1
|
Heck KL, Yi Y, Thornton D, Zheng J, Calderón AI. A comparative metabolomics analysis of Açaí (Euterpe oleracea Mart.) fruit, food powder, and botanical dietary supplement extracts. PHYTOCHEMICAL ANALYSIS : PCA 2025; 36:394-408. [PMID: 38965051 DOI: 10.1002/pca.3416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Euterpe oleracea Mart. (açaí) is a botanical of interest to many who seek functional foods that provide antioxidant and anti-inflammatory properties. Cancer patients are increasingly taking botanical dietary supplements containing açaí to complement their conventional therapeutics, which may lead to serious adverse events. Before testing our açaí extracts in vitro for botanical-drug interactions, the goal is to chemically characterize our extracts for compounds whose biological activity in açaí is unknown. OBJECTIVE The objective of this work was to develop a chemical fingerprinting method for untargeted characterization of açaí samples from a variety of sources, including food products and botanical dietary supplement capsules, made with multiple extraction solvents. METHODS An optimized LC-MS method was generated for in-depth untargeted fingerprinting of chemical constituents in açaí extracts. Statistical analysis models were used to describe relationships between the açaí extracts based on molecular features found in both positive and negative mode ESI. RESULTS In an attempt to elucidate the differences in metabolites among açaí extracts from different cultivars, we identified or tentatively identified 173 metabolites from the 16 extracts made from 6 different sources. Of these compounds, there are 138 reported in açaí for the first time. Statistical models showed similar yet distinct differences between the extracts tested based on the polarity of compounds present and the origin of the source material. CONCLUSION A high-resolution mass spectrometry method was generated that allowed us to greatly characterize 16 complex extracts made from different sources of açaí with different extraction solvent polarities.
Collapse
Affiliation(s)
- Kabre L Heck
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Yuyan Yi
- Department of Mathematics and Statistics, College of Science and Mathematics, Auburn University, AL, USA
| | - Destini Thornton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jingyi Zheng
- Department of Mathematics and Statistics, College of Science and Mathematics, Auburn University, AL, USA
| | - Angela I Calderón
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| |
Collapse
|
2
|
Varada S, Chamberlin SR, Bui L, Brandes MS, Gladen-Kolarsky N, Harris CJ, Hack W, Neff CJ, Brumbach BH, Soumyanath A, Quinn JF, Gray NE. Oral Asiatic Acid Improves Cognitive Function and Modulates Antioxidant and Mitochondrial Pathways in Female 5xFAD Mice. Nutrients 2025; 17:729. [PMID: 40005058 PMCID: PMC11858387 DOI: 10.3390/nu17040729] [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: 01/23/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Extracts of the plant Centella asiatica can enhance mitochondrial function, promote antioxidant activity and improve cognitive deficits. Asiatic acid (AA) is one of the constituent triterpene compounds present in the plant. In this study, we explore the effects of AA on brain mitochondrial function, antioxidant response and cognition in a beta-amyloid (Aβ)-overexpressing 5xFAD mouse line. Methods: Six- to seven-month-old 5xFAD mice were treated with 1% AA for 4 weeks. In the last week of treatment, associative memory was assessed along with mitochondrial bioenergetics and the expression of mitochondrial and antioxidant response genes from isolated cortical synaptosomes. The Aβ plaque burden was also evaluated. Results: AA treatment resulted in improvements in associative memory in female 5xFAD mice without altering the Aβ plaque burden. Cortical mitochondrial function and mitochondrial gene expression were increased in the AA-treated female 5xFAD mice, as was the expression of antioxidant genes. More modest effects of AA on cortical mitochondrial function and mitochondrial and antioxidant gene expression were observed in male 5xFAD mice. Conclusions: Oral AA treatment improved cognitive and mitochondrial function and activated antioxidant in Aβ-overexpressing mice. These changes occurred independent of alterations in Aβ plaque burden, suggesting that AA could have translational therapeutic relevance in later-stage AD when plaques are well established.
Collapse
Affiliation(s)
- Samantha Varada
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Stephen R. Chamberlin
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Lillie Bui
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Mikah S. Brandes
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Noah Gladen-Kolarsky
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Christopher J. Harris
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Wyatt Hack
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Cody J. Neff
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Barbara H. Brumbach
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
- Department of Neurology and Parkinson’s Disease Research Education and Clinical Care Center (PADRECC), VA Portland Healthcare System, Portland, OR 97239, USA
| | - Nora E. Gray
- Department of Neurology, Oregon Health and Science University, Portland, OR 97239, USA; (S.V.); (A.S.); (J.F.Q.)
| |
Collapse
|
3
|
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
|
4
|
Matthews DG, Khorani M, Bobe G, Caruso M, Magana AA, Gray NE, Quinn JF, Stevens JF, Maier CS, Soumyanath A. Centella asiatica improves cognitive function and alters the hippocampal metabolome of aged Tg2576 and wild-type mice. J Alzheimers Dis Rep 2024; 8:1611-1638. [PMID: 40034352 PMCID: PMC11863750 DOI: 10.1177/25424823241296740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 09/23/2024] [Indexed: 03/05/2025] Open
Abstract
Background Alzheimer's disease (AD) is a growing public health problem in the aging population, with limited treatment options. We previously reported that Centella asiatica herb water extract (CAW) attenuates cognitive decline in murine models of AD and aging. Objective To explore changes in the hippocampal metabolome associated with CAW's modulation of cognitive function and amyloid-β (Aβ) plaque load in aged Tg2576 and wild-type (WT) mice. Methods We compared cognitive function, hippocampal Aβ plaque burden, and hippocampal metabolite profile in 20-month-old Tg2576 female mice and their WT littermates following 3-5 weeks treatment with CAW (0, 200, or 1000 mg/kg/d p.o.). Cognitive testing included contextual fear response (CFR) and novel object recognition task (NORT). Aβ plaque burden was measured via immunohistochemistry. Metabolomic profiles of mouse hippocampi were obtained using liquid chromatography coupled with high resolution tandem mass spectrometry. Results CAW treatment resulted in dose-related improvements in CFR and NORT performance of Tg2576 and WT mice. However, while CFR correlated with neurosignaling and glycosylated ceramide levels, NORT performance correlated with lysophosphatidylcholines and oxidized metabolites, and Aβ accumulation was linked to elevated excitatory and suppressed inhibitory neurotransmission. Only a subset of the metabolite changes induced by CAW in Tg2576 mice represented a reversal of metabolite differences between Tg2576 and WT mice, suggesting the involvement of other pathways in CAW's cognitive effects. Conclusions Mechanisms underlying CAW's cognitive effects extend beyond reversing metabolic effects of Aβ accumulation. The data support the potential use of CAW to manage memory challenges in aged individuals with or without AD.
Collapse
Affiliation(s)
- Donald G Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
| | - Mona Khorani
- Department of Chemistry, Oregon State University, Corvallis, OR, USA
| | - Gerd Bobe
- 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
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
| | - Joseph F Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, 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
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & 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 & Science University, Portland, OR, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, USA
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
5
|
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
|
6
|
McAfee A, Magaña AA, Foster LJ, Hoover SE. Differences in honeybee queen pheromones revealed by LC-MS/MS: Reassessing the honest signal hypothesis. iScience 2024; 27:110906. [PMID: 39391732 PMCID: PMC11465126 DOI: 10.1016/j.isci.2024.110906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/08/2024] [Accepted: 09/05/2024] [Indexed: 10/12/2024] Open
Abstract
The honest signal hypothesis posits that social insect queens emit pheromonal signals that convey information about fecundity, which workers use to make decisions around investing in direct or indirect fitness. We used liquid chromatography-tandem mass spectrometry to measure honeybee (Apis mellifera) queen retinue pheromone (QRP) in relation to age, laying status, and acceptance using a protocol that enables concurrent metabolomic and lipidomic analyses. Older queens produced higher levels of the QRP components 9-R-hydroxydec-2(E)-enoic acid (9(R)-HDA), linolenic acid (LEA), and 4-hydroxy-3-methoxyphenylethanol (HVA) compared to younger queens, with HVA also correlating with ovary mass. However, ovary mass was not an influential metric for worker decision-making around queen acceptance; therefore, the relationship between HVA and ovary mass is merely an "honest signal" of a non-influential metric. Parallel metabolomic and lipidomic analyses showed that samples cluster according to queen age and mating status, but not ovary mass, revealing many other physiological changes occurring in the queen's early life.
Collapse
Affiliation(s)
- Alison McAfee
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Department of Applied Ecology, North Carolina State University, Raleigh, NC, USA
| | - Armando Alcazar Magaña
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Leonard J. Foster
- Michael Smith Laboratories, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
- Life Sciences Institute, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Shelley E. Hoover
- Department of Biological Sciences, University of Lethbridge, Lethbridge, AB, Canada
| |
Collapse
|
7
|
Black B, da Silva LBR, Hu G, Qu X, Smith DFQ, Magaña AA, Horianopoulos LC, Caza M, Attarian R, Foster LJ, Casadevall A, Kronstad JW. Glutathione-mediated redox regulation in Cryptococcus neoformans impacts virulence. Nat Microbiol 2024; 9:2084-2098. [PMID: 38956248 PMCID: PMC11930340 DOI: 10.1038/s41564-024-01721-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 05/07/2024] [Indexed: 07/04/2024]
Abstract
The fungal pathogen Cryptococcus neoformans is well adapted to its host environment. It has several defence mechanisms to evade oxidative and nitrosative agents released by phagocytic host cells during infection. Among them, melanin production is linked to both fungal virulence and defence against harmful free radicals that facilitate host innate immunity. How C. neoformans manipulates its redox environment to facilitate melanin formation and virulence is unclear. Here we show that the antioxidant glutathione is inextricably linked to redox-active processes that facilitate melanin and titan cell production, as well as survival in macrophages and virulence in a murine model of cryptococcosis. Comparative metabolomics revealed that disruption of glutathione biosynthesis leads to accumulation of reducing and acidic compounds in the extracellular environment of mutant cells. Overall, these findings highlight the importance of redox homeostasis and metabolic compensation in pathogen adaptation to the host environment and suggest new avenues for antifungal drug development.
Collapse
Affiliation(s)
- Braydon Black
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Leandro Buffoni Roque da Silva
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Guanggan Hu
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xianya Qu
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Daniel F Q Smith
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Armando Alcázar Magaña
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Metabolomics Core Facility, Life Sciences Institute, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Linda C Horianopoulos
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Wisconsin Energy Institute, University of Wisconsin-Madison, Madison, WI, USA
| | - Mélissa Caza
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Larissa Yarr Medical Microbiology Laboratory, Kelowna General Hospital, Kelowna, British Columbia, Canada
| | - Rodgoun Attarian
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Pfizer Canada, Kirkland, Quebec, Canada
| | - Leonard J Foster
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
- Metabolomics Core Facility, Life Sciences Institute, Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Arturo Casadevall
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - James W Kronstad
- The Michael Smith Laboratories, Departments of Microbiology and Immunology, and Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
8
|
Rowe K, Gray NE, Zweig JA, Law A, Techen N, Maier CS, Soumyanath A, Kretzschmar D. Centella asiatica and its caffeoylquinic acid and triterpene constituents increase dendritic arborization of mouse primary hippocampal neurons and improve age-related locomotion deficits in Drosophila. FRONTIERS IN AGING 2024; 5:1374905. [PMID: 39055970 PMCID: PMC11269084 DOI: 10.3389/fragi.2024.1374905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Accepted: 06/12/2024] [Indexed: 07/28/2024]
Abstract
Introduction Centella asiatica (CA) is known in Ayurvedic medicine as a rejuvenating herb with particular benefits in the nervous system. Two groups of specialized metabolites found in CA and purported to contribute to its beneficial effects are triterpenes (TTs) and caffeoylquinic acids (CQAs). In order to evaluate the role and interactions of TTs and CQAs in the effects of CA, we examined the neurotrophic effects of a water extract of CA (CAW) and combinations of its TT and CQA components in mouse primary hippocampal neurons in vitro and in Drosophila melanogaster flies in vivo. Methods Primary hippocampal neurons were isolated from mouse embryos and exposed in vitro for 5 days to CAW (50 μg/mL), mixtures of TTs, CQAs or TT + CQA components or to 4 TTs or 8 individual CQA compounds of CAW. Dendritic arborization was evaluated using Sholl analysis. Drosophila flies were aged to 28 days and treated for 2 weeks with CAW (10 mg/mL) in the food, mixtures of TTs, CQAs or TT + CQA and individual TT and CQA compounds. TTs and CQAs were tested at concentrations matching their levels in the CAW treatment used. After 2 weeks of treatment, Drosophila aged 42 days were evaluated for phototaxis responses. Results In mouse primary hippocampal neurons, CAW (50 μg/mL), the TT mix, CQA mix, all individual TTs and most CQAs significantly increased dendritic arborization to greater than control levels. However, the TT + CQA combination significantly decreased dendritic arborization. In Drosophila, a marked age-related decline in fast phototaxis response was observed in both males and females over a 60 days period. However, resilience to this decline was afforded in both male and female flies by treatment from 28 days onwards with CAW (10 mg/mL), or equivalent concentrations of mixed TTs, mixed CQAs and a TT + CQA mix. Of all the individual compounds, only 1,5-diCQA slowed age-related decline in phototaxis in male and female flies. Discussion This study confirmed the ability of CAW to increase mouse neuronal dendritic arborization, and to provide resilience to age-related neurological decline in Drosophila. The TT and CQA components both contribute to these effects but do not have a synergistic effect. While individual TTs and most individual CQAs increased dendritic arborization at CAW equivalent concentrations, in the Drosophila model, only 1,5-diCQA was able to slow down the age-related decline in phototaxis. This suggests that combinations (or potentially higher concentrations) of the other compounds are needed to provide resilience in this model.
Collapse
Affiliation(s)
- Karon Rowe
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Jonathan A. Zweig
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Alexander Law
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Natascha Techen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- National Center for Natural Products Research, University of Mississippi, Oxford, MS, United States
| | - Claudia S. Maier
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Department of Neurology, Oregon Health and Science University, Portland, OR, United States
| | - Doris Kretzschmar
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR, United States
- Oregon Institute of Occupational Health Sciences, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
9
|
Gray NE, Hack W, Brandes MS, Zweig JA, Yang L, Marney L, Choi J, Magana AA, Cerruti N, McFerrin J, Koike S, Nguyen T, Raber J, Quinn JF, Maier CS, Soumyanath A. Amelioration of age-related cognitive decline and anxiety in mice by Centella asiatica extract varies by sex, dose and mode of administration. FRONTIERS IN AGING 2024; 5:1357922. [PMID: 38770167 PMCID: PMC11102990 DOI: 10.3389/fragi.2024.1357922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 04/10/2024] [Indexed: 05/22/2024]
Abstract
Background: A water extract (CAW) of the Ayurvedic plant Centella asiatica administered in drinking water has been shown to improve cognitive deficits in mouse models of aging and neurodegenerative diseases. Here the effects of CAW administered in drinking water or the diet on cognition, measures of anxiety and depression-like behavior in healthy aged mice are compared. Methods: Three- and eighteen-month-old male and female C57BL6 mice were administered rodent AIN-93M diet containing CAW (0, 0.2, 0.5 or 1% w/w) to provide 0, 200 mg/kg/d, 500 mg/kg/d or 1,000 mg/kg/d CAW for a total of 5 weeks. An additional group of eighteen-month-old mice were treated with CAW (10 mg/mL) in their drinking water CAW for a total of 5 weeks to deliver the same exposure of CAW as the highest dietary dose (1,000 mg/kg/d). CAW doses delivered were calculated based on food and water consumption measured in previous experiments. In the fourth and fifth weeks, mice underwent behavioral testing of cognition, anxiety and depression (n = 12 of each sex per treatment group in each test). Results: Aged mice of both sexes showed cognitive deficits relative to young mice while only female aged mice showed increased anxiety compared to the young female mice and no differences in depression were observed between the different ages. CAW (1,000 mg/kg/d) in the drinking water improved deficits in aged mice in learning, executive function and recognition memory in both sexes and attenuated the increased measures of anxiety observed in the aged female mice. However, CAW in the diet only improved executive function in aged mice at the highest dose (1,000 mg/kg/d) in both sexes and did so less robustly than when given in the water. There were no effects of CAW on depression-like behavior in aged animals regardless of whether it was administered in the diet or the water. Conclusions: These results suggest that CAW can ameliorate age-related changes in measures of anxiety and cognition and that the mode of administration is important for the effects of CAW on resilience to these age-related changes.
Collapse
Affiliation(s)
- Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Wyatt Hack
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Mikah S. Brandes
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Jonathan A. Zweig
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Liping Yang
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Luke Marney
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Jaewoo Choi
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Armando Alcazar Magana
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Natasha Cerruti
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Oregon’s Wild Harvest, Redmond, OR, United States
| | - Janis McFerrin
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Oregon’s Wild Harvest, Redmond, OR, United States
| | - Seiji Koike
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Thuan Nguyen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Jacob Raber
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Behavioral Neuroscience, Oregon Health & Science University, Portland, OR, United States
| | - Joseph F. Quinn
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Healthcare System, Portland, OR, United States
| | - Claudia S. Maier
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
10
|
Speers AB, Wright KM, Brandes MS, Kedjejian N, Matthews DG, Caruso M, Harris CJ, Koike S, Nguyen T, Quinn JF, Soumyanath A, Gray NE. Mode of administration influences plasma levels of active Centella asiatica compounds in 5xFAD mice while markers of neuroinflammation remain unaltered. Front Neurosci 2024; 18:1277626. [PMID: 38591068 PMCID: PMC10999680 DOI: 10.3389/fnins.2024.1277626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 03/13/2024] [Indexed: 04/10/2024] Open
Abstract
Introduction A water extract of Centella asiatica (L.) Urban [Apiaceae] (CAW) has demonstrated cognitive-enhancing effects in mouse models of Alzheimer's disease and aging, the magnitude of which is influenced by whether CAW is delivered in the drinking water or the diet. These cognitive benefits are accompanied by improvements in oxidative stress and mitochondrial function in the brain, two pathways related to the neuroinflammatory response. The effect of CAW on neuroinflammation, however, has not been directly studied. Here, we investigated the effect of CAW on neuroinflammation in 5xFAD mice and compared plasma levels of CAW's active compounds following two modes of CAW administration. Methods Eight-to-nine-month-old male and female 5xFAD mice and their wild-type littermates were administered CAW in their diet or drinking water (0 or 1,000 mg/kg/day) for five weeks. Immunohistochemistry was performed for β-amyloid (Aβ), glial fibrillary acidic protein (GFAP), and Griffonia simplicifolia lectin I (GSL I) in the cortex and hippocampus. Gene expression of inflammatory mediators (IL-6, TNFα, IL-1β, TREM2, AIF1, CX3CR1, CX3CL1, CD36, C3AR1, RAGE, CCR6, CD3E) was measured in the deep grey matter. Results CAW decreased cortical Aβ plaque burden in female 5xFAD mice administered CAW in the drinking water but had no effect on Aβ plaques in other treatment groups. CAW did not impact elevated levels of GFAP or GSL I in 5xFAD mice, regardless of sex, brain region, or mode of CAW administration. In the deep grey matter, CAW increased C3AR1 expression in 5xFAD females administered CAW in the drinking water and decreased IL-1β expression in 5xFAD males administered CAW in the diet. CAW had no effect, however, on gene expression levels of any other inflammatory mediator in the deep grey, for either sex or mode of CAW administration. Mice administered CAW in the drinking water versus the diet had significantly higher plasma levels of CAW compounds. Discussion CAW had little impact on the neuroinflammatory markers selected for evaluation in the present study, suggesting that the cognitive benefits of CAW may not be mediated by an anti-inflammatory effect or that additional molecular markers are needed to fully characterize the effect of CAW on neuroinflammation.
Collapse
Affiliation(s)
- Alex B. Speers
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Kirsten M. Wright
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Mikah S. Brandes
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nareg Kedjejian
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Christopher J. Harris
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Seiji Koike
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Thuan Nguyen
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- School of Public Health, Oregon Health & Science University-Portland State University, Portland, OR, United States
| | - Joseph F. Quinn
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, United States
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR, United States
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
11
|
Alcázar Magaña A, Vaswani A, Brown KS, Jiang Y, Alam MN, Caruso M, Lak P, Cheong P, Gray NE, Quinn JF, Soumyanath A, Stevens JF, Maier CS. Integrating High-Resolution Mass Spectral Data, Bioassays and Computational Models to Annotate Bioactives in Botanical Extracts: Case Study Analysis of C. asiatica Extract Associates Dicaffeoylquinic Acids with Protection against Amyloid-β Toxicity. Molecules 2024; 29:838. [PMID: 38398590 PMCID: PMC10892090 DOI: 10.3390/molecules29040838] [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: 12/30/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Rapid screening of botanical extracts for the discovery of bioactive natural products was performed using a fractionation approach in conjunction with flow-injection high-resolution mass spectrometry for obtaining chemical fingerprints of each fraction, enabling the correlation of the relative abundance of molecular features (representing individual phytochemicals) with the read-outs of bioassays. We applied this strategy for discovering and identifying constituents of Centella asiatica (C. asiatica) that protect against Aβ cytotoxicity in vitro. C. asiatica has been associated with improving mental health and cognitive function, with potential use in Alzheimer's disease. Human neuroblastoma MC65 cells were exposed to subfractions of an aqueous extract of C. asiatica to evaluate the protective benefit derived from these subfractions against amyloid β-cytotoxicity. The % viability score of the cells exposed to each subfraction was used in conjunction with the intensity of the molecular features in two computational models, namely Elastic Net and selectivity ratio, to determine the relationship of the peak intensity of molecular features with % viability. Finally, the correlation of mass spectral features with MC65 protection and their abundance in different sub-fractions were visualized using GNPS molecular networking. Both computational methods unequivocally identified dicaffeoylquinic acids as providing strong protection against Aβ-toxicity in MC65 cells, in agreement with the protective effects observed for these compounds in previous preclinical model studies.
Collapse
Affiliation(s)
- Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (A.V.); (M.N.A.); (P.L.); (P.C.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (N.E.G.); (A.S.); (J.F.S.)
- Life Sciences Institute, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Ashish Vaswani
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (A.V.); (M.N.A.); (P.L.); (P.C.)
| | - Kevin S. Brown
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA;
- School of Chemical, Biological, and Environmental Engineering, Oregon State University, 116 Johnson Hall, 105 SW 26th Street, Corvallis, OR 97331, USA
| | - Yuan Jiang
- Department of Statistics, Oregon State University, Corvallis, OR 97331, USA;
| | - Md Nure Alam
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (A.V.); (M.N.A.); (P.L.); (P.C.)
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (M.C.); (J.F.Q.)
| | - Parnian Lak
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (A.V.); (M.N.A.); (P.L.); (P.C.)
| | - Paul Cheong
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (A.V.); (M.N.A.); (P.L.); (P.C.)
| | - Nora E. Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (N.E.G.); (A.S.); (J.F.S.)
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (M.C.); (J.F.Q.)
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (M.C.); (J.F.Q.)
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR 97239, USA
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (N.E.G.); (A.S.); (J.F.S.)
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (M.C.); (J.F.Q.)
| | - Jan F. Stevens
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (N.E.G.); (A.S.); (J.F.S.)
- 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.); (A.V.); (M.N.A.); (P.L.); (P.C.)
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA; (N.E.G.); (A.S.); (J.F.S.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| |
Collapse
|
12
|
Yang L, Marney L, Magana AA, Choi J, Wright K, Mcferrin J, Gray NE, Soumyanath A, Stevens JF, Maier CS. Quantification of Caffeoylquinic Acids and Triterpenes as Targeted Bioactive Compounds of Centella asiatica in Extracts and Formulations by Liquid Chromatography Mass Spectrometry. JOURNAL OF CHROMATOGRAPHY OPEN 2023; 4:100091. [PMID: 37789901 PMCID: PMC10544816 DOI: 10.1016/j.jcoa.2023.100091] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Centella asiatica (CA) is a culinary vegetable and well-known functional food that is widely used as a medicinal herb and dietary supplement. CA is rich in pentacyclic triterpenes (TTs), including asiaticoside (AS), madecassoside (MS) and the related aglycones asiatic acid (AA), madecassic acid (MA). Traditionally, TTs have been associated with the bioactivity and health promoting effect of CA. Recently, mono-caffeoylquinic acids (MonoCQAs) and di-caffeoylquinic acids (DiCQAs) have been found to contribute to the bioactivity of CA as well. This work reports an analytical strategy based on liquid chromatography coupled to multiple reaction monitoring mass spectrometry (LC-MRM-MS) for the simultaneous rapid and accurate quantification of 12 bioactive compounds in CA, namely AS, MS, AA, MA, 5-CQA, 4-CQA, 3-CQA, 1,3-DiCQA, 3,4-DiCQA, 1,5-DiCQA, 3,5-DiCQA, 4,5-DiCQA. Method selectivity, accuracy, precision, repeatability, robustness, linearity range, limit of detection (LOD), and limit of quantitation (LOQ) were validated. The validated LC-MRM-MS method has been successfully applied to quantify the 12 bioactive compounds in CA aqueous extracts and two related formulations: a standardized CA product (CAP) used in a phase I clinical trial and formulated CA rodent diets used in preclinical studies. The validated method allows us to support the standardization of CA products used for clinical trials and conduct routine LC-MRM-MS analyses of formulated preclinical diets to confirm correct levels of CA phytochemical markers.
Collapse
Affiliation(s)
- Liping Yang
- Department of Chemistry, Oregon State University, Corvallis, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
| | - Luke Marney
- Department of Chemistry, Oregon State University, Corvallis, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
| | - Armando Alcazar Magana
- Department of Chemistry, Oregon State University, Corvallis, OR
- Linus Pauling Institute, Oregon State University, Corvallis, OR
- Life Sciences Institute, University of British Columbia, Vancouver BC, Canada
| | - Jaewoo Choi
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
- Linus Pauling Institute, Oregon State University, Corvallis, OR
| | - Kirsten Wright
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
- Department of Neurology, Oregon Health and Science University, Portland, OR
| | | | - Nora E Gray
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
- Department of Neurology, Oregon Health and Science University, Portland, OR
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
- Department of Neurology, Oregon Health and Science University, Portland, OR
| | - Jan F Stevens
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
- Linus Pauling Institute, Oregon State University, Corvallis, OR
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR
| | - Claudia S Maier
- Department of Chemistry, Oregon State University, Corvallis, OR
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health and Science University, Portland, OR
- Linus Pauling Institute, Oregon State University, Corvallis, OR
| |
Collapse
|
13
|
Holvoet H, Long DM, Yang L, Choi J, Marney L, Poeck B, Maier CS, Soumyanath A, Kretzschmar D, Strauss R. Chlorogenic Acids, Acting via Calcineurin, Are the Main Compounds in Centella asiatica Extracts That Mediate Resilience to Chronic Stress in Drosophila melanogaster. Nutrients 2023; 15:4016. [PMID: 37764799 PMCID: PMC10537055 DOI: 10.3390/nu15184016] [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: 08/18/2023] [Revised: 09/13/2023] [Accepted: 09/15/2023] [Indexed: 09/29/2023] Open
Abstract
Common symptoms of depressive disorders include anhedonia, sleep problems, and reduced physical activity. Drugs used to treat depression mostly aim to increase serotonin signaling but these can have unwanted side effects. Depression has also been treated by traditional medicine using plants like Centella asiatica (CA) and this has been found to be well tolerated. However, very few controlled studies have addressed CA's protective role in depression, nor have the active compounds or mechanisms that mediate this function been identified. To address this issue, we used Drosophila melanogaster to investigate whether CA can improve depression-associated symptoms like anhedonia and decreased climbing activity. We found that a water extract of CA provides resilience to stress induced phenotypes and that this effect is primarily due to mono-caffeoylquinic acids found in CA. Furthermore, we describe that the protective function of CA is due to a synergy between chlorogenic acid and one of its isomers also present in CA. However, increasing the concentration of chlorogenic acid can overcome the requirement for the second isomer. Lastly, we found that chlorogenic acid acts via calcineurin, a multifunctional phosphatase that can regulate synaptic transmission and plasticity and is also involved in neuronal maintenance.
Collapse
Affiliation(s)
- Helen Holvoet
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, 55128 Mainz, Germany; (H.H.)
| | - Dani M. Long
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA (L.Y.); (J.C.); (A.S.)
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Liping Yang
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA (L.Y.); (J.C.); (A.S.)
- 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.Y.); (J.C.); (A.S.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Luke Marney
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA (L.Y.); (J.C.); (A.S.)
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
| | - Burkhard Poeck
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, 55128 Mainz, Germany; (H.H.)
| | - Claudia S. Maier
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA (L.Y.); (J.C.); (A.S.)
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Amala Soumyanath
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA (L.Y.); (J.C.); (A.S.)
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA
| | - Doris Kretzschmar
- BENFRA Botanical Dietary Supplements Research Center, Oregon Health & Science University, Portland, OR 97239, USA (L.Y.); (J.C.); (A.S.)
- Oregon Institute of Occupational Health Sciences, Oregon Health & Science University, Portland, OR 97239, USA
| | - Roland Strauss
- Institut für Entwicklungsbiologie und Neurobiologie, Johannes Gutenberg-Universität Mainz, 55128 Mainz, Germany; (H.H.)
| |
Collapse
|
14
|
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: 2] [Impact Index Per Article: 1.0] [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
|
15
|
Bouranis JA, Beaver LM, Jiang D, Choi J, Wong CP, Davis EW, Williams DE, Sharpton TJ, Stevens JF, Ho E. Interplay between Cruciferous Vegetables and the Gut Microbiome: A Multi-Omic Approach. Nutrients 2022; 15:nu15010042. [PMID: 36615700 PMCID: PMC9824405 DOI: 10.3390/nu15010042] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/14/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Brassica vegetables contain a multitude of bioactive compounds that prevent and suppress cancer and promote health. Evidence suggests that the gut microbiome may be essential in the production of these compounds; however, the relationship between specific microbes and the abundance of metabolites produced during cruciferous vegetable digestion are still unclear. We utilized an ex vivo human fecal incubation model with in vitro digested broccoli sprouts (Broc), Brussels sprouts (Brus), a combination of the two vegetables (Combo), or a negative control (NC) to investigate microbial metabolites of cruciferous vegetables. We conducted untargeted metabolomics on the fecal cultures by LC-MS/MS and completed 16S rRNA gene sequencing. We identified 72 microbial genera in our samples, 29 of which were significantly differentially abundant between treatment groups. A total of 4499 metabolomic features were found to be significantly different between treatment groups (q ≤ 0.05, fold change > 2). Chemical enrichment analysis revealed 45 classes of compounds to be significantly enriched by brassicas, including long-chain fatty acids, coumaric acids, and peptides. Multi-block PLS-DA and a filtering method were used to identify microbe−metabolite interactions. We identified 373 metabolites from brassica, which had strong relationships with microbes, such as members of the family Clostridiaceae and genus Intestinibacter, that may be microbially derived.
Collapse
Affiliation(s)
- John A. Bouranis
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Laura M. Beaver
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Duo Jiang
- Department of Statistics, Oregon State University, Corvallis, OR 97331, USA
| | - Jaewoo Choi
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Carmen P. Wong
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Edward W. Davis
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Center for Quantitative Life Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - David E. Williams
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR 97331, USA
| | - Thomas J. Sharpton
- Department of Statistics, Oregon State University, Corvallis, OR 97331, USA
- Department of Microbiology, Oregon State University, Corvallis, OR 97331, USA
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA
| | - Emily Ho
- School of Biological and Population Health Sciences, Oregon State University, Corvallis, OR 97331, USA
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
- Correspondence:
| |
Collapse
|
16
|
Paemanee A, Rattanabunyong S, Ketngamkum Y, Siriwaseree J, Pongpamorn P, Romyanon K, Tangphatsornruang S, Kuaprasert B, Choowongkomon K. Mass spectrometry and synchrotron-FTIR microspectroscopy reveal the anti-inflammatory activity of Bua Bok extracts. PHYTOCHEMICAL ANALYSIS : PCA 2022; 33:1086-1098. [PMID: 35790045 DOI: 10.1002/pca.3161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 06/15/2022] [Accepted: 06/16/2022] [Indexed: 06/15/2023]
Abstract
INTRODUCTION Bua Bok or Centella asiatica (CA) is an Asian vegetable with anti-inflammatory benefits. Asiaticoside, asiatic acid, madecassoside and madecassic have been characterised as major active ingredients with a wide range of pharmacological advantages. In manufacturing processes, high-performance liquid chromatography (HPLC) and liquid chromatography-mass spectrometry (LCMS) are used to routinely determine the active compounds in raw materials. OBJECTIVES This research aims to explore anti-inflammatory properties, characterise metabolites and observe the biochemical changes of the inflammatory induced macrophages after pretreatment with the potential extracted fractions. METHODS Bua Bok leaf extracts were prepared. Macrophages were pretreated with non-toxic fractions to determine the anti-inflammatory action. Tentative metabolites of effective fractions were identified by LC-MS. Synchrotron fourier-transform infrared (S-FTIR) microspectroscopy was utilised to observe the biochemical change of the lipopolysaccharide (LPS)-induced cells after pretreatment with potential fractions. RESULTS Fractions of ethyl acetate, 30% and 100% ethanol highly increased the nitrile scavenging and suppressed the function of phospholipase A2 . Fractions of 70% and 100% ethanol strongly decreased nitric oxide production. The comparison of 39 chemical compounds was presented. The change of proteins was improved after pretreatment of macrophages with fraction 70% ethanol. Fraction of 100% ethanol revealed the lipid accumulation was lower than 70% ethanol and diclofenac. CONCLUSION While the anti-inflammatory actions of 70% and 100% ethanol were similar. S-FTIR expressed they inhibited inflammatory response with the distinct features of biomolecules. The S-FTIR, LC-MS and biological assay confidently provided the efficient strategies to inform the advantage of herbal extract on cellular organisation instead of a single compound.
Collapse
Affiliation(s)
- Atchara Paemanee
- National Omics Centre, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | | | - Yanisa Ketngamkum
- National Omics Centre, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | | | - Ponkanok Pongpamorn
- National Omics Centre, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | - Kanokwan Romyanon
- National Centre for Genetic Engineering and Biotechnology, National Science and Technology Development Agency (NSTDA), Pathum Thani, Thailand
| | | | - Buabarn Kuaprasert
- Research Facility Department, Synchrotron Light Research Institute (Public Organisation), Nakhon Ratchasima, Thailand
| | - Kiattawee Choowongkomon
- Department of Biochemistry, Faculty of Science, Kasetsart University, Bangkok, Thailand
- Centre for Advanced Studies in Nanotechnology for Chemical, Food and Agricultural Industries, KU Institute for Advanced Studies, Kasetsart University, Bangkok, Thailand
| |
Collapse
|
17
|
Keller RM, Beaver LM, Prater MC, Truong L, Tanguay RL, Stevens JF, Hord NG. Nitrate exposure reprograms hepatic amino acid and nutrient sensing pathways prior to exercise: A metabolomic and transcriptomic investigation in zebrafish (Danio rerio). Front Mol Biosci 2022; 9:903130. [PMID: 35928228 PMCID: PMC9343839 DOI: 10.3389/fmolb.2022.903130] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 06/28/2022] [Indexed: 11/13/2022] Open
Abstract
Scope: Nitrate supplementation is a popular ergogenic aid that improves exercise performance by reducing oxygen consumption during exercise. We investigated the effect of nitrate exposure and exercise on metabolic pathways in zebrafish liver.Materials and methods: Fish were exposed to sodium nitrate (606.9 mg/L), or control water, for 21 days and analyzed at intervals during an exercise test. We utilized untargeted liquid chromatography-tandem mass spectrometry (LC-MS/MS) analysis and measured gene expression of 24 genes central to energy metabolism and redox signaling.Results: We observed a greater abundance of metabolites involved in endogenous nitric oxide (NO) metabolism and amino acid metabolism in nitrate-treated liver at rest, compared to rested controls. In the absence of exercise, nitrate treatment upregulated expression of genes central to nutrient sensing (pgc1a), protein synthesis (mtor) and purine metabolism (pnp5a and ampd1) and downregulated expression of genes involved in mitochondrial fat oxidation (acaca and cpt2).Conclusion: Our data support a role for sub-chronic nitrate treatment in the improvement of exercise performance, in part, by improving NO bioavailability, sparing arginine, and modulating hepatic gluconeogenesis and glycolytic capacity in the liver.
Collapse
Affiliation(s)
- Rosa M. Keller
- University of California, San Francisco, San Francisco, CA, United States
| | - Laura M. Beaver
- University of California, San Francisco, San Francisco, CA, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
| | - Mary C. Prater
- Department of Foods and Nutrition, College of Family and Consumer Sciences, University of Georgia, Athens, GA, United States
| | - Lisa Truong
- Sinnhuber Aquatic Research Laboratory and the Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Robyn L. Tanguay
- Sinnhuber Aquatic Research Laboratory and the Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- College of Pharmacy, Oregon State University, Corvallis, OR, United States
| | - Norman G. Hord
- OU Health, Harold Hamm Diabetes Center, Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
- *Correspondence: Norman G. Hord,
| |
Collapse
|
18
|
Huang L, Alcazar Magana A, Skinkis PA, Osborne J, Qian YL, Qian MC. Composition of Pinot Noir Wine from Grapevine Red Blotch Disease-Infected Vines Managed with Exogenous Abscisic Acid Applications. Molecules 2022; 27:molecules27144520. [PMID: 35889392 PMCID: PMC9321972 DOI: 10.3390/molecules27144520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/25/2022] [Accepted: 07/08/2022] [Indexed: 12/10/2022] Open
Abstract
Grapevine red blotch disease (GRBD) has negative effects on grape development and impacts berry ripening. Abscisic acid (ABA) is a plant growth regulator involved in the initiation of berry ripening. Exogenous abscisic acid application was compared to an unsprayed control on GRBD-positive Pinot noir vines during two vintages, and the total monomeric anthocyanin, total phenolics, phenolic composition, and volatile profile were measured in wines. In addition, untargeted metabolites were profiled using high-resolution LC-MS/MS. Results showed that the wine composition varied by vintage year and was not consistent with ABA application. Wines from the ABA treatment had a lower total anthocyanin and total phenolic content in one year. The untargeted high-resolution LC-MS/MS analysis showed a higher abundance of phenolic compounds in ABA wines in 2019, but lower in 2018. The wine volatile compounds of ABA treatments varied by vintage. There were higher levels of free β-damascenone, β-ionone, nerol, and several fermentation-derived esters, acids, and alcohols in ABA wines, but these were not observed in 2019. Lower 3-isobutyl-2-methoxypyrazine (IBMP) was also observed in wines with ABA treatment in 2019. The results demonstrated that ABA application to the fruit zones did not consistently mitigate the adverse impacts of GRBD on Pinot noir wines.
Collapse
Affiliation(s)
- Ling Huang
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA; (L.H.); (A.A.M.); (J.O.); (Y.L.Q.)
| | - Armando Alcazar Magana
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA; (L.H.); (A.A.M.); (J.O.); (Y.L.Q.)
| | - Patricia A. Skinkis
- Department of Horticulture, Oregon State University, Corvallis, OR 97331, USA;
- Oregon Wine Research Institute, Oregon State University, Corvallis, OR 97331, USA
| | - James Osborne
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA; (L.H.); (A.A.M.); (J.O.); (Y.L.Q.)
- Oregon Wine Research Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Yanping L. Qian
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA; (L.H.); (A.A.M.); (J.O.); (Y.L.Q.)
| | - Michael C. Qian
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97331, USA; (L.H.); (A.A.M.); (J.O.); (Y.L.Q.)
- Oregon Wine Research Institute, Oregon State University, Corvallis, OR 97331, USA
- Correspondence: ; Tel.: +1-541-737-9114
| |
Collapse
|
19
|
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: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/14/2022] [Accepted: 01/17/2022] [Indexed: 11/24/2022] Open
Abstract
Centella asiatica is reputed in Eastern medicine to improve cognitive function in humans. Preclinical studies have demonstrated that aqueous extracts of C. asiatica improve cognition in mouse models of aging and Alzheimer's disease (AD) through the modulation of mitochondrial biogenesis and nuclear factor-erythroid-2-related factor 2 (Nrf2)-dependent antioxidant response genes. This randomized, double-blind, crossover Phase I trial explored the oral bioavailability and pharmacokinetics of key compounds from two doses (2 g and 4 g) of a standardized C. asiatica aqueous extract product (CAP), over 10 h, in four mildly demented older adults on cholinesterase inhibitor therapy. The analysis focused on triterpenes (TTs) and caffeoylquinic acids (CQAs), which are known to contribute to C. asiatica's neurological activity. The acute safety of CAP and the effects on NRF2 gene expression in peripheral blood mononuclear cells were evaluated. Single administration of 2 g or 4 g of CAP was safe and well-tolerated. The TT aglycones, asiatic acid and madecassic acid, were identified in plasma and urine, while the parent glycosides, asiaticoside and madecassoside, although abundant in CAP, were absent in plasma and had limited renal excretion. Similarly, mono- and di-CQAs showed delayed absorption and limited presence in plasma or urine, while the putative metabolites of these compounds showed detectable plasma pharmacokinetic profiles and urinary excretion. CAP elicited a temporal change in NRF2 gene expression, mirroring the TT aglycone's pharmacokinetic curve in a paradoxical dose-dependent manner. The oral bioavailability of active compounds or their metabolites, NRF2 target engagement, and the acute safety and tolerability of CAP support the validity of using CAP in future clinical studies.
Collapse
Affiliation(s)
- Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Melissa Bollen
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Jason David
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Alex B. Speers
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Mikah S. Brandes
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Armando Alcázar Magaña
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA;
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Christine McClure
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| | - Jan F. Stevens
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR 97331, USA;
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Claudia S. Maier
- Department of Chemistry, Oregon State University, Corvallis, OR 97331, USA; (A.A.M.); (C.S.M.)
- Linus Pauling Institute, Oregon State University, Corvallis, OR 97331, USA
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
- Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, OR 97239, USA
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR 97239, USA; (K.M.W.); (M.B.); (J.D.); (A.B.S.); (M.S.B.); (N.E.G.); (C.M.); (J.F.Q.)
| |
Collapse
|
20
|
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
|
21
|
Speers AB, García-Jaramillo M, Feryn A, Matthews DG, Lichtenberg T, Caruso M, Wright KM, Quinn JF, Stevens JF, Maier CS, Soumyanath A, Gray NE. Centella asiatica Alters Metabolic Pathways Associated With Alzheimer's Disease in the 5xFAD Mouse Model of ß-Amyloid Accumulation. Front Pharmacol 2021; 12:788312. [PMID: 34975484 PMCID: PMC8717922 DOI: 10.3389/fphar.2021.788312] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Accepted: 11/30/2021] [Indexed: 11/13/2022] Open
Abstract
Centella asiatica is an herb used in Ayurvedic and traditional Chinese medicine for its beneficial effects on brain health and cognition. Our group has previously shown that a water extract of Centella asiatica (CAW) elicits cognitive-enhancing effects in animal models of aging and Alzheimer's disease, including a dose-related effect of CAW on memory in the 5xFAD mouse model of ß-amyloid accumulation. Here, we endeavor to elucidate the mechanisms underlying the effects of CAW in the brain by conducting a metabolomic analysis of cortical tissue from 5xFAD mice treated with increasing concentrations of CAW. Tissue was collected from 8-month-old male and female 5xFAD mice and their wild-type littermates treated with CAW (0, 200, 500, or 1,000 mg/kg/d) dissolved in their drinking water for 5 weeks. High-performance liquid chromatography coupled to high-resolution mass spectrometry analysis was performed and relative levels of 120 annotated metabolites were assessed in the treatment groups. Metabolomic analysis revealed sex differences in the effect of the 5xFAD genotype on metabolite levels compared to wild-type mice, and variations in the metabolomic response to CAW depending on sex, genotype, and CAW dose. In at least three of the four treated groups (5xFAD or wild-type, male or female), CAW (500 mg/kg/d) significantly altered metabolic pathways related to purine metabolism, nicotinate and nicotinamide metabolism, and glycerophospholipid metabolism. The results are in line with some of our previous findings regarding specific mechanisms of action of CAW (e.g., improving mitochondrial function, reducing oxidative stress, and increasing synaptic density). Furthermore, these findings provide new information about additional, potential mechanisms for the cognitive-enhancing effect of CAW, including upregulation of nicotinamide adenine dinucleotide in the brain and modulation of brain-derived neurotrophic factor. These metabolic pathways have been implicated in the pathophysiology of Alzheimer's disease, highlighting the therapeutic potential of CAW in this neurodegenerative disease.
Collapse
Affiliation(s)
- Alex B. Speers
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Manuel García-Jaramillo
- Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, OR, United States
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Alicia Feryn
- OHSU-PSU School of Public Health, Oregon Health & Science University, Portland, OR, United States
| | - Donald G. Matthews
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Talia Lichtenberg
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Maya Caruso
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Kirsten M. Wright
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Joseph F. Quinn
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
- Parkinson’s Disease Research Education and Clinical Care Center, Veterans’ Administration Portland Health Care System, Portland, OR, United States
| | - Jan F. Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Pharmaceutical Sciences, Oregon State University, Corvallis, OR, United States
| | - Claudia S. Maier
- Linus Pauling Institute, Oregon State University, Corvallis, OR, United States
- Department of Chemistry, Oregon State University, Corvallis, OR, United States
| | - Amala Soumyanath
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| | - Nora E. Gray
- Department of Neurology, Oregon Health & Science University, Portland, OR, United States
| |
Collapse
|
22
|
Ferraris Q, Alcazar A, Qian MC. Profiling polar lipids in whey protein phospholipid concentrate by LC-HRMS/MS. Food Chem 2021; 374:131495. [PMID: 34776307 DOI: 10.1016/j.foodchem.2021.131495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/10/2021] [Accepted: 10/26/2021] [Indexed: 11/28/2022]
Abstract
Bioactive polar lipids in the milk fat globular membrane can be recovered and enriched during whey protein processing into a co-product called whey protein phospholipid concentrate (WPPC). A food-grade solvent successfully extracted polar lipids from powdered dairy products, and lipids can be fractionated under temperature-induced crystallization. This study investigates the specific lipid species present in ethanol extracted lipid residues from commercially available WPPC using a UPLC-Q-TOF-MS/MS lipidomics method. In general, sphingomyelins and phosphatidylcholines were retained in the polar lipid enriched fraction. Sphingomyelin was found to be a rich source of long chain fatty acids. Several glycosphingolipids, glucosyl-, galactosyl-, lactosyl-, and galabiosylceramide, were also detected in WPPC; these species were observed to crystallize away from other polar lipids during fractionation. Correlation analysis supported the claim that majority of polar lipids recovered in a total lipid extract using ethanol were retained in a polar lipid enriched residue after fractional crystallization.
Collapse
Affiliation(s)
- Quintin Ferraris
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97330, United States
| | - Armando Alcazar
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97330, United States; Department of Chemistry, Oregon State University, Corvallis, OR 97330, United States
| | - Michael C Qian
- Department of Food Science and Technology, Oregon State University, Corvallis, OR 97330, United States.
| |
Collapse
|
23
|
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: 103] [Impact Index Per Article: 25.8] [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
|
24
|
Keller RM, Beaver LM, Reardon PN, Prater MC, Truong L, Robinson MM, Tanguay RL, Stevens JF, Hord NG. Nitrate-induced improvements in exercise performance are coincident with exuberant changes in metabolic genes and the metabolome in zebrafish ( Danio rerio) skeletal muscle. J Appl Physiol (1985) 2021; 131:142-157. [PMID: 34043471 PMCID: PMC8325611 DOI: 10.1152/japplphysiol.00185.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 05/19/2021] [Accepted: 05/24/2021] [Indexed: 11/22/2022] Open
Abstract
Dietary nitrate supplementation improves exercise performance by reducing the oxygen cost of exercise and enhancing skeletal muscle function. However, the mechanisms underlying these effects are not well understood. The purpose of this study was to assess changes in skeletal muscle energy metabolism associated with exercise performance in a zebrafish model. Fish were exposed to sodium nitrate (60.7 mg/L, 303.5 mg/L, 606.9 mg/L), or control water, for 21 days and analyzed at intervals (5, 10, 20, 30, 40 cm/s) during a 2-h strenuous exercise test. We measured oxygen consumption during an exercise test and assessed muscle nitrate concentrations, gene expression, and the muscle metabolome before, during, and after exercise. Nitrate exposure reduced the oxygen cost of exercise and increased muscle nitrate concentrations at rest, which were reduced with increasing exercise duration. In skeletal muscle, nitrate treatment upregulated expression of genes central to nutrient sensing (mtor), redox signaling (nrf2a), and muscle differentiation (sox6). In rested muscle, nitrate treatment increased phosphocreatine (P = 0.002), creatine (P = 0.0005), ATP (P = 0.0008), ADP (P = 0.002), and AMP (P = 0.004) compared with rested-control muscle. Following the highest swimming speed, concentration of phosphocreatine (P = 8.0 × 10-5), creatine (P = 6.0 × 10-7), ATP (P = 2.0 × 10-6), ADP (P = 0.0002), and AMP (P = 0.004) decreased compared with rested nitrate muscle. Our data suggest nitrate exposure in zebrafish lowers the oxygen cost of exercise by changing the metabolic programming of muscle prior to exercise and increasing availability of energy-rich metabolites required for exercise.NEW & NOTEWORTHY We show that skeletal muscle nitrate concentration is higher with supplementation at rest and was lower in groups with increasing exercise duration in a zebrafish model. The higher availability of nitrate at rest is associated with upregulation of key nutrient-sensing genes and greater availability of energy-producing metabolites (i.e., ATP, phosphocreatine, glycolytic intermediates). Overall, nitrate supplementation may lower oxygen cost of exercise through improved fuel availability resulting from metabolic programming of muscle prior to exercise.
Collapse
Affiliation(s)
- Rosa M Keller
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Laura M Beaver
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
| | - Patrick N Reardon
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
- Nuclear Magnetic Resonance Facility, Oregon State University, Corvallis, Oregon
| | - Mary C Prater
- Department of Foods and Nutrition, College of Family and Consumer Sciences, University of Georgia, Athens, Georgia
| | - Lisa Truong
- Sinnhuber Aquatic Research Laboratory and the Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Matthew M Robinson
- School of Biological and Population Health Sciences, College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| | - Robyn L Tanguay
- Sinnhuber Aquatic Research Laboratory and the Department of Environmental and Molecular Toxicology, Oregon State University, Corvallis, Oregon
| | - Jan F Stevens
- Linus Pauling Institute, Oregon State University, Corvallis, Oregon
- College of Pharmacy, Oregon State University, Corvallis, Oregon
| | - Norman G Hord
- OU Health, Harold Hamm Diabetes Center, Department of Nutritional Sciences, College of Allied Health, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
25
|
Speers AB, Cabey KA, Soumyanath A, Wright KM. Effects of Withania somnifera (Ashwagandha) on Stress and the Stress- Related Neuropsychiatric Disorders Anxiety, Depression, and Insomnia. Curr Neuropharmacol 2021; 19:1468-1495. [PMID: 34254920 PMCID: PMC8762185 DOI: 10.2174/1570159x19666210712151556] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/19/2021] [Accepted: 06/28/2021] [Indexed: 11/22/2022] Open
Abstract
BACKGROUND Withania somnifera (WS), also known as Ashwagandha, is commonly used in Ayurveda and other traditional medicine systems. WS has seen an increase in worldwide usage due to its reputation as an adaptogen. This popularity has elicited increased scientific study of its biological effects, including a potential application for neuropsychiatric and neurodegenerative disorders. OBJECTIVE This review aims to provide a comprehensive summary of preclinical and clinical studies examining the neuropsychiatric effects of WS, specifically its application in stress, anxiety, depression, and insomnia. METHODS Reports of human trials and animal studies of WS were collected primarily from the PubMed, Scopus, and Google Scholar databases. RESULTS WS root and leaf extracts exhibited noteworthy anti-stress and anti-anxiety activity in animal and human studies. WS also improved symptoms of depression and insomnia, though fewer studies investigated these applications. WS may alleviate these conditions predominantly through modulation of the hypothalamic-pituitary-adrenal and sympathetic-adrenal-medullary axes, as well as through GABAergic and serotonergic pathways. While some studies link specific withanolide components to its neuropsychiatric benefits, there is evidence for the presence of additional, as yet unidentified, active compounds in WS. CONCLUSION While benefits were seen in the reviewed studies, significant variability in the WS extracts examined prevents a consensus on the optimum WS preparation or dosage for treating neuropsychiatric conditions. WS generally appears safe for human use; however, it will be important to investigate potential herb-drug interactions involving WS if used alongside pharmaceutical interventions. Further elucidation of active compounds of WS is also needed.
Collapse
Affiliation(s)
| | | | - Amala Soumyanath
- Address correspondence to these authors at the Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA; Tel/Fax: +1-503-494-6882, +1-503-494-7499; E-mails: ;
| | - Kirsten M. Wright
- Address correspondence to these authors at the Department of Neurology, Oregon Health and Science University, Portland, Oregon, USA; Tel/Fax: +1-503-494-6882, +1-503-494-7499; E-mails: ;
| |
Collapse
|
26
|
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: 17] [Impact Index Per Article: 4.3] [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
|
27
|
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: 32] [Impact Index Per Article: 6.4] [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
|
28
|
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: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Centella asiatica (CA) shows considerable promise for development as a botanical drug for cognitive decline. Its primary bioactive components include triterpene glycosides asiaticoside and madecassoside and their corresponding aglycones asiatic acid and madecassic acid. Exploration of the bioactivity of CA's caffeoylquinic acids is ongoing. In this study, an aqueous extract of CA (CAW-R61J) was evaluated for drug interaction potential through inhibition or induction of P450 enzymes, as required by the US Food and Drug Administration. CAW-R61J was assessed for induction potential of CYP1A2, CYP2B6, and CYP3A4 using transporter-certified cryopreserved human hepatocytes in sandwich culture. Gene expression of these target P450s was quantified, and enzyme activities were determined to confirm gene expression results. No induction was observed up to 16.7 µg/ml CAW-R61J (equivalent to 1.1 µM asiaticoside, 0.8 µM madecassoside, 0.09 µM asiatic acid, and 0.12 µM madecassic acid). Reversible and time-dependent inhibitory effects of CAW-R61J on CYP1A2, CYP2B6, CYP2C8, CYP2C9, CYP2C19, CYP2D6, and CYP3A4/5 were evaluated using human liver microsomes. CAW-R61J showed weak reversible inhibition of most of the P450 forms tested, with the strongest being CYP2C9 (IC50 of 330 µg/ml). CAW-R61J (≤1000 µg/ml) was not a time-dependent inhibitor of any of these P450 enzymes. In summary, CAW-R61J had no, or only a weak impact, on P450 induction and inhibition in vitro. The clinical relevance of these results will depend on the in vivo concentration of CAW-R61J components achieved in humans. Plasma triterpene concentrations measured in our recent clinical studies suggest minimal risk of P450-mediated drug interactions by these components. SIGNIFICANCE STATEMENT: A preparation of Centella asiatica is currently under clinical development for the prevention or treatment of cognitive decline. The US Food and Drug Administration required an evaluation of its potential for drug interactions mediated through drug-metabolizing enzymes. This in vitro study revealed minimal induction or inhibition of a range of P450 enzymes, including CYP3A4, by the C. asiatica extract, suggesting a low potential for drug interactions modulated by P450 metabolism.
Collapse
Affiliation(s)
- Kirsten M Wright
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Armando Alcazar Magana
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Ronald M Laethem
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Caroline L Moseley
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Troy T Banks
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Claudia S Maier
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Jan F Stevens
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Joseph F Quinn
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| | - Amala Soumyanath
- Department of Neurology, Oregon Health and Science University, Portland, Oregon (K.M.W., J.F.Q., A.S.); Departments of Chemistry (A.A.M., C.S.M.) and Pharmaceutical Sciences (J.F.S.) and Linus Pauling Institute (A.A.M., J.F.S.), Oregon State University, Corvallis, Oregon; BioIVT, Durham, North Carolina (R.M.L., C.L.M., T.T.B.); and Department of Neurology, Veterans Affairs Portland Health Care System Center, Portland, Oregon (J.F.Q.)
| |
Collapse
|