1
|
Chen HJ, Chen YF, Chen JF, Qian K, Zhu YY, Fang L, Zhang Y, Yang T, Wang GW, Huang PT. Kuanxiong Aerosol Attenuates Ischemic Stroke Injury via Modulation of the TRPV1 Channel. Chin J Integr Med 2025:10.1007/s11655-024-3669-9. [PMID: 39792345 DOI: 10.1007/s11655-024-3669-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVE To evaluate the therapeutic effects of Kuanxiong Aerosol (KXA) on ischemic stroke with reperfusion and elucidate the underlying pharmacological mechanisms. METHODS In vivo pharmacological effects on ischemic stroke with reperfusion was evaluated using the transient middle cerebral artery occlusion (t-MCAO) mice model. To evaluate short-term outcome, 30 mice were randomly divided into vehicle group (n=15) and KXA group (n=15). Mice in KXA and vehicle groups received 69 mg KXA and vehicle for 1 day, respectively. To evaluate long-term outcome, 35 mice were randomly divided into sham group (n=5), vehicle group (n=15), and KXA group (n=15). Mice in KXA and vehicle groups received 69 mg KXA and vehicle for 7 days, respectively. Pathological changes in the brain were observed by 2,3,5-triphenyltetrazolium chloride or Nissl stainings, and behavioral assessments, including the Modified Neurologic Severity Score, Bederson score, rotarod test, and adhesive removal test were conducted. The penetration ability of KXA and KX (KXA without propellants) through the blood-brain barrier was assessed both in vitro using a transwell model and in vivo. Furthermore, in vitro effects of KX (5, 10, and 20 µL/L) on oxygen and glucose deprivation/re-oxygenation (OGD/R)-induced injury, transient receptor potential vanilloid type 1 (TRPV1) modulation, calcium influx, and mitochondrial function were explored through Western blot, CCK-8 assay, JC-1 staining, calcium imaging, adenosine triphosphate (ATP) and antioxidant measurements. RESULTS In in vivo experiments, KXA reduced brain infarct volume and neuron loss in t-MCAO mice. Behavioral assessments showed marked improvement in the neurological deficit of t-MCAO mice with KXA treatment (P<0.05 or P<0.01). Additionally, in vitro findings indicated that KX ameliorated OGD/R-induced injury through TRPV1 channel modulation. KX increased cell viability in OGD/R-treated SH-SY5Y cells and prevented OGD/R-induced calcium overload by downregulating TRPV1 expression and constraining calcium influx through TRPV1 (P<0.05 or P<0.01). Furthermore, KXA maintained the membrane potential and function of mitochondria in OGD/R-treated SH-SY5Y cells. CONCLUSIONS KXA could attenuate ischemic stroke injury through TRPV1 channel modulation, indicating its potential as a promising therapeutic option for stroke in clinical practice.
Collapse
Affiliation(s)
- Hong-Jian Chen
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Ye-Feng Chen
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Ji-Fan Chen
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Kai Qian
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Yang-Yang Zhu
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Lei Fang
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Ying Zhang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Tao Yang
- Postdoctoral Research Center, Zhejiang SuKean Pharmaceutical Co., Ltd., Hangzhou, 311225, China
| | - Guo-Wei Wang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China
| | - Pin-Tong Huang
- Department of Ultrasound in Medicine, the Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, 310009, China.
| |
Collapse
|
2
|
Yan X, David SD, Du G, Li W, Liang D, Nie S, Ge M, Wang C, Qiao J, Li Y, Caiyin Q. Biological Properties of Sandalwood Oil and Microbial Synthesis of Its Major Sesquiterpenoids. Biomolecules 2024; 14:971. [PMID: 39199359 PMCID: PMC11352278 DOI: 10.3390/biom14080971] [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: 07/09/2024] [Revised: 07/26/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
Sandalwood essential oil is extracted from the heartwood part of mature sandalwood and is known for its pleasant fragrance and exceptional medicinal activities, including antimicrobial, antitumor, and anti-inflammatory properties. The (Z)-α-santalol and (Z)-β-santalol are the most vital ingredients contributing to sandalwood oil's bioactivities and unique woody odor characteristics. Metabolic engineering strategies have shown promise in transforming microorganisms such as yeast and bacteria into effective cell factories for enhancing the production of vital sesquiterpenes (santalene and santalol) found in sandalwood oil. This review aims to summarize sources of sandalwood oil, its components/ingredients, and its applications. It also highlights the biosynthesis of santalene and santalol and the various metabolic engineering strategies employed to reconstruct and enhance santalene and santalol biosynthesis pathways in heterologous hosts.
Collapse
Affiliation(s)
- Xiaoguang Yan
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Sichone Daniel David
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Guangzhao Du
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Weiguo Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Dongmei Liang
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Shengxin Nie
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Mingyue Ge
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
| | - Chen Wang
- Zhejiang Key Laboratory of Alternative Technologies for Fine Chemicals Process, Shaoxing University, Shaoxing 312000, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Zhejiang Institute, Tianjin University, Shaoxing 312300, China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Yanni Li
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, China
- Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, Tianjin 300072, China
| |
Collapse
|
3
|
Xiaoying M, Zhiming H, Tao Y, Jun X, Ying Z, Na G, Xun C, Guoli L, Hong W. Elucidating the molecular mechanisms underlying anti-inflammatory effects of Morchella esculenta in the arachidonic acid metabolic pathway by network pharmacology and molecular docking. Sci Rep 2023; 13:15881. [PMID: 37741847 PMCID: PMC10517965 DOI: 10.1038/s41598-023-42658-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 09/13/2023] [Indexed: 09/25/2023] Open
Abstract
Morchella esculenta is an edible fungus with a uniquely delicious flavor and remarkable benefits for health. Herein, the molecular mechanism underlying the anti-inflammatory effects of Morchella esculenta was elucidated using molecular docking and network pharmacology. NPASS, Super-pred, SEA, Swiss Target Prediction, GeneCards, DisGeNET, Omim database, and STRING platform were used to select anti-inflammatory targets and construct target protein interaction networks using the active ingredients of Morchella esculenta. The OmicShare cloud platform was used to analyze GO functions and KEGG pathways related to the target, and the AutoDock Vina software was used to perform molecular docking and molecular dynamics (MD) simulation on the main target. Based on Cytoscape's "Network Analysis", the degree was used to identify potential key targets, and different inflammatory transcriptome data sets were used to evaluate core targets showing clinical significance. The active ingredient of Morchella esculenta identified from the NPASS database was EOYA, which had 43 anti-inflammatory targets, including NR1I2, PTGS1, PTGS2, CYP4F2, CYP3A4, TLR4, MAPK1, PLA2G4A, and PTPN11, and was mainly implicated in arachidonic acid metabolism, vascular endothelial growth factor signal pathway, and sphingomyelin signal transduction pathway, indicating that the anti-inflammatory effects of EOYA were mainly related to these biological processes. The degree was used to select 9 potential effective targets, namely NR1I2, PTGS1, PTGS2, CYP4F2, CYP3A4, TLR4, MAPK1, PLA2G4A, and PTPN11, among which NR1I2, PTGS1, PTGS2, PLA2G4A, MAPK1, CYP3A4, and TLR4 showed clinical significance. Molecular docking results showed that (E)-Octadec-11-En-9-Ynoic Acid (EOYA) could spontaneously bind to the 9 core targets, and the binding fractions of NR1I2, PTGS1, PTGS2, CYP4F2, and CYP3A4 were the highest. The MD simulation results showed that EYOA did indeed bind well NR1I2 to PTGS2, and the complex has high stability. Morchella esculenta can regulate the activity of prostaglandin endoperoxide synthetase, and affect the biosynthesis of prostaglandins, thereby impacting the metabolic pathway of arachidonic acid.
Collapse
Affiliation(s)
- Ma Xiaoying
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Huo Zhiming
- Information Center, Guidaojiaotong Polytechnic Institute, Shenyang, 110161, China
| | - Yang Tao
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Xiao Jun
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Zhao Ying
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Gong Na
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Chen Xun
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Liu Guoli
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China
| | - Wang Hong
- The Institute of Edible Fungi, Liaoning Academy of Agricultural Sciences, Shenyang, 110161, China.
| |
Collapse
|
4
|
Gao X, Zhang H, Li K, Shi Y, Guo X, Wang L, Li D. Sandalwood seed oil improves insulin sensitivity in high-fat/high-sucrose diet-fed rats associated with altered intestinal microbiota and its metabolites. Food Funct 2021; 12:9739-9749. [PMID: 34664591 DOI: 10.1039/d1fo02239c] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Sandalwood seed oil (SSO), rich in ximenynic acid, is extracted from the seed kernels of Santalum spicatum. The current work aimed to clarify the potential mechanisms of SSO in preventing insulin resistance (IR) by investigating the intestinal microbiota and its metabolites. Fifty male Sprague-Dawley rats were randomly divided into a standard chow group (N), and four high-fat/high-sucrose (HFHS) diet-fed groups plus 7% of SSO, fish oil (FO), linseed oil (LO) or sunflower oil (SO), respectively. After 12 weeks, the feces were collected and subsequently the rats were sacrificed for collecting blood and tissues. The results indicated that the SSO, FO and LO groups had a lower ratio of Firmicutes to Bacteroidetes (F/B) and lower levels of Actinobacteria phylum in their feces compared to the SO group. HOMA-IR was positively correlated with F/B (r = 0.63) and Actinobacteria (r = 0.64). At the genus level, beneficial bacteria, including Oscillospira, Clostridium, Turicibacter, Ruminococcus and Coprococcus, were more abundant, while destructive bacteria, such as Collinsella, were less abundant in the SSO group than in the SO group. The concentrations of fecal short-chain fatty acids (SCFAs) were higher, and the serum LPS and trimethylamine N-oxide (TMAO) were lower in the SSO, FO and LO groups than the SO group. In addition, SCFAs were negatively (r: -0.45 to -0.82), and LPS (r: 0.12 to 0.42) and TMAO (r: 0.32 to 0.49) were positively correlated with HOMA-IR and serum IL-1β, IL-6 and TNF-α. In summary, the prevention effect of SSO on HFHS induced IR was associated with altered intestinal microbiota composition and the production of microbial metabolites.
Collapse
Affiliation(s)
- Xiang Gao
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China. .,College of Life Sciences, Qingdao University, Qingdao, China
| | - Huijun Zhang
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| | - Kelei Li
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| | - Yan Shi
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| | - Xiaofei Guo
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| | - Ling Wang
- College of Food Science and Technology, Huazhong Agricultural University, Wuhan, China
| | - Duo Li
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| |
Collapse
|
5
|
Majewski G, Craw J, Falla T. Accelerated Barrier Repair in Human Skin Explants Induced with a Plant-Derived PPAR-α Activating Complex via Cooperative Interactions. Clin Cosmet Investig Dermatol 2021; 14:1271-1293. [PMID: 34566418 PMCID: PMC8458040 DOI: 10.2147/ccid.s325967] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2021] [Accepted: 08/28/2021] [Indexed: 12/27/2022]
Abstract
Background Peroxisome proliferator-activated receptors (PPARs) govern epidermal lipid synthesis and metabolism. In skin, PPAR activation has been shown to regulate genes responsible for permeability barrier homeostasis, epidermal differentiation, lipid biosynthesis, and inflammation. Objective Given the known dermatologic benefits of PPARs, we set out to discover a naturally derived, multi-molecule complex that would be superior to the more commonly formulated conjugated linoleic acids (CLAs). We hypothesized that a complex may be capable of modulating PPAR-α by cooperative or multi-ligand binding interactions to accelerate skin barrier repair. Methods To achieve this, we assembled a novel PPAR-α agonist complex, referred to as RFV3, from a combination of small molecules routinely used in Ayurvedic medicine and accepted in cosmetic and topical over-the-counter dermatologic products. We tested RFV3’s potential as a PPAR-α agonist by evaluating its transcriptional response, ligand binding affinity to PPAR-α, gene expression profiles and barrier repair properties in human skin explant models. Results We assembled RFV3 by solubilizing two standardized plant extracts in a suitable solvent and induced a significant transcriptional response in PPAR-α luciferase reporter assay. Furthermore, transcriptome profiling of RFV3-treated epidermal substitutes revealed expressed genes consistent with known targets of PPAR-α, including those involved in epidermal barrier repair. In addition, in silico modeling demonstrated differential co-binding affinities of RFV3 to PPAR-α compared with those of the endogenous ligands (CLAs) and a synthetic PPAR-α agonist. Lastly, delipidated skin explant models confirmed accelerated barrier repair activity with significant increases in ceramides, filaggrin and transglutaminase-1 after treatment. Conclusion These findings suggest that the RFV3 complex successfully mimics a PPAR-α agonist and induces synthesis of skin barrier lipids and proteins consistent with known PPAR pathways.
Collapse
Affiliation(s)
- George Majewski
- Rodan & Fields, San Francisco, CA, 94105, USA.,Present Affiliation: Contrast Product Development, Walnut, CA, 91789, USA
| | - John Craw
- Rodan & Fields, San Francisco, CA, 94105, USA
| | | |
Collapse
|
6
|
Zhang H, Gao X, Li K, Liu Y, Hettiarachichi DS, Sunderland B, Li D. Sandalwood seed oil ameliorates hepatic insulin resistance by regulating the JNK/NF-κB inflammatory and PI3K/AKT insulin signaling pathways. Food Funct 2021; 12:2312-2322. [PMID: 33617622 DOI: 10.1039/d0fo03051a] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Sandalwood (santalum spicatum) seed oil (SSO) is rich in ximenynic acid. The aim of the present study was to investigate the effect of SSO on high-fat/high-sucrose diet (HFHSD) induced insulin resistance (IR) in comparison with fish oil (FO), sunflower oil (SO) and linseed oil (LO). Fifty male Sprague-Dawley rats were randomly divided into five dietary groups: standard chow diet (controls), HFHSD plus 7% SSO, HFHSD plus 7% FO, HFHSD plus 7% SO and HFHSD plus 7% LO. After 12 weeks of feeding, the rats were sacrificed, and the serum parameters, hepatic lipids and underlying molecular mechanisms were studied. SSO, FO or LO significantly prevented glucose intolerance, hyperglycaemia, obesity, and hepatic lipid accumulation, and decreased the homeostasis model assessment of IR (HOMA-IR) and the serum levels of pro-inflammatory factors (IL-6, IL-1β and TNF-α) compared with SO. In addition, SSO activated the PI3K/AKT insulin signaling pathway and down-regulated the JNK/NF-κB inflammatory signaling pathway in the liver. In summary, our results proved that SSO exerted an ameliorative effect on IR by regulating the hepatic inflammation related blockage of the insulin signaling pathway in the rats.
Collapse
Affiliation(s)
- Huijun Zhang
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| | - Xiang Gao
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China. and College of Life Sciences, Qingdao University, Qingdao, China
| | - Kelei Li
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China.
| | - Yandi Liu
- School of Pharmacy, Curtin University, Perth, Australia
| | | | | | - Duo Li
- Institute of Nutrition & Health, College of Public Health, Qingdao University, Qingdao, China. and Department of Food Science and Nutrition, Zhejiang University, Hangzhou, China
| |
Collapse
|
7
|
Cai F, Liu Y, Hettiarachichi D, Wang F, Li J, Sunderland B, Li D. Ximenynic Acid Regulation of n-3 PUFA Content in Liver and Brain. Lifestyle Genom 2020; 13:64-73. [DOI: 10.1159/000502773] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 08/12/2019] [Indexed: 11/19/2022] Open
|
8
|
Yuan H, Gong J, Tang K, Huang J, Xiao G, Lv J. Milk oligopeptide inhibition of (α)-tocopherol fortified linoleic acid oxidation. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2019. [DOI: 10.1080/10942912.2019.1657888] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Affiliation(s)
- Haina Yuan
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Zhejiang Provincial Key Lab for Chem&Bio Processing Technology of Farm Produces, Hangzhou, ZJ, China
| | - Jinyan Gong
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Zhejiang Provincial Key Lab for Chem&Bio Processing Technology of Farm Produces, Hangzhou, ZJ, China
| | - Kun Tang
- Department of Endocrinology, The Affiliated Hospital of Guizhou Medical University, Guiyang, GZ, China
| | - Jinge Huang
- Department of Biological Engineering, Utah State University, Logan, UT, USA
| | - Gongnian Xiao
- School of Biological and Chemical Engineering, Zhejiang University of Science and Technology, Zhejiang Provincial Collaborative Innovation Center of Agricultural Biological Resources Biochemical Manufacturing, Zhejiang Provincial Key Lab for Chem&Bio Processing Technology of Farm Produces, Hangzhou, ZJ, China
| | - Jianmin Lv
- Laboratory Animal Research Center, Zhejiang Chinese Medical University, Hangzhou, ZJ, China
| |
Collapse
|
9
|
Orchard A, van Vuuren SF, Viljoen AM, Kamatou G. The in vitro antimicrobial evaluation of commercially essential oils and their combinations against acne. Int J Cosmet Sci 2018; 40:226-243. [PMID: 29574906 DOI: 10.1111/ics.12456] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2017] [Accepted: 02/28/2018] [Indexed: 01/22/2023]
Abstract
OBJECTIVE The study investigated the efficacy of commercial essential oil combinations against the two pathogens responsible for acne with the aim to identify synergy and favourable oils to possibly use in a blend. MATERIALS AND METHODS Antimicrobial activity was assessed using the minimum inhibitory concentration (MIC) assay against Staphylococcus epidermidis (ATCC 2223) and Propionibacterium acnes (ATCC 11827), and the fractional inhibitory concentration index (ΣFIC) was calculated. Combinations displaying synergistic interactions were further investigated at varied ratios and the results plotted on isobolograms. RESULTS From the 408 combinations investigated, 167 combinations were identified as displaying noteworthy antimicrobial activity (MIC value ≤1.00 mg mL-1 ). Thirteen synergistic interactions were observed against S. epidermidis, and three synergistic combinations were observed against P. acnes. It was found that not one of the synergistic interactions identified were based on the combinations recommended in the layman's aroma-therapeutic literature. Synergy was evident rather from leads based on antimicrobial activity from previous studies, thus emphasizing the importance of scientific validation. Leptospermum scoparium J.R.Forst. and G.Forst (manuka) was the essential oil mostly involved in synergistic interactions (four) against S. epidermidis. Cananga odorata (Lam.) Hook.f. and Thomson (ylang ylang) essential oil was also frequently involved in synergy where synergistic interactions could be observed against both pathogens. The combination with the lowest MIC value against both acne pathogens was Vetiveria zizanioides Stapf (vetiver) with Cinnamomum verum J.Presl (cinnamon bark) (MIC values 0.19-0.25 mg mL-1 ). Pogostemon patchouli Benth. (patchouli), V. zizanioides, C. verum and Santalum spp. (sandalwood) could be identified as the oils that contributed the most noteworthy antimicrobial activity towards the combinations. The different chemotypes of the essential oils used in the combinations predominantly resulted in similar antimicrobial activity. CONCLUSIONS The investigated essential oil combinations resulted in at least 50% of the combinations displaying noteworthy antimicrobial activity. Most of the synergistic interactions do not necessarily correspond to the recommended layman's aroma-therapeutic literature, which highlights a need for scientific validation of essential oil antimicrobial activity. No antagonism was observed.
Collapse
Affiliation(s)
- A Orchard
- Department of Pharmacy and Pharmacology, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| | - S F van Vuuren
- Department of Pharmacy and Pharmacology, University of the Witwatersrand, 7 York Road, Parktown, 2193, Johannesburg, South Africa
| | - A M Viljoen
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
- Department of Pharmaceutical Sciences, SAMRC Herbal Drugs Research Unit, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| | - G Kamatou
- Department of Pharmaceutical Sciences, Faculty of Science, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
- Department of Pharmaceutical Sciences, SAMRC Herbal Drugs Research Unit, Tshwane University of Technology, Private Bag X680, Pretoria, 0001, South Africa
| |
Collapse
|
10
|
Bommareddy A, Brozena S, Steigerwalt J, Landis T, Hughes S, Mabry E, Knopp A, VanWert AL, Dwivedi C. Medicinal properties of alpha-santalol, a naturally occurring constituent of sandalwood oil: review. Nat Prod Res 2017; 33:527-543. [PMID: 29130352 DOI: 10.1080/14786419.2017.1399387] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Alpha-santalol is a naturally occurring sesquiterpene that is derived from sandalwood oil. Its wide range of health benefits have been attributed to the modulation of various signalling pathways involved in the development of a particular disease. For example, the antitumour and cancer preventive properties of alpha-santalol have been shown to involve cell death induction through apoptosis and cell cycle arrest in various cancer models. A marked decrease in inflammatory markers have also been shown with alpha-santalol administration in skin tissue models. The current review is aimed at bringing the most recent advances of alpha-santalol against various disease-specific models and highlighting its associated mechanistic details.
Collapse
Affiliation(s)
- Ajay Bommareddy
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Sarah Brozena
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - James Steigerwalt
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Terra Landis
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Sarah Hughes
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Erica Mabry
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Aaron Knopp
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Adam L VanWert
- a Department of Pharmaceutical Sciences, Nesbitt School of Pharmacy , Wilkes University , Wilkes-Barre , PA , USA
| | - Chandradhar Dwivedi
- b Department of Pharmaceutical Sciences, College of Pharmacy , South Dakota State University , Brookings , South Dakota
| |
Collapse
|
11
|
Anjum S, Gupta A, Sharma D, Dalal P, Gupta B. Skin compatibility and antimicrobial studies on biofunctionalized polypropylene fabric. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2016; 69:1043-50. [DOI: 10.1016/j.msec.2016.08.009] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 07/22/2016] [Accepted: 08/03/2016] [Indexed: 12/18/2022]
|
12
|
Cai F, Li J, Liu Y, Zhang Z, Hettiarachchi DS, Li D. Effect of ximenynic acid on cell cycle arrest and apoptosis and COX-1 in HepG2 cells. Mol Med Rep 2016; 14:5667-5676. [PMID: 27840952 DOI: 10.3892/mmr.2016.5920] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 07/22/2016] [Indexed: 11/06/2022] Open
Abstract
Ximenynic acid is a conjugated enyne fatty acid, which is currently of interest due to its anti-inflammatory activity. Due to the association between inflammation and cancer, the present study was designed to investigate the anti‑cancer activity of ximenynic acid in the HepG2 human hepatoma cell line and the underlying mechanisms. The current study demonstrated the anti‑proliferation and pro‑apoptosis activities of ximenynic acid by cell viability assay and flow cytometry analysis. The expression of anti‑apoptosis protein silent information regulator T1 (SIRT1) was significantly suppressed by ximenynic acid. Furthermore, ximenynic acid blocked G1/S phase transition by inhibiting the protein expression of the cell cycle‑associated protein general control of amino acid synthesis yeast homolog like 2 (GCN5L2), and the mRNA expression of cyclin D3 and cyclin E1. Furthermore, ximenynic acid suppressed the expression of angiogenesis‑associated genes, including vascular endothelial growth factor (VEGF)‑B and VEGF‑C. Finally, ximenynic acid significantly inhibited the expression of cyclooxygenase‑1 (COX‑1) mRNA and protein, however COX‑2 expression was not reduced. The results of the present study suggested that ximenynic acid may inhibit growth of HepG2 cells by selective inhibition of COX‑1 expression, which leads to cell cycle arrest, and alters the apoptosis pathway and expression of angiogenic factors. The current study aimed to investigate whether ximenynic acid might be developed as novel anticancer agent.
Collapse
Affiliation(s)
- Fang Cai
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| | - Jianying Li
- Institute of Developmental and Regenerative Biology, Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - Yandi Liu
- School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth 02042G, Australia
| | - Zunyi Zhang
- Institute of Developmental and Regenerative Biology, Hangzhou Normal University, Hangzhou, Zhejiang 310000, P.R. China
| | - D S Hettiarachchi
- School of Pharmacy, Curtin Health Innovation Research Institute, Curtin University, Perth 02042G, Australia
| | - Duo Li
- Department of Food Science and Nutrition, Zhejiang University, Hangzhou, Zhejiang 310058, P.R. China
| |
Collapse
|
13
|
Shinagawa FB, Santana FCD, Mancini-Filho J. Efeito do óleo de semente de uva prensado a frio nos marcadores bioquímicos e perfil inflamatório de ratos. REV NUTR 2015. [DOI: 10.1590/1415-52732015000100006] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Objetivo: O objetivo deste trabalho foi avaliar o efeito do consumo crônico do óleo de semente de uva, obtido do mercado brasileiro, nos marcadores bioquímicos e inflamatórios de ratos saudáveis. Métodos: Ratos Wistar, recém-desmamados e saudáveis, receberam por 65 dias óleo de semente de uva e soja em duas concentrações (3 e 6 mL/kg de peso corporal). Os parâmetros avaliados foram a ingestão alimentar, peso corporal e dos tecidos hepático, cerebral e adiposo retroperitonial; neste último, foi ainda realizado o perfil de ácidos graxos. A análise dos parâmetros bioquímicos, peroxidação lipídica e perfil inflamatório através da quantificação das citocinas TNF-α, IL-10 e IL-6 foi realizada no soro. Resultados: O óleo de semente de uva, independentemente da dose administrada, promoveu maior acúmulo de gordura no tecido hepático e aumento nos níveis de peroxidação lipídica do soro. Verificou-se que, quando consumido na maior dose, houve maior incorporação do ácido graxo linoleico no tecido adiposo retroperitonial. Modificações nos parâmetros bioquímicos e inflamatórios séricos não foram observadas. Conclusão: O consumo de óleo de semente de uva não provocou alterações metabólicas significantes em nenhuma das doses administradas ainda que se tenha observado uma elevação nos níveis de peroxidação lipídica sérica.
Collapse
|