were divided into six groups and administered with donepezil 1 mg/kg/day, CA (200, 400, and 800 mg/kg/day) and D-gal 60 mg/kg/day + AlCl3 200 mg/kg/day for 10 weeks. The ethology of the rats was evaluated by the Morris water maze test. The levels of acetylcholinesterase (AChE), phosphorylated tau (P-tau), malondialdehyde (MDA) and activities of superoxide dismutase (SOD), in the hippocampus and cerebral cortex were estimated by enzyme-linked immunosorbent assay (ELISA). Additionally, the ultrastructure of the prefrontal cortex of the rats’ was observed using transmission electron microscopy (TEM). Rats administered with D-gal/AlCl3 exhibited cognitive deficits, decreased activities of SOD, and marked increase in AChE and MDA levels. Further, prominent alterations in the ultrastructure of the prefrontal cortex were observed. Conversely, co-administration of CA with D-gal/AlCl3 improved cognitive impairment, decreased AChE levels, attenuated the oxidative stress in hippocampus and cerebral cortex, and prevented ultrastructural alteration of neurons in the prefrontal cortex. Irrespective of the dose of CA administered, the protective effects were comparable to donepezil. In conclusion, this study suggests that CA attenuated the cognitive deficits in rats by restoring cholinergic function, attenuating oxidative stress, and preventing the morphological aberrations.
Collapse
Affiliation(s)
- Samaila Musa Chiroma
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
- Department of Human Anatomy, Faculty of Basic Medical Sciences, University of Maiduguri, 600230 Maiduguri, Borno State, Nigeria.
| | | | - Che Norma Mat Taib
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| | - Zulkhairi Amom
- Faculty of Health Sciences, Universiti Teknologi Mara (UiTM) Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| | - Saravanan Jagadeesan
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
- Department of Human Anatomy, Universiti Tunku Abdul Rahman (UTAR), Bandar Sungai Long, 43000 Kajang, Selangor, Malaysia.
| | - Mohd Ilham Adenan
- Atta-ur-Rahman Institute for Natural Product Discovery, Universiti Teknologi Mara (UiTM) Kampus Puncak Alam, 42300 Bandar Puncak Alam, Selangor, Malaysia.
| | - Onesimus Mahdi
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
- Department of Human Anatomy, College of Medical Sciences, Gombe State University, 760211 Gombe, Gombe State, Nigeria.
| | - Mohamad Aris Mohd Moklas
- Department of Human Anatomy, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia.
| |
Collapse
|