1
|
Chamberlin SR, Zweig JA, Neff CJ, Marney L, Choi J, Yang L, Maier CS, Soumyanath A, McWeeney S, Gray NE. Multi-Omics Analysis in Mouse Primary Cortical Neurons Reveals Complex Positive and Negative Biological Interactions Between Constituent Compounds of Centella asiatica. Pharmaceuticals (Basel) 2024; 18:19. [PMID: 39861082 PMCID: PMC11768890 DOI: 10.3390/ph18010019] [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: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/27/2025] Open
Abstract
Background: A water extract of the Ayurvedic plant Centella asiatica (L.) Urban, family Apiaceae (CAW), improves cognitive function in mouse models of aging and Alzheimer's disease and affects dendritic arborization, mitochondrial activity, and oxidative stress in mouse primary neurons. Triterpenes (TT) and caffeoylquinic acids (CQA) are constituents associated with these bioactivities of CAW, although little is known about how interactions between these compounds contribute to the plant's therapeutic benefit. Methods: Mouse primary cortical neurons were treated with CAW or equivalent concentrations of four TT combined, eight CQA combined, or these twelve compounds combined (TTCQA). Treatment effects on the cell transcriptome (18,491 genes) and metabolome (192 metabolites) relative to vehicle control were evaluated using RNAseq and metabolomic analyses, respectively. Results: Extensive differentially expressed genes (DEGs) were seen with all treatments, as well as evidence of interactions between compounds. Notably, many DEGs seen with TT treatment were not observed in the TTCQA condition, possibly suggesting CQA reduced the effects of TT. Moreover, additional gene activity seen with CAW as compared to TTCQA indicates the presence of additional compounds in CAW that further modulate TTCQA interactions. Weighted Gene Correlation Network Analysis (WGCNA) identified 4 gene co-expression modules altered by treatments that were associated with extracellular matrix organization, fatty acid metabolism, cellular response to stress and stimuli, and immune function. Compound interaction patterns were seen at the eigengene level in these modules. Interestingly, in metabolomics analysis, the TTCQA treatment saw the highest number of changes in individual metabolites (20), followed by CQA (15), then TT (8), and finally CAW (3). WGCNA analysis found two metabolomics modules with significant eigenmetabolite differences for TT and CQA and possible compound interactions at this level. Conclusions: Four gene expression modules and two metabolite modules were altered by the four treatment types applied. This methodology demonstrated the existence of both negative and positive interactions between TT, CQA, and additional compounds found in CAW on the transcriptome and metabolome of mouse primary cortical neurons.
Collapse
Affiliation(s)
- Steven R. Chamberlin
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA;
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA (A.S.); (N.E.G.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
| | - Jonathan A. Zweig
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA (A.S.); (N.E.G.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
| | - Cody J. Neff
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA (A.S.); (N.E.G.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
| | - Luke Marney
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Jaewoo Choi
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Liping Yang
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Claudia S. Maier
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA (A.S.); (N.E.G.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
| | - Shannon McWeeney
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR 97239, USA;
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97239, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA (A.S.); (N.E.G.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (L.M.); (C.S.M.)
| |
Collapse
|
2
|
Moreira P, Macedo J, Matos P, Bicker J, Fortuna A, Figueirinha A, Salgueiro L, Batista MT, Silva A, Silva S, Resende R, Branco PC, Cruz MT, Pereira CF. Effect of bioactive extracts from Eucalyptus globulus leaves in experimental models of Alzheimer's disease. Biomed Pharmacother 2024; 181:117652. [PMID: 39486370 DOI: 10.1016/j.biopha.2024.117652] [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: 08/01/2024] [Revised: 10/19/2024] [Accepted: 10/28/2024] [Indexed: 11/04/2024] Open
Abstract
Current therapies for Alzheimer's disease (AD) do not delay its progression, therefore, novel disease-modifying strategies are urgently needed. Recently, an increasing number of compounds from natural origin with protective properties against AD have been identified. Mixtures or extracts obtained from natural products containing several bioactive compounds have multifunctional properties and have drawn the attention because multiple AD pathways can be simultaneously modulated. This study evaluated the in vitro and in vivo effect of the essential oil (EO) obtained from the hydrodistillation of Eucalyptus globulus leaves, and an extract obtained from the hydrodistillation residual water (HRW). It was observed that EO and HRW have anti-inflammatory effect in brain immune cells modeling AD, namely lipopolysaccharide (LPS)- and amyloid-beta (Aβ)-stimulated microglia. In cell models that mimic AD-related neuronal dysfunction, HRW attenuated Aβ secretion and Aβ-induced mitochondrial dysfunction. Since the HRW's major components did not cross the blood-brain barrier, both EO and HRW were administered to the APP/PS1 transgenic AD mouse model by an intranasal route, which reduced cortical and hippocampal Aβ levels, and to rescue memory deficits and anxiety-like behaviors. Finally, HRW and EO were found to regulate cholesterol levels in aged mice after intranasal administration, suggesting that these extracts can reduce hypercholesterolemia and avoid risk for AD development. Overall, findings support a protective role of E. globulus extracts against AD‑like pathology and cognitive impairment highlighting the underlying mechanisms. These extracts obtained from underused forest biomass could be useful to develop nutraceutical supplements helpful to avoid AD risk and to prevent its progression.
Collapse
Affiliation(s)
- Patrícia Moreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal.
| | - Jéssica Macedo
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Patrícia Matos
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Joana Bicker
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Fortuna
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Artur Figueirinha
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; LAQV, REQUIMTE, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Lígia Salgueiro
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Maria Teresa Batista
- CIEPQPF, Research Center for Chemical Processes Engineering and Forest Products, Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Ana Silva
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Sónia Silva
- Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal; iCBR-Coimbra Institute for Clinical and Biomedical Research, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Rosa Resende
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal
| | - Pedro Costa Branco
- RAIZ-Forest and Paper Research Institute, Eixo, Aveiro 3800-783, Portugal
| | - Maria Teresa Cruz
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Pharmacy, University of Coimbra, Coimbra 3000-548, Portugal
| | - Cláudia Fragão Pereira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra 3004-504, Portugal; Faculty of Medicine, University of Coimbra, Coimbra 3000-548, Portugal.
| |
Collapse
|
3
|
Chamberlin S, Zweig JA, Neff CJ, Marney L, Choi J, Yang L, Maier CS, Soumyanath A, McWeeney S, Gray NE. Multi-omics analysis in mouse primary cortical neurons reveals complex positive and negative biological interactions between constituent compounds in Centella asiatica. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.04.621595. [PMID: 39574684 PMCID: PMC11580974 DOI: 10.1101/2024.11.04.621595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/10/2024]
Abstract
Background A water extract of the Ayurvedic plant Centella asiatica (CAW) improves cognitive function in mouse models of aging and Alzheimer's disease, and affects dendritic arborization, mitochondrial activity and oxidative stress in mouse primary neurons. Triterpenes (TT) and caffeoylquinic acids (CQA) are constituents associated with these bioactivities of CAW although little is known about how interactions between these compounds contribute to the plant's therapeutic benefit. Methods Mouse primary cortical neurons were treated with CAW, or equivalent concentrations of four TT combined, eight CQA combined, or these twelve compounds combined (TTCQA). Treatment effects on the cell transcriptome (18,491 genes) and metabolome (192 metabolites) relative to vehicle control were evaluated using RNAseq and metabolomic analyses respectively. Results Extensive differentially expressed genes (DEGs) were seen with all treatments, as well as evidence of interactions between compounds. Notably many DEGs seen with TT treatment were not observed in the TTCQA condition, possibly suggesting CQA reduced the effects of TT. Moreover, additional gene activity seen with CAW as compared to TTCQA indicate the presence of additional compounds in CAW that further modulate TTCQA interactions. Weighted Gene Correlation Network Analysis (WGCNA) identified 4 gene co-expression modules altered by treatments that were associated with extracellular matrix organization, fatty acid metabolism, cellular response to stress and stimuli, and immune function. Compound interaction patterns were seen at the eigengene level in these modules. Interestingly, in metabolomics analysis, the TTCQA treatment saw the highest number of changes in individual metabolites (20), followed by CQA (15), then TT (8) and finally CAW (3). WGCNA analysis found two metabolomics modules with significant eigenmetabolite differences for TT and CQA, and possible compound interactions at this level. Conclusions Four gene expression modules and two metabolite modules were altered by the four types of treatments applied. This methodology demonstrated the existence of both negative and positive interactions between TT, CQA and additional compounds found in CAW on the transcriptome and metabolome of mouse primary cortical neurons.
Collapse
Affiliation(s)
- Steve Chamberlin
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR
- Department of Neurology, Oregon Health & Science University, Portland, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
| | - Jonathan A Zweig
- Department of Neurology, Oregon Health & Science University, Portland, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
| | - Cody J Neff
- Department of Neurology, Oregon Health & Science University, Portland, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
| | - Luke Marney
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
- Department of Chemistry, Oregon State University, Corvallis, OR
| | - Jaewoo Choi
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
- Department of Chemistry, Oregon State University, Corvallis, OR
| | - Liping Yang
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
- Department of Chemistry, Oregon State University, Corvallis, OR
| | - Claudia S Maier
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
- Department of Chemistry, Oregon State University, Corvallis, OR
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
| | - Shannon McWeeney
- Department of Medical Informatics and Clinical Epidemiology, Oregon Health & Science University, Portland, OR
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR
| | - Nora E Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University (OHSU) Portland, OR
| |
Collapse
|
4
|
Eze FN, Bunyapongpan A, Prapunpoj P. Neuroprotective effect against amyloidogenic transthyretin aggregates - Induced cytotoxicity on human neuroblastoma cell by phenolic-rich Centella asiatica extract. Heliyon 2024; 10:e39159. [PMID: 39640739 PMCID: PMC11620063 DOI: 10.1016/j.heliyon.2024.e39159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 10/08/2024] [Accepted: 10/08/2024] [Indexed: 12/07/2024] Open
Abstract
Transthyretin (ATTR) amyloidosis is a progressive and life-threatening neurodegenerative disease caused by aggregation of the plasma transport protein, transthyretin, for which treatment is rare and cure unavailable. Centella asiatica is a small edible herb with a long history of neurological application in ethnomedicine. This work investigated whether hydrophilic extract of C. asiatica (CAB) could suppress the toxic effects of transthyretin amyloid aggregate (TTRa) in cell model derived from the same in vivo target. TTRa was prepared via thermal-induced aggregation. Chemical cross-linking and Tricine-SDS-PAGE, Thioflavin-T fluorescence, and TEM analyses confirmed that TTRa matched the profile of TTRL55P nonfibrillar amyloid aggregates. PrestoBlue cell viability assay revealed that exposure of IMR-32 human neuroblastoma cells to TTRa (2-8 μM) resulted in significant cytotoxicity. Conversely, exposure of IMR-32 cells to CAB did not adversely affect their viability. In addition, when IMR-32 cells were co-treated with TTRa and varied concentrations of CAB, the toxic effect of TTRa was significantly (p < 0.01) inhibited dose-dependently. The extract was found to possess potent radical scavenging effects, and quantitative RP-HPLC analysis showed that asiaticoside and phenolics were its main components. The cytoprotective effect against TTRa, antioxidant property, and good safety profile collectively suggest that CAB could be applied in the development of nutraceuticals or therapeutics against transthyretin amyloidosis.
Collapse
Affiliation(s)
- Fredrick Nwude Eze
- Drug Delivery System Excellence Center, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
- Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Apinna Bunyapongpan
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Porntip Prapunpoj
- Department of Biochemistry, Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| |
Collapse
|
5
|
Firdaus Z, Gutti G, Ganeshpurkar A, Kumar A, Krishnamurthy S, Singh SK, Singh TD. Centella asiatica improves memory and executive function in middle-aged rats by controlling oxidative stress and cholinergic transmission. JOURNAL OF ETHNOPHARMACOLOGY 2024; 325:117888. [PMID: 38336185 DOI: 10.1016/j.jep.2024.117888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 01/31/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Centella asiatica (L.) Urban, is a medicinal herb with rich history of traditional use in Indian subcontinent. This herb has been valued for its diverse range of medicinal properties including memory booster, and also as a folk treatment for skin diseases, wound healing and mild diuretic. AIM OF STUDY Aging is a gradual and continuous process of natural decay in the biological systems, including the brain. This work aims to evaluate the effectiveness of ethanolic extract of Centella asiatica (CAE) on age-associated cognitive impairments in rats, as well as the underlying mechanism. MATERIAL AND METHODS Rats were allocated into five distinct groups of 5 animals each: Young rats (3 months old rats), middle-aged (m-aged) rats (13-14 months old), and the remaining three groups were comprised of m-aged rats treated with different concentrations of CAE, viz., 150, 300, and 450 mg/kg b. w., orally for 42 days. Y-maze, open field, novel object recognition, and elevated plus maze tests were used to assess animal behavior. The malondialdehyde (MDA), superoxide dismutase (SOD), and acetylcholinesterase (AChE) assays; and H&E staining were done in the rat brain to assess the biochemical and structural changes. CAE was also subjected to HPLC analysis, in vitro antioxidant and anti-cholinergic activity. The active compounds of CAE were docked with AChE and BuChE in molecular docking study. RESULTS The results showed that CAE treatment improves behavioral performance; attenuates the age-associated increase in MDA content, SOD, and AChE activity; and reduces neuronal loss. In vitro study showed that CAE has concentration-dependent antioxidant and anti-AChE activity. Furthermore, the presence of Asiatic acid and Madecassic acid in CAE and their good binding with cholinergic enzymes (in silico) also suggest the anticholinergic effect of CAE. CONCLUSION The findings of the current study show that the anticholinergic and antioxidant effects of CAE are attributable to the presence of Asiatic acid and Madecassic acid, which not only provide neuroprotection against age-associated cognitive decline but also reverse it.
Collapse
Affiliation(s)
- Zeba Firdaus
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Gopichand Gutti
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, 221005, India
| | - Ankit Ganeshpurkar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, 221005, India
| | - Ashok Kumar
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, 221005, India
| | - Sairam Krishnamurthy
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, 221005, India
| | - Sushil Kumar Singh
- Department of Pharmaceutical Engineering and Technology, Indian Institute of Technology, BHU, Varanasi, 221005, India
| | - Tryambak Deo Singh
- Department of Medicinal Chemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
6
|
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
|
7
|
Jusril NA, Mukhtar SM, Abu Bakar SI, Md Saad WM, Wen NK, Adenan MI. Nanoemulsion of Ethanolic Extract of Centella asiatica (NanoSECA)Ameliorates Learning and Memory Performance by EnhancingCholinergic Activities, Increasing Antioxidative Levels, and AttenuatingOxidative Stress Markers in Adult Rats. Pharm Nanotechnol 2024; 12:329-339. [PMID: 37653639 DOI: 10.2174/2211738511666230831122844] [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: 02/08/2023] [Revised: 06/27/2023] [Accepted: 07/24/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Centella asiatica (C. asiatica) has long been traditionally used as a memory enhancer. Nanoemulsion of ethanolic extract C. asiatica (NanoSECA) has been developed to improve brain functions. However, the effect of NanoSECA on enhancing memory and cognitive functions remains unexplored. OBJECTIVES This research aimed to investigate the potential of NanoSECA on cognitive tasks and memory enhancement pathways in a normal adult rat model. METHODS Thirty male Sprague Dawley rats (7-8 weeks old) were randomly subjected to five groups (n=six per group). Treatment groups were supplemented with NanoSECA and ethanolic extract of C. asiatica (SECA) for 28 days by oral gavages. Different brain sections were isolated, homogenized, and tested for acetylcholinesterase, antioxidants (glutathione and malondialdehyde), and anti-inflammatory agents (nitric oxide, tumour necrosis factor-α, and prostaglandin E2). RESULTS NanoSECA supplementation markedly enhanced the acetylcholine, glutathione levels and reduced a distinct diminution in plasma activities of acetylcholinesterase, malondialdehyde, nitric oxide, prostaglandin E, and tumor necrosis factor-α levels. CONCLUSION NanoSECA can be used as a memory enhancer through enhanced cholinergic activity, increased antioxidant level, and reduced oxidative stress.
Collapse
Affiliation(s)
- Nor Atiqah Jusril
- School of Agriculture Science & Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut Campus, 22200, Besut, Terengganu, Malaysia
| | - Shahida Muhammad Mukhtar
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Level 9, FF3, Puncak Alam Campus, Universiti Teknologi MARA, Puncak Alam, 42300, Selangor Darul Ehsan, Malaysia
| | - Syahrul Imran Abu Bakar
- School of Agriculture Science & Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut Campus, 22200, Besut, Terengganu, Malaysia
- Atta-ur-Rahman Institute for Natural Product Discovery (AuRIns), Level 9, FF3, Puncak Alam Campus, Universiti Teknologi MARA, Puncak Alam, 42300, Selangor Darul Ehsan, Malaysia
| | - Wan Mazlina Md Saad
- Centre of Medical Laboratory Technology, Faculty of Health Sciences, Universiti Teknologi MARA, Puncak Alam, 42300, Selangor Darul Ehsan, Malaysia
| | - Ng Kwok Wen
- Faculty of Pharmacy, Quest International University, Jalan Raja Permaisuri Bainun, 30250, Ipoh, Perak, Malaysia
| | - Mohd Ilham Adenan
- School of Agriculture Science & Biotechnology, Faculty of Bioresources and Food Industry, Universiti Sultan Zainal Abidin, Besut Campus, 22200, Besut, Terengganu, Malaysia
- Faculty of Applied Sciences, Universiti Teknologi MARA Pahang Branch, Bandar Tun Abdul Razak, 26400, Jengka, Pahang Darul Makmur, Malaysia
| |
Collapse
|
8
|
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
|
9
|
Ho E, Drake VJ, Michels AJ, Nkrumah-Elie YM, Brown LL, Scott JM, Newman JW, Shukitt-Hale B, Soumyanath A, Chilton FH, Lindemann SR, Shao A, Mitmesser SH. Perspective: Council for Responsible Nutrition Science in Session. Optimizing Health with Nutrition-Opportunities, Gaps, and the Future. Adv Nutr 2023; 14:948-958. [PMID: 37270030 PMCID: PMC10509435 DOI: 10.1016/j.advnut.2023.05.015] [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: 03/30/2023] [Revised: 05/20/2023] [Accepted: 05/30/2023] [Indexed: 06/05/2023] Open
Abstract
Achieving optimal health is an aspirational goal for the population, yet the definition of health remains unclear. The role of nutrition in health has evolved beyond correcting malnutrition and specific deficiencies and has begun to focus more on achieving and maintaining 'optimal' health through nutrition. As such, the Council for Responsible Nutrition held its October 2022 Science in Session conference to advance this concept. Here, we summarize and discuss the findings of their Optimizing Health through Nutrition - Opportunities and Challenges workshop, including several gaps that need to be addressed to advance progress in the field. Defining and evaluating various indices of optimal health will require overcoming these key gaps. For example, there is a strong need to develop better biomarkers of nutrient status, including more accurate markers of food intake, as well as biomarkers of optimal health that account for maintaining resilience-the ability to recover from or respond to stressors without loss to physical and cognitive performance. In addition, there is a need to identify factors that drive individualized responses to nutrition, including genotype, metabotypes, and the gut microbiome, and to realize the opportunity of precision nutrition for optimal health. This review outlines hallmarks of resilience, provides current examples of nutritional factors to optimize cognitive and performance resilience, and gives an overview of various genetic, metabolic, and microbiome determinants of individualized responses.
Collapse
Affiliation(s)
- Emily Ho
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon; Nutrition Program, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon.
| | - Victoria J Drake
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
| | | | | | - LaVerne L Brown
- National Institutes of Health, Office of Dietary Supplements, Bethesda, Maryland
| | - Jonathan M Scott
- Consortium for Health and Military Performance, Department of Military and Emergency Medicine, F. Edward Hébert School of Medicine, Uniformed Services University, Bethesda, Maryland
| | - John W Newman
- United States Department of Agriculture, Agricultural Research Service, Western Human Nutrition Research Center, Davis, California
| | - Barbara Shukitt-Hale
- United States Department of Agriculture, Agricultural Research Service, Human Nutrition Research Center on Aging at Tufts University, Boston, Massachusetts
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Floyd H Chilton
- Center for Precision Nutrition and Wellness, University of Arizona, Tucson, Arizona; School of Nutritional Sciences and Wellness, College of Agriculture and Life Sciences, University of Arizona, Tucson, Arizona
| | - Stephen R Lindemann
- Whistler Center for Carbohydrate Research, Department of Food Science, Purdue University, West Lafayette, Indiana
| | - Andrew Shao
- ChromaDex External Research Program, Los Angeles, California
| | | |
Collapse
|
10
|
Wright KM, Bollen M, David J, Mepham B, Alcázar Magaña A, McClure C, Maier CS, Quinn JF, Soumyanath A. Bioanalytical method validation and application to a phase 1, double-blind, randomized pharmacokinetic trial of a standardized Centella asiatica (L.) Urban water extract product in healthy older adults. Front Pharmacol 2023; 14:1228030. [PMID: 37680716 PMCID: PMC10481538 DOI: 10.3389/fphar.2023.1228030] [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: 05/24/2023] [Accepted: 08/03/2023] [Indexed: 09/09/2023] Open
Abstract
Introduction: Centella asiatica is an herbaceous plant reputed in Eastern medicine to improve memory. Preclinical studies have shown that C. asiatica aqueous extract (CAW) improves neuronal health, reduces oxidative stress, and positively impacts learning and cognition. This study aimed to develop and validate bioanalytical methods for detecting known bioactive compounds from C. asiatica in human biological matrices and apply them to a human pharmacokinetic trial in healthy older adults. Methods: High performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS) was used for detecting triterpenes and caffeoylquinic acids from C. asiatica, or their metabolites, in human plasma and urine. Validation parameters including linearity, precision, accuracy, recovery and thermal stability were evaluated. The method was applied to a Phase I, randomized, double-blind, crossover trial of two doses (2 or 4 g) of a standardized C. asiatica water extract product (CAP) in eight healthy older adults. Pharmacokinetic parameters were measured over a 12-h post administration period and acute safety was assessed. Results: The method satisfied US Food & Drug Administration criteria for linearity and recovery of the analytes of interest in human plasma and urine. The method also satisfied criteria for precision and accuracy at medium and high concentrations. Single administration of 2 and 4 g of CAP was well tolerated and safe in healthy older adults. The parent triterpene glycosides, asiaticoside and madecassoside, were not detected in plasma and in minimal amounts in urinary excretion analyses, while the aglycones, asiatic acid and madecassic acid, showed readily detectable pharmacokinetic profiles. Similarly, the di-caffeoylquinic acids and mono-caffeoylquinic acids were detected in low quantities, while their putative metabolites showed readily detectable pharmacokinetic profiles and urinary excretion. Discussion: This method was able to identify and calculate the concentration of triterpenes and caffeoylquinic acids from C. asiatica, or their metabolites, in human plasma and urine. The oral absorption of these key compounds from CAP, and its acute safety in healthy older adults, support the use of this C. asiatica product in future clinical trials.
Collapse
Affiliation(s)
- Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Melissa Bollen
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
| | - Jason David
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Bridgette Mepham
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Christine McClure
- Department of Neurology, Oregon Health & Science University, 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
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Veterans Affairs Portland Healthcare System Center, Department of Neurology, Portland, OR, United States
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
11
|
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: 2.7] [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
|
12
|
Srivastava V, Mathur D, Rout S, Mishra BK, Pannu V, Anand A, Anand A. Ayurvedic Herbal Therapies: A Review of Treatment and Management of Dementia. Curr Alzheimer Res 2022; 19:568-584. [PMID: 35929620 DOI: 10.2174/1567205019666220805100008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/18/2022] [Accepted: 04/26/2022] [Indexed: 01/27/2023]
Abstract
Dementia has been characterized by atypical neurological syndromes and several cognitive deficits, such as extended memory loss, strange behavior, unusual thinking, impaired judgment, impotence, and difficulty with daily living activities. Dementia is not a disease, but it is caused by several neurodegenerative diseases, such as Alzheimer's, Parkinson's, and Lewy's bodies. Several drugs and remedies are indicated for alleviating unusual cognitive decline, but no effective pharmacological treatment regimens are available without side effects. Herbal drugs or traditional medicines like Ayurveda have been known for facilitating and corroborating the balance between mind, brain, body, and environment. Ayurvedic therapy comprises 600 herbal formulas, 250 single plant remedies, and natural and holistic health-giving treatments that relieve dementia in patients and increase vitality. Ayurvedic Rasayana herbs [rejuvenating elements] strengthen the brain cells, enhance memory, and decrease stress. The current medicine scenario in the treatment of dementia has prompted the shift in exploring the efficacy of ayurvedic medicine, its safety, and its efficiency. This review presents the literature on several herbal treatments for improving dementia symptomatology and patients' quality of life.
Collapse
Affiliation(s)
- Vinod Srivastava
- College of Health and Behavioral Sciences, Fort Hays State University, Hays, Kansas 67601, USA
| | - Deepali Mathur
- Department of Neurology, Apollo Hospitals, Bhubaneswar, Odisha, India
| | - Soumyashree Rout
- Department of Neurology, Apollo Hospitals, Bhubaneswar, Odisha, India
| | | | - Viraaj Pannu
- Department of Internal Medicine, Jersey Shore University Medical Center, Neptune, New Jersey, USA
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Akshay Anand
- Neuroscience Research Lab, Department of Neurology, PGIMER, Chandigarh, India
| |
Collapse
|
13
|
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: 3.3] [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
|
14
|
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: 0.3] [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
|
15
|
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
|
16
|
Haroon HB, Mukherjee D, Anbu J, Teja BV. Thiolated Chitosan-Centella asiatica Nanocomposite: A Potential Brain Targeting Strategy Through Nasal Route. AAPS PharmSciTech 2021; 22:251. [PMID: 34668091 DOI: 10.1208/s12249-021-02131-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 08/27/2021] [Indexed: 01/02/2023] Open
Abstract
The major challenge associated with the treatment of neurological disorders is the inefficiency of drugs to enter the Central Nervous System (CNS). Polymer-drug conjugates are now being tailored to overcome this hindrance associated with conventional drugs. The study aimed at developing polymer hybrid nasal nanocomposite for enhanced delivery of Centella to the CNS. Thiolated chitosan was complexed with Centella to form a composite using EDAC hydrochloride. The composite was characterized by FTIR, XRD, NMR, and MS. Further, this composite was converted into a nanoformulation by the ionic-gelation method, characterized, and subjected to ex vivo permeation studies. Additionally, MTT assay was performed using Human Uumbilical cord Vein Endothelial Cells (HUVECs) mimicking Blood-Brain Barrier (BBB) to establish the safety of nanocomposite. The targeting efficacy was predicted by molecular docking studies against receptors associated with BBB. The FTIR, XRD, NMR, and MS studies confirmed the chemical conjugation of thiolated chitosan with Centella. Nanocomposite characterization through SEM, AFM, and DLS confirmed the size and stability of the developed nanocomposite having a zeta potential of - 14.5 mV and PDI of 0.260. The nanocomposite showed no signs of nasal ciliotoxicity and good permeation of 89.44 ± 1.75% (mean ± SD, n = 3) at 8 h across the nasal mucosa. MTT assay showed that the nanocomposite had lesser toxicity compared to the free drug (IC50 of Centella-269.1 μg/mL and IC50 of CTC nanocomposite-485.375 μg/mL). The affinity of polymer to the BBB receptors as proved by docking studies suggests the ability of polymer-based nanocomposite to concentrate in the brain post nasal administration.
Collapse
|
17
|
Magaña AA, Kamimura N, Soumyanath A, Stevens JF, Maier CS. Caffeoylquinic acids: chemistry, biosynthesis, occurrence, analytical challenges, and bioactivity. THE PLANT JOURNAL : FOR CELL AND MOLECULAR BIOLOGY 2021; 107:1299-1319. [PMID: 34171156 PMCID: PMC9084498 DOI: 10.1111/tpj.15390] [Citation(s) in RCA: 89] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/15/2021] [Accepted: 06/19/2021] [Indexed: 05/02/2023]
Abstract
Caffeoylquinic acids (CQAs) are specialized plant metabolites we encounter in our daily life. Humans consume CQAs in mg-to-gram quantities through dietary consumption of plant products. CQAs are considered beneficial for human health, mainly due to their anti-inflammatory and antioxidant properties. Recently, new biosynthetic pathways via a peroxidase-type p-coumaric acid 3-hydroxylase enzyme were discovered. More recently, a new GDSL lipase-like enzyme able to transform monoCQAs into diCQA was identified in Ipomoea batatas. CQAs were recently linked to memory improvement; they seem to be strong indirect antioxidants via Nrf2 activation. However, there is a prevalent confusion in the designation and nomenclature of different CQA isomers. Such inconsistencies are critical and complicate bioactivity assessment since different isomers differ in bioactivity and potency. A detailed explanation regarding the origin of such confusion is provided, and a recommendation to unify nomenclature is suggested. Furthermore, for studies on CQA bioactivity, plant-based laboratory animal diets contain CQAs, which makes it difficult to include proper control groups for comparison. Therefore, a synthetic diet free of CQAs is advised to avoid interferences since some CQAs may produce bioactivity even at nanomolar levels. Biotransformation of CQAs by gut microbiota, the discovery of new enzymatic biosynthetic and metabolic pathways, dietary assessment, and assessment of biological properties with potential for drug development are areas of active, ongoing research. This review is focused on the chemistry, biosynthesis, occurrence, analytical challenges, and bioactivity recently reported for mono-, di-, tri-, and tetraCQAs.
Collapse
Affiliation(s)
- Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
| | - Naofumi Kamimura
- Department of Bioengineering, Nagaoka University of Technology, Nagaoka, Niigata, Japan
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
- Department of Neurology, Oregon Health and Science University, Portland, OR, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, USA
| |
Collapse
|
18
|
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: 1.5] [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
|
19
|
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: 29] [Impact Index Per Article: 5.8] [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
|
20
|
Simultaneous analysis of five triterpenes in Centella asiatica by high performance liquid chromatography with cyclodextrins as the mobile phase additives. Sci Rep 2020; 10:18577. [PMID: 33122688 PMCID: PMC7596059 DOI: 10.1038/s41598-020-75554-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022] Open
Abstract
Triterpenes are considered the major active components in Centella asiatica (L.) Urb. (C. asiatica), such as asiatic acid, madecassic acid, asiaticoside, madecassoside and asiaticoside B. It is difficult to simultaneously determine five triterpenes because of madecassoside isomers (madecassoside and asiaticoside B), and the great polarity difference between triterpene acid and triterpene glycoside. In this study, a simple high performance liquid chromatography method with isocratic elution employing cyclodextrins (CDs) as the mobile phase additives was developed to determine five triterpenes in C. asiatica. Various factors affecting triterpenes retention in the C18 column, such as the nature of CDs, γ-CD concentration, acetonitrile percentage and temperature, were studied. Experimental results showed that γ-CD, as an effective mobile phase additive, could markedly reduce the retention of triterpenes (especially asiatic acid and madecassic acid), and improve the separation for madecassoside and asiaticoside B. The elution of five triterpenes could be achieved on an ODS C18 column within 30 min using the acetonitrile-0.2% phosphoric acid contained 4.0 mM γ-CD (20:80, v/v) mixture as the mobile phase. The retention modification of triterpenes may be attributed to the formation of the triterpenes-γ-CD inclusion complexes. The optimized method was successfully applied for simultaneous determination of five triterpenes in C. asiatica.
Collapse
|
21
|
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: 0.8] [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
|
22
|
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: 95] [Impact Index Per Article: 19.0] [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
|
23
|
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: 167] [Impact Index Per Article: 33.4] [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
|
24
|
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: 3.0] [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
|
25
|
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: 5.5] [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
|
26
|
Quinn JF. Lost in Translation? Finding Our Way To Effective Alzheimer's Disease Therapies. J Alzheimers Dis 2019; 64:S33-S39. [PMID: 29758942 DOI: 10.3233/jad-179930] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Efforts over the past two decades to develop effective disease-modifying treatments for Alzheimer's disease have been disappointing, while parallel efforts in another chronic neurologic disease, multiple sclerosis, have been remarkably productive. In an effort to advance development of therapeutics for Alzheimer's disease, these two fields are contrasted in terms of the utility of animal models, definition of study populations, and utility of biomarkers. Possible solutions are suggested, and the review concludes with description of some active peer-reviewed, publicly funded clinical studies which address some of the identified weaknesses in past clinical trials for age-related dementia.
Collapse
Affiliation(s)
- Joseph F Quinn
- Oregon Health and Science University, Portland VA Medical Center, Department of Neurology, Portland, OR, USA
| |
Collapse
|
27
|
Gray NE, Zweig JA, Caruso M, Zhu JY, Wright KM, Quinn JF, Soumyanath A. Centella asiatica attenuates hippocampal mitochondrial dysfunction and improves memory and executive function in β-amyloid overexpressing mice. Mol Cell Neurosci 2018; 93:1-9. [PMID: 30253196 DOI: 10.1016/j.mcn.2018.09.002] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/17/2018] [Accepted: 09/20/2018] [Indexed: 12/21/2022] Open
Abstract
Centella asiatica is a medicinal plant used to enhance memory. We have previously shown that a water extract of Centella asiatica (CAW) attenuates β-amyloid (Aβ)-induced spatial memory deficits in mice and improves neuronal health. Yet the effect of CAW on other cognitive domains remains unexplored as does its in vivo mechanism of improving Aβ-related cognitive impairment. This study investigates the effects of CAW on learning, memory and executive function as well as mitochondrial function and antioxidant response in the 5xFAD model of Aβ accumulation. Seven month old 5xFAD female mice were treated with CAW (2 mg/mL) in their drinking water for two weeks prior to behavioral testing. Learning, memory and executive function were assessed using the object location memory task (OLM), conditioned fear response (CFR) and odor discrimination reversal learning (ODRL) test. Mitochondrial function was profiled using the Seahorse XF platform in hippocampal mitochondria isolated from these animals and tissue was harvested for assessment of mitochondrial, antioxidant and synaptic proteins. CAW improved performance in all behavioral tests in the 5xFAD but had no effect on WT animals. Hippocampal mitochondrial function was improved and hippocampal and cortical expression of mitochondrial genes was increased in CAW-treated 5xFAD mice. Gene expression of the transcription factor NRF2, as well as its antioxidant target enzymes, was also increased with CAW treatment in both WT and 5xFAD mice. CAW treatment also decreased Aβ-plaque burden in the hippocampus of treated 5xFAD mice but had no effect on plaques in the cortex. These data show that CAW can improve many facets of Aβ-related cognitive impairment in 5xFAD mice. Oral treatment with CAW also attenuates hippocampal mitochondrial dysfunction in these animals. Because mitochondrial dysfunction and oxidative stress accompany cognitive impairment in many pathological conditions beyond Alzheimer's disease, this suggests potentially broad therapeutic utility of CAW.
Collapse
Affiliation(s)
- Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA.
| | - 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
| | - Jennifer Y Zhu
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Kirsten M Wright
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA
| |
Collapse
|
28
|
Binti Mohd Yusuf Yeo NA, Muthuraju S, Wong JH, Mohammed FR, Senik MH, Zhang J, Yusof SR, Jaafar H, Adenan ML, Mohamad H, Tengku Muhammad TS, Abdullah JM. Hippocampal amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid GluA1 (AMPA GluA1) receptor subunit involves in learning and memory improvement following treatment with Centella asiatica extract in adolescent rats. Brain Behav 2018; 8:e01093. [PMID: 30105867 PMCID: PMC6160644 DOI: 10.1002/brb3.1093] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Revised: 07/02/2018] [Accepted: 07/06/2018] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Centella asiatica is an herbal plant that contains phytochemicals that are widely believed to have positive effects on cognitive function. The adolescent stage is a critical development period for the maturation of brain processes that encompass changes in physical and psychological systems. However, the effect of C. asiatica has not been extensively studied in adolescents. The aim of this study was therefore to investigate the effects of a C. asiatica extract on the enhancement of learning and memory in adolescent rats. METHODS The locomotor activity, learning, and memory were assessed by using open field test and water T-maze test. This study also examined changes in neuronal cell morphology using cresyl violet and apoptosis staining. We also performed immunohistochemical study to analyse the expression of the glutamate AMPA receptor (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) GluA1 subunit and the GABA receptor (γ-Aminobutyric Acid) subtype GABAA α1 subunit in the hippocampus of the same animals. RESULTS We found no significant changes in locomotor activity (p > 0.05). The water T-maze data showed that 30 mg/kg dose significantly (p < 0.05) improved learning, memory, and the memory consolidation phase but had no effect on reversal learning (p > 0.05). Histological data revealed no neuronal morphological changes. Immunohistochemical analysis revealed increased expression of the AMPA GluA1 receptor subunit but there was no effect on GABAA receptor α1 subunit expression in the CA1 and CA2 subregions of the hippocampus. CONCLUSIONS The C. asiatica extract therefore improved hippocampus-dependent spatial learning and memory in a dose-dependent manner in rats through the GluA1-containing AMPA receptor in the CA1 and CA2 sub regions of the hippocampus.
Collapse
Affiliation(s)
- Nor Aqilah Binti Mohd Yusuf Yeo
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| | - Sangu Muthuraju
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| | - Jia Hui Wong
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| | - Faruque Reza Mohammed
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| | - Mohd Harizal Senik
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| | - Jingli Zhang
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| | | | - Hasnan Jaafar
- Department of Pathology, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia
| | - Mohd Llham Adenan
- Faculty of Applied Sciences, Universiti Teknologi MARA, Shah Alam, Selangor, Malaysia
| | - Habsah Mohamad
- Institute of Marine Biotechnology, Universiti Malaysia Terengganu, Terengganu, Malaysia
| | | | - Jafri Malin Abdullah
- Center for Neuroscience Services and Research(P3Neuro), Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Jalan Hospital Universiti Sains Malaysia, Kota Bharu, Kelantan, Malaysia.,Department of Neurosciences, Hospital Universiti Sains Malaysia, Jalan Hospital USM, Kota Bharu, Kelantan, Malaysia
| |
Collapse
|
29
|
Gray NE, Zweig JA, Caruso M, Martin MD, Zhu JY, Quinn JF, Soumyanath A. Centella asiatica increases hippocampal synaptic density and improves memory and executive function in aged mice. Brain Behav 2018; 8:e01024. [PMID: 29920983 PMCID: PMC6043711 DOI: 10.1002/brb3.1024] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/28/2018] [Revised: 05/07/2018] [Accepted: 05/08/2018] [Indexed: 12/27/2022] Open
Abstract
INTRODUCTION Centella asiatica is a plant used for centuries to enhance memory. We have previously shown that a water extract of Centella asiatica (CAW) attenuates age-related spatial memory deficits in mice and improves neuronal health. Yet the effect of CAW on other cognitive domains remains unexplored as does its mechanism of improving age-related cognitive impairment. This study investigates the effects of CAW on a variety of cognitive tasks as well as on synaptic density and mitochondrial and antioxidant pathways. METHODS Twenty-month-old CB6F1 mice were treated with CAW (2 mg/ml) in their drinking water for 2 weeks prior to behavioral testing. Learning, memory, and executive function were assessed using the novel object recognition task (NORT), object location memory task (OLM), and odor discrimination reversal learning (ODRL) test. Tissue was collected for Golgi analysis of spine density as well as assessment of mitochondrial, antioxidant, and synaptic proteins. RESULTS CAW improved performance in all behavioral tests suggesting effects on hippocampal and cortical dependent memory as well as on prefrontal cortex mediated executive function. There was also an increase in synaptic density in the treated animals, which was accompanied by increased expression of the antioxidant response gene NRF2 as well as the mitochondrial marker porin. CONCLUSIONS These data show that CAW can increase synaptic density as well as antioxidant and mitochondrial proteins and improve multiple facets of age-related cognitive impairment. Because mitochondrial dysfunction and oxidative stress also accompany cognitive impairment in many pathological conditions this suggests a broad therapeutic utility of CAW.
Collapse
Affiliation(s)
- Nora E Gray
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Jonathan A Zweig
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Cell, Developmental and Cancer Biology, Oregon Health and Science University, Portland, Oregon
| | - Maya Caruso
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Marjoen D Martin
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| | - Jennifer Y Zhu
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, Oregon
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon.,Department of Neurology and Parkinson's Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, Oregon
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, Oregon
| |
Collapse
|
30
|
Centella asiatica Attenuates Mitochondrial Dysfunction and Oxidative Stress in A β-Exposed Hippocampal Neurons. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:7023091. [PMID: 28883904 PMCID: PMC5572603 DOI: 10.1155/2017/7023091] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 06/16/2017] [Accepted: 06/27/2017] [Indexed: 12/17/2022]
Abstract
Centella asiatica has been used for centuries to enhance memory. We have previously shown that a water extract of Centella asiatica (CAW) protects against the deleterious effects of amyloid-β (Aβ) in neuroblastoma cells and attenuates Aβ-induced cognitive deficits in mice. Yet, the neuroprotective mechanism of CAW has yet to be thoroughly explored in neurons from these animals. This study investigates the effects of CAW on neuronal metabolism and oxidative stress in isolated Aβ-expressing neurons. Hippocampal neurons from amyloid precursor protein overexpressing Tg2576 mice and wild-type (WT) littermates were treated with CAW. In both genotypes, CAW increased the expression of antioxidant response genes which attenuated the Aβ-induced elevations in reactive oxygen species (ROS) and lipid peroxidation in Tg2576 neurons. CAW also improved mitochondrial function in both genotypes and increased the expression of electron transport chain enzymes and mitochondrial labeling, suggesting an increase in mitochondrial content. These data show that CAW protects against mitochondrial dysfunction and oxidative stress in Aβ-exposed hippocampal neurons which could contribute to the beneficial effects of the extract observed in vivo. Since CAW also improved mitochondrial function in the absence of Aβ, these results suggest a broader utility for other conditions where neuronal mitochondrial dysfunction occurs.
Collapse
|