1
|
Sathiyamoorthi E, Lee J, Ramesh MD, M R, Sandhanasamy D, Nguyen ND, Shanmuganathan R. Biodiesel production from eggshells derived bio-nano CaO catalyst-Microemulsion fuel blends for up-gradation of biodiesel. ENVIRONMENTAL RESEARCH 2024; 260:119626. [PMID: 39019143 DOI: 10.1016/j.envres.2024.119626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 07/12/2024] [Accepted: 07/14/2024] [Indexed: 07/19/2024]
Abstract
The utilization of bio-oil derived from biomass presents a promising alternative to fossil fuels, though it faces challenges when directly applied in diesel engines. Microemulsification has emerged as a viable strategy to enhance bio-oil properties, facilitating its use in hybrid fuels. This study explores the microemulsification of Jatropha bio-oil with ethanol, aided by a surfactant, to formulate a hybrid liquid fuel. Additionally, a bio-nano CaO heterogeneous catalyst synthesized from eggshells is employed to catalyse the production of Jatropha biodiesel from the microemulsified fuel using microwave irradiation. The catalyst is characterized through UV-Vis, XRD, and SEM analysis. The investigation reveals a significant reduction in CO, CO2, and NOX emissions with the utilization of microemulsion-based biodiesel blends. Various blends of conventional diesel, Jatropha biodiesel, and ethanol are prepared with different ethanol concentrations (5, 10, and 20 wt%). Engine performance parameters, including fuel consumption, NOX emission, and brake specific fuel consumption, are analyzed. Results indicate that the conventional diesel/Jatropha biodiesel/ethanol (10 wt%) blend exhibits superior performance compared to conventional diesel, Jatropha biodiesel, and other blends. The fuel consumption of the conventional diesel/Jatropha biodiesel/ethanol (10 wt%) blend is measured at 554.6 g/h, surpassing that of conventional diesel and other biodiesel blends. The presence of water (0.14 %) in the blend reduces the heating value, consequently increasing the energy requirement. CO and CO2 emissions for the conventional diesel/Jatropha biodiesel/ethanol (10 wt%) blend are notably lower compared to conventional C-18 hydrocarbons and various biodiesel blends. These findings accentuate the efficacy of the microemulsion process in enhancing fuel characteristics and reducing emissions. Further investigations could explore optimizing the emulsifying agents and their impact on engine performance and emission characteristics, contributing to the advancement of sustainable fuel technologies.
Collapse
Affiliation(s)
- Ezhaveni Sathiyamoorthi
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - M D Ramesh
- Instituto de Alta Investigación, Universidad de Tarapacá, Arica, 1000000, Chile
| | - Rithika M
- Center for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, India
| | - Devanesan Sandhanasamy
- Department of Physics and Astronomy, College of Science, King Saud University, P. O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - N D Nguyen
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam; School of Engineering & Technology, Duy Tan University, Da Nang, Vietnam
| | - Rajasree Shanmuganathan
- Institute of Research and Development, Duy Tan University, Da Nang, Vietnam; School of Engineering & Technology, Duy Tan University, Da Nang, Vietnam.
| |
Collapse
|
2
|
Benito-Vázquez I, Garrido-Romero M, Hontoria-Caballo G, García-García C, Díez-Municio M, Moreno FJ. Carob ( Ceratonia siliqua) Flour as Source of Bioactive Compounds: Production, Characterization and Nutraceutical Value. Foods 2024; 13:3024. [PMID: 39410059 PMCID: PMC11475722 DOI: 10.3390/foods13193024] [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: 08/22/2024] [Revised: 09/12/2024] [Accepted: 09/18/2024] [Indexed: 10/20/2024] Open
Abstract
Carob (Ceratonia siliqua) seeds are rich in diverse bioactive compounds, including galactomannan, β-sitosterol, unsaturated fatty acids and proteins with bioactive peptides in their sequence. This study delineates the compositional characterization of six carob seed flour types derived from different production processes, providing valuable insights for designing tailored nutraceutical products based on desired bioactive compound profiles. Our analysis indicated that a higher purity of galactomannan resulted in a greater mannose/galactose ratio, which increased the linearity of the galactomannan polymer and could enhance interchain interaction, thereby increasing aggregation capacity. A higher viscosity could potentially increase the capacity of galactomannan to create satiety and lower cholesterol levels. Among the different tested flours, those whose main compound was the endosperm were optimal for containing high galactomannan content, whereas those derived from the germ were ideal for having high concentrations of fatty acids (i.e., oleic and linoleic acids) and β-sitosterol. The presence of these lipids in carob flours could offer cardiovascular and metabolic health benefits, contributing synergistically. Additionally, flours that contain the germ have beneficial peptides included in proteins like glycinin and conglutin with potential anticholesterolemic and antidiabetic properties. This work provides different methods for obtaining carob flours rich in bioactive compounds, offering the nutraceutical industry a framework to select the best option for industrial-scale production.
Collapse
Affiliation(s)
- Iván Benito-Vázquez
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (M.G.-R.)
- Pharmactive Biotech Products SLU, Faraday 7, 28049 Madrid, Spain; (G.H.-C.); (M.D.-M.)
| | - Manuel Garrido-Romero
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (M.G.-R.)
- Pharmactive Biotech Products SLU, Faraday 7, 28049 Madrid, Spain; (G.H.-C.); (M.D.-M.)
| | - Gema Hontoria-Caballo
- Pharmactive Biotech Products SLU, Faraday 7, 28049 Madrid, Spain; (G.H.-C.); (M.D.-M.)
| | - Carlos García-García
- Centro de Biología Molecular Severo Ochoa, CBM (CSIC-UAM), Nicolás Cabrera, 1, 28049 Madrid, Spain;
| | - Marina Díez-Municio
- Pharmactive Biotech Products SLU, Faraday 7, 28049 Madrid, Spain; (G.H.-C.); (M.D.-M.)
| | - F. Javier Moreno
- Instituto de Investigación en Ciencias de la Alimentación, CIAL (CSIC-UAM), Nicolás Cabrera 9, 28049 Madrid, Spain; (I.B.-V.); (M.G.-R.)
| |
Collapse
|
3
|
González-Montoya M, Vargas-Vargas MA, Torres-Isidro O, García-Berumen CI, Cuiniche-Méndez MG, Saavedra-Molina A, Ontiveros-Rodríguez JC, García-Gutiérrez HA, Calderón-Cortés E, Cortés-Rojo C. Evaluation of Unsaponifiable Fraction of Avocado Oil on Liver and Kidney Mitochondrial Function in Rats Fed a High-Fat and High-Carbohydrate Diet. Metabolites 2024; 14:431. [PMID: 39195527 PMCID: PMC11487379 DOI: 10.3390/metabo14080431] [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: 07/03/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/29/2024] Open
Abstract
High-fat and high-carbohydrate (HF-HC) diets induce metabolic syndrome via mitochondrial dysfunction and oxidative stress. We have previously shown that this may be prevented by avocado oil, a source of bioactive molecules with antioxidant properties. However, it is unknown if these effects are mediated by the unsaponifiable fraction of avocado oil (UFAO). Thus, we tested if this fraction improves glucose metabolism, bioenergetics and oxidative stress in mitochondria from the kidney and liver of rats fed an HF-HC diet. We found that 12 weeks of an HF-HC diet impaired glucose utilization and increased insulin resistance, which was prevented by UFAO administration. The HF-HC diet decreased respiration, membrane potential and electron transport chain (ETC) function in liver and kidney mitochondria. These mitochondrial dysfunctions were prevented by UFAO intake. Unexpectedly, UFAO increased ROS levels in the mitochondria of control animals and did not decrease them in rats with an HF-HC diet; however, UFAO protects liver and kidney mitochondria from iron-induced oxidative stress. These findings suggest that impairments in glucose metabolism and mitochondrial function by an HF-HC diet may be prevented by UFAO, without decreasing ROS generation but protecting mitochondria from oxidative damage.
Collapse
Grants
- 7973911 Programa Proyectos de Investigación financiados 2024, Coordinación de Investigación Científica, Universidad Michoacana de San Nicolás de Hidalgo, México.
- ICTI-PICIR23-063 Instituto de Ciencia, Tecnología e Innovación - Gobierno del Estado de Michoacán, México.
- ICTI PICIR23-028 Instituto de Ciencia, Tecnología e Innovación - Gobierno del Estado de Michoacán, México.
- 472544 Estancias Posdoctorales por Mexico 2022, Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT)
- 589763 Estancias Posdoctorales por Mexico 2023, Consejo Nacional de Humanidades, Ciencia y Tecnología (CONAHCYT)
Collapse
Affiliation(s)
- Marcela González-Montoya
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - Manuel Alejandro Vargas-Vargas
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - Olin Torres-Isidro
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - Claudia Isabel García-Berumen
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - María Guadalupe Cuiniche-Méndez
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - Alfredo Saavedra-Molina
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - Julio Cesar Ontiveros-Rodríguez
- Consejo Nacional de Humanidades, Ciencias y Tecnologías-Instituto de Investigaciones Químico Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico;
| | - Hugo A. García-Gutiérrez
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| | - Elizabeth Calderón-Cortés
- Facultad de Enfermería, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58020, MC, Mexico;
| | - Christian Cortés-Rojo
- Instituto de Investigaciones Químico-Biológicas, Universidad Michoacana de San Nicolás de Hidalgo, Morelia 58030, MC, Mexico; (M.A.V.-V.); (O.T.-I.); (C.I.G.-B.); (M.G.C.-M.); (A.S.-M.); (H.A.G.-G.)
| |
Collapse
|
4
|
Zhang P, Liu N, Xue M, Zhang M, Xiao Z, Xu C, Fan Y, Qiu J, Zhang Q, Zhou Y. β-Sitosterol Reduces the Content of Triglyceride and Cholesterol in a High-Fat Diet-Induced Non-Alcoholic Fatty Liver Disease Zebrafish ( Danio rerio) Model. Animals (Basel) 2024; 14:1289. [PMID: 38731293 PMCID: PMC11083524 DOI: 10.3390/ani14091289] [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: 02/25/2024] [Revised: 04/01/2024] [Accepted: 04/20/2024] [Indexed: 05/13/2024] Open
Abstract
OBJECTIVE Non-alcoholic fatty liver disease (NAFLD) is strongly associated with hyperlipidemia, which is closely related to high levels of sugar and fat. β-sitosterol is a natural product with significant hypolipidemic and cholesterol-lowering effects. However, the underlying mechanism of its action on aquatic products is not completely understood. METHODS A high-fat diet (HFD)-induced NAFLD zebrafish model was successfully established, and the anti-hyperlipidemic effect and potential mechanism of β-sitosterol were studied using oil red O staining, filipin staining, and lipid metabolomics. RESULTS β-sitosterol significantly reduced the accumulation of triglyceride, glucose, and cholesterol in the zebrafish model. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis showed that differential lipid molecules in β-sitosterol mainly regulated the lipid metabolism and signal transduction function of the zebrafish model. β-sitosterol mainly affected steroid biosynthesis and steroid hormone biosynthesis in the zebrafish model. Compared with the HFD group, the addition of 500 mg/100 g of β-sitosterol significantly inhibited the expression of Ppar-γ and Rxr-α in the zebrafish model by at least 50% and 25%, respectively. CONCLUSIONS β-sitosterol can reduce lipid accumulation in the zebrafish model of NAFLD by regulating lipid metabolism and signal transduction and inhibiting adipogenesis and lipid storage.
Collapse
Affiliation(s)
- Peng Zhang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China;
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Naicheng Liu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China;
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Mingyang Xue
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
| | - Mengjie Zhang
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China;
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Zidong Xiao
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
| | - Chen Xu
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
| | - Yuding Fan
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
| | - Junqiang Qiu
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China;
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Qinghua Zhang
- Key Laboratory of Exploration and Utilization of Aquatic Genetic Resources, Ministry of Education, Shanghai Ocean University, Shanghai 201306, China;
- National Pathogen Collection Center for Aquatic Animals, Shanghai Ocean University, Shanghai 201306, China
| | - Yong Zhou
- Yangtze River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Wuhan 430223, China; (P.Z.); (N.L.); (M.X.); (M.Z.); (Z.X.); (C.X.); (Y.F.)
| |
Collapse
|
5
|
Wang L, Liu Y, Shen G, He G, Qiu S, Li B. Mechanisms of Si-Wu Decoction in the treatment of ulcerative colitis revealed by network pharmacology and experimental verification. JOURNAL OF ETHNOPHARMACOLOGY 2023; 317:116847. [PMID: 37356743 DOI: 10.1016/j.jep.2023.116847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/19/2023] [Accepted: 06/23/2023] [Indexed: 06/27/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Si-Wu Decoction (SWD) is a traditional Chinese medicine decoction. SWD is commonly used to treat blood deficiency syndrome. It is also used to treat some ulcerative colitis (UC) patients now, but the mechanism of action remains unclear. AIM OF THE STUDY This study explored the efficacy and mechanism of action of SWD in treating UC based on network pharmacology and related experimental validation. MATERIALS AND METHODS Several databases were used to screen SWD for major active ingredients, targets of the ingredients, and UC disease genes. Cytoscape 3.8.2 software was used for topological analysis to construct the drug-compound-disease gene-target relationship network. The String database platform was used to construct the target protein interaction network. The DAVID (Database for Annotation, Visualization and Integrated Discovery) database was used to perform the GO (Gene Ontology) and KEGG (Kyoto Encyclopedia of Genes and Genomes) enrichment analysis for the key targets. DSS (Dextran Sulfate Sodium)-induced UC mouse model was used to evaluate the in-vivo activity of SWD. Western Blot analysis and quantitative polymerase chain reaction were performed to verify the targets in the related pathways. RESULTS Network pharmacology revealed that the SWD targeted pathway network involved 12 core targets and 15 major pathways. SWD may play a part by targeting key targets such as nuclear factor-kappaB (NF-κB), Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) pathway, and several mitogenic pathways. We showed that SWD largely restored the colorectal structure in UC model mice. Compared to the model group, the SWD group showed reduced infiltration of inflammatory cells. SWD significantly decreased the mRNA levels of IL-6 (Interleukin-6), TNF-α (Tumor necrosis factor-alpha), IL-1b (Interleukin-1beta) and other pro-inflammatory factors. Western Blot results showed that SWD concentration-dependently inhibited STAT3 and NF-κB activation in DSS-treated colon tissue. CONCLUSION Our findings suggest that SWD treats UC by inhibiting STAT3 and NF-κB signaling pathways, reducing the expression of inflammatory cytokines, and improving epithelial repair in experimental colitis, thus shedding light on the mechanisms by which SWD exerts its effects on UC.
Collapse
Affiliation(s)
- Lin Wang
- Institute of Traditional Chinese Medicine and Stem Cell Research, College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Ya Liu
- Institute of Traditional Chinese Medicine and Stem Cell Research, College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Guiyan Shen
- Institute of Traditional Chinese Medicine and Stem Cell Research, College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Guanlin He
- Institute of Traditional Chinese Medicine and Stem Cell Research, College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, China.
| | - Shoutao Qiu
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Baojie Li
- Bio-X Institutes, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
6
|
Abo-Zaid OA, Moawed FS, Ismail ES, Ahmed ESA. β-Sitosterol mitigates hepatocyte apoptosis by inhibiting endoplasmic reticulum stress in thioacetamide-induced hepatic injury in γ-irradiated rats. Food Chem Toxicol 2023; 172:113602. [PMID: 36610474 DOI: 10.1016/j.fct.2023.113602] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/06/2023]
Abstract
The endoplasmic reticulum (ER) controls many biological functions besides maintaining the function of liver cells. Various studies reported the role of the ER stress and UPR signaling pathway in various liver diseases via triggering hepatocytes apoptosis. This study aims to investigate the suppressive effect of β-sitosterol (βS) on apoptosis associated with liver injury and ER stress. METHODS Liver damage in rats was induced by TAA (150 mg/kg I.P twice a week/3 weeks) and γ-irradiation (single dose 3.5 Gy) and treated with βS (20 mg/kg daily for 30 days). Serum aminotransferase activity, lipid profile and lipid metabolic factors were measured beside liver oxidative stress and inflammatory markers. Moreover, the hepatic expression of ER stress markers (inositol-requiring enzyme 1 alpha (IRE1α), X-box-binding protein 1 (XBP1) and CCAAT/enhancer binding protein homologous protein (CHOP) and apoptotic markers were detected together with histopathological examination. RESULTS βS diminished the aminotransferase activity, the oxidative stress markers as well as the inflammatory mediators. Furthermore, βS lowered the circulating TG and TC and the hepatic lipotoxicity via the suppression of lipogenesis (Srebp-1c) and improved the β-oxidation (Pparα and Cpt1a) together with the mitochondrial biogenesis (Pgc-1 α). Moreover, the upregulated levels of ER stress markers were reduced upon treatment with βS, which consequently attenuated hepatic apoptosis. CONCLUSION βS relieves hepatic injury, ameliorates mitochondrial biogenesis, and reduces lipotoxicity and apoptosis via inhibition of CHOP and ER stress response.
Collapse
Affiliation(s)
- Omayma Ar Abo-Zaid
- Biochemistry and Molecular Biology Department, Faculty of Vet. Med. Benha University, Egypt.
| | - Fatma Sm Moawed
- Health Radiation Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| | - Effat Soliman Ismail
- Biochemistry and Molecular Biology Department, Faculty of Vet. Med. Benha University, Egypt.
| | - Esraa S A Ahmed
- Radiation Biology Research, National Center for Radiation Research and Technology, Egyptian Atomic Energy Authority, Cairo, Egypt.
| |
Collapse
|
7
|
Bi Y, Liang H, Han X, Li K, Zhang W, Lai Y, Wang Q, Jiang X, Zhao X, Fan H. β-Sitosterol Suppresses LPS-Induced Cytokine Production in Human Umbilical Vein Endothelial Cells via MAPKs and NF- κB Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2023; 2023:9241090. [PMID: 36636603 PMCID: PMC9831711 DOI: 10.1155/2023/9241090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 11/02/2022] [Accepted: 11/03/2022] [Indexed: 01/05/2023]
Abstract
Atherosclerosis (AS) is an inflammatory disease, whose occurrence and development mechanism is related to a great number of inflammatory cytokines. β-sitosterol (BS), a natural compound extracted from numerous vegetables and plant medicines, has been suggested to improve AS, but the underlying mechanism remains vague. This work focused on investigating how BS affected the lipopolysaccharide (LPS)-treated human umbilical vein endothelial cells (HUVECs) and further exploring the potential targets and mechanisms through network pharmacology (NP) and molecular docking (MD). According to in vitro experiments, LPS resulted in an increase in the expression of inflammatory cytokines like tumor necrosis factor-α (TNF-α), cyclooxygenase-2 (Cox-2), and interleukin-6 (IL-6). Besides, secretion of IL-6, interleukin-1β (IL-1β), and TNF-α also increased in HUVECs, whereas BS decreased the expression and secretion of these cytokines. NP analysis revealed that the improvement effect of BS on AS was the result of its comprehensive actions targeting 99 targets and 42 pathways. In this network, MAPKs signaling pathway was the core pathway, whereas MAPK1, MAPK8, MAPK14, and NFKB1 were the hub targets. MD analysis also successfully validated the interactions between BS and these targets. Moreover, verification test results indicated that BS downregulated the abnormal expression and activation of MAPKs and NF-κB signaling pathways in LPS-treated cells, including p38, JNK, ERK, NF-κB, and IκB-α phosphorylation expressions. Furthermore, p65 nuclear translocation was also regulated by BS treatment. In conclusion, the BS-related mechanisms in treating AS are possibly associated with inflammatory response inhibition by regulating MAPKs and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Yiming Bi
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510140, China
| | - Hongfeng Liang
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Xin Han
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Kongzheng Li
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Wei Zhang
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Yigui Lai
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Qiang Wang
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
| | - Xuefeng Jiang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xiaoshan Zhao
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Huijie Fan
- Department of Traditional Chinese Medicine, People's Hospital of Yangjiang, Yangjiang 529500, China
- Key Laboratory of Ministry of Education for TCM Viscera-State Theory and Applications, Liaoning University of Traditional Chinese Medicine, Shenyang 110032, China
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| |
Collapse
|
8
|
Wang D, Koh E, Lee KA, Chung HS. Chemical constituents from
Betula schmidtii
and their free radical scavenging, tyrosinase inhibitory, and neuroprotective activities. B KOREAN CHEM SOC 2022. [DOI: 10.1002/bkcs.12650] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022]
Affiliation(s)
- Da‐Hye Wang
- College of Science and Technology Duksung Women's University Seoul Republic of Korea
| | - Eun‐Hie Koh
- College of Science and Technology Duksung Women's University Seoul Republic of Korea
| | - Kyung Ae Lee
- Department of Food and Nutrition Anyang University Anyang Republic of Korea
| | - Ha Sook Chung
- College of Science and Technology Duksung Women's University Seoul Republic of Korea
| |
Collapse
|
9
|
Long L, Gao C, Qiu J, Yang L, Wei H, Zhou Y. Fatty acids and nutritional components of the seed oil from Wangmo red ball Camellia oleifera grown in the low-heat valley of Guizhou, China. Sci Rep 2022; 12:16554. [PMID: 36192507 PMCID: PMC9530227 DOI: 10.1038/s41598-022-20576-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/15/2022] [Indexed: 12/02/2022] Open
Abstract
Wangmo red ball Camellia oleifera is the main Camellia species cultivated for oil in the low-heat valley of Guizhou, China. In this study, we evaluated the comprehensive nutritional value of Wangmo C. oleifera seed oil through fatty acid and nutritional component analyses. Twenty excellent Wangmo C. oleifera plants with stable yield and disease resistance were selected from the Camellia oleifera germplasm resource garden in the low-heat valley site of Guizhou University. The unit crown yield, fatty acid content of the seed oil, fatty acid composition and functional nutrients were determined, and the oil quality was comprehensively evaluated using principal component analysis. The fatty acid content of C. oleifera seed oil was 35.03–53.47%, suggesting likely popularization and wide application prospects. The fatty acids included SFAs, MUFAs and PUFAs, and the oleic acid content was 80%, indicating a highly stable and nutritious oil. The oil was also rich in carotenoids, polyphenols, flavonoids, β-sitosterol, squalene and α-Ve, with average content of 7.404 mg/kg, 16.062 mg/kg, 0.401 g/100 g, 265.087 mg/kg, 129.315 mg/kg and 21.505 mg/100 g, respectively. However, the correlations among the nutritional indices were weak. PCA showed that germplasms GH7, GH43, GH28, GH8 and GH31 exhibited the top five nutritional qualities. The rankings in this study provide data for identifying excellent Wangmo C. oleifera plants with high nutritional quality. Additionally, this study provides a valuable reference for the research and development of high-end edible oil and a theoretical basis for the development of economic forest species in low-heat valley areas across the world.
Collapse
Affiliation(s)
- Li Long
- Institute for Forest Resources and Environment of Guizhou, Key Laboratory of Forest Cultivation in Plateau Mountain of Guizhou Province, College of Forestry, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Chao Gao
- Institute for Forest Resources and Environment of Guizhou, Key Laboratory of Forest Cultivation in Plateau Mountain of Guizhou Province, College of Forestry, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China.
| | - Jie Qiu
- Institute for Forest Resources and Environment of Guizhou, Key Laboratory of Forest Cultivation in Plateau Mountain of Guizhou Province, College of Forestry, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Lu Yang
- Institute for Forest Resources and Environment of Guizhou, Key Laboratory of Forest Cultivation in Plateau Mountain of Guizhou Province, College of Forestry, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Hongli Wei
- Institute for Forest Resources and Environment of Guizhou, Key Laboratory of Forest Cultivation in Plateau Mountain of Guizhou Province, College of Forestry, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| | - Yunchao Zhou
- Institute for Forest Resources and Environment of Guizhou, Key Laboratory of Forest Cultivation in Plateau Mountain of Guizhou Province, College of Forestry, Guizhou University, Jiaxiu South Road, Guiyang, 550025, China
| |
Collapse
|
10
|
Peng X, Xia Y, Xie J, Liu H, Fan L, Yu C, Ni X. Mechanism of Thunberg Fritillaria in treating endometriosis based on network pharmacology and the effect of Peiminine on the MEK/ERK pathway. Am J Transl Res 2022; 14:6196-6209. [PMID: 36247281 PMCID: PMC9556459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 07/03/2022] [Indexed: 06/16/2023]
Abstract
OBJECTIVE To explore the mechanism of Thunberg Fritillaria in treating endometriosis (EMs) based on network pharmacology and the effect of Peiminine on the MEK/ERK pathway. METHODS We applied Chinese medicine system pharmacology analysis platform (TCMSP) database and literature search to screen the main chemical components of Fritillaria thunbergii Miq and created a Vanny map from the databases of TCMSP, GENECARDS, Online Mendelian Inheritance in Man (OMIM), and some others. The STRING database was used to construct the protein interaction network of Fritillaria thunbergii Miq and EMs. The overlapping targets and enriched pathways were discovered using the cells of the innate immune annotation database (DAVID) and the Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses. To test the mechanism of Peiminine, the active ingredients of Fritillaria thunbergii, in the therapy of EMs, we designed cell assays and animal research. EMs mouse models were treated with several therapies, including fibrosis inhibitor in Peiminine by utilizing Hematoxylin-eosin staining (HE staining), MASSON staining, Immunohistochemistry, Immunofluorescence, quantitative real-time PCR (qRT-PCR) experiment, and Western blotting test. We evaluated the anti-endometriotic effects of Peiminine using 12Z human endometriotic cells. Cell Counting Kit 8 was used to assess the vitality of 12z cells (CCK8). We evaluated the migration ability of 12z cells by cell scratch test. RESULTS The effective active ingredients of Fritillaria thunbergii Miq in the treatment of EMs are Pelargonidin, Beta-sitosterol syringaresinol, Peimisine Pelargonidin-3, 5-diglucoside Ziebeimine Zhebeiresinol Verticine Solatubin OSI-2040 Chaksine Peiminine Peiminoside Peiminoside_qt, and 6-Methoxyl-2-acetyl-3-methyl-1, 4-naphthoquinone-8-O-beta-D-glucopyranoside. The critical targets for Fritillaria thunbergii Miq treating EMs are NOS2/PTGS1/AR/PPARG/PTGS2/NCOA2/RXRA/PGR/NR3C1/NCOA1/SLC6A4/OPRM1/BCL2 and ESR1. The results of GO function and KEGG enrichment analysis showed that the role pathway was estrogen-related signaling and thyroid hormone-related signaling. The expression of E-cadherin was decreased in EMs while MEK1/2, P-ERK, N-cadherin and vimentin were all increased in MASSON, immunofluorescence, Real-time PCR and Western blotting. In epithelial 12Z cells, high concentrations of Peiminine can block cell activity and migration, which is directly related to blocking cell fibrosis. CONCLUSION Overall, this study partially verified the network pharmacological prediction that Peiminine regulates the MAPK pathway in inhibiting 12Z cell proliferation and migration, and finally protects against EMs.
Collapse
Affiliation(s)
- Xia Peng
- Department of Gynecology, Shanghai Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200071, China
| | - Yue Xia
- Department of Gynecology, Shanghai Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200071, China
| | - Jiani Xie
- Department of Gynecology, Shanghai Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200071, China
| | - Honglin Liu
- Department of Gynecology, Shanghai Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200071, China
| | - Limin Fan
- The Institute for Biomedical Engineering and Nano Science Tongji University School of MedicineNo. 1239, Siping Road, Shanghai 200092, China
| | - Chaoqin Yu
- Department of Traditional Chinese Gynecology, Changhai Hospital Affiliated to Naval Medical UniversityShanghai 200071, China
| | - Xiaorong Ni
- Department of Gynecology, Shanghai Traditional Chinese Medicine Hospital, Shanghai University of Traditional Chinese MedicineShanghai 200071, China
| |
Collapse
|
11
|
Pei J, Prasad M, Mohamed Helal G, El-Sherbiny M, Abdelmonem Elsherbini DM, Rajagopal P, Palanisamy CP, Veeraraghavan VP, Jayaraman S, Surapaneni KM. Beta-Sitosterol Facilitates GLUT4 Vesicle Fusion on the Plasma Membrane via the Activation of Rab/IRAP/Munc 18 Signaling Pathways in Diabetic Gastrocnemius Muscle of Adult Male Rats. Bioinorg Chem Appl 2022; 2022:7772305. [PMID: 35992048 PMCID: PMC9388314 DOI: 10.1155/2022/7772305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/21/2022] [Indexed: 12/18/2022] Open
Abstract
Nutritional overload in the form of high-fat and nonglycolysis sugar intake contributes towards the accelerated creation of reactive oxygen species (ROS), hyperglycemia, and dyslipidemia. Glucose absorption and its subsequent oxidation processes in fat and muscle tissues alter as a consequence of these modifications. Insulin resistance (IR) caused glucose transporter 4 (GLUT4) translocation to encounter a challenge that manifested itself as changes in glycolytic pathways and insulin signaling. We previously found that beta (β)-sitosterol reduces IR in fat tissue via IRS-1/PI3K/Akt facilitated signaling due to its hypolipidemic and hypoglycemic activity. The intention of this research was to see whether the phytosterol β-sitosterol can aid in the translocation of GLUT4 in rats fed on high-fat diet (HFD) and sucrose by promoting Rab/IRAP/Munc 18 signaling molecules. The rats were labeled into four groups, namely control rats, HFD and sucrose-induced diabetic control rats, HFD and sucrose-induced diabetic rats given oral dose of 20 mg/kg body wt./day of β-sitosterol treatment for 30 days, and HFD and sucrose-induced diabetic animals given oral administration of 50 mg/kg body wt./day metformin for 30 days. Diabetic rats administered with β-sitosterol and normalized the titers of blood glucose, serum insulin, serum testosterone, and the status of insulin tolerance and oral glucose tolerance. In comparison with the control group, β-sitosterol effectively regulated both glycolytic and gluconeogenesis enzymes. Furthermore, qRT-PCR analysis of the mRNA levels of key regulatory genes such as SNAP23, VAMP-2, syntaxin-4, IRAP, vimentin, and SPARC revealed that β-sitosterol significantly regulated the mRNA levels of the above genes in diabetic gastrocnemius muscle. Protein expression analysis of Rab10, IRAP, vimentin, and GLUT4 demonstrated that β-sitosterol had a positive effect on these proteins, resulting in effective GLUT4 translocation in skeletal muscle. According to the findings, β-sitosterol reduced HFD and sucrose-induced IR and augmented GLUT4 translocation in gastrocnemius muscle through insulin signaling modulation via Rab/IRAP/Munc 18 and glucose metabolic enzymes. The present work is the first of its kind to show that β-sitosterol facilitates GLUT4 vesicle fusion on the plasma membrane via Rab/IRAP/Munc 18 signaling molecules in gastrocnemius muscle.
Collapse
Affiliation(s)
- JinJin Pei
- Qinba State Key Laboratory of Biological Resources and Ecological Environment, 2011 QinLing-Bashan Mountains, Bioresources Comprehensive Development C. I. C, Shaanxi Province Key Laboratory of Bio-Resources, College of Bioscience and Bioengineering, Shaanxi University of Technology, Hanzhong 723001, Shaanxi, China
| | - Monisha Prasad
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Ghada Mohamed Helal
- Department of Medical Biochemistry, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed El-Sherbiny
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Dalia Mahmoud Abdelmonem Elsherbini
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Jouf University, P.O. Box 2014, Sakaka, Saudi Arabia
- Department of Anatomy, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Ponnulakshmi Rajagopal
- Central Research Laboratory, Meenakshi Academy of Higher Education and Research (Deemed to be University), Chennai 600078, India
| | - Chella Perumal Palanisamy
- State Key Laboratory of Biobased Material and Green Papermaking, College of Food Science and Engineering, Qilu University of Technology, Shandong Academy of Science, Jinan 250353, China
| | - Vishnu Priya Veeraraghavan
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Selvaraj Jayaraman
- Centre of Molecular Medicine and Diagnostics (COMManD), Department of Biochemistry, Saveetha Dental College & Hospital, Saveetha Institute of Medical & Technical Sciences, Saveetha University, Chennai 600077, India
| | - Krishna Mohan Surapaneni
- Departments of Biochemistry,Molecular Virology,Medical Education,Research,Clinical Skills & Simulation, Panimalar Medical College Hospital & Research Institute, Varadharajapuram, Poonamallee, Chennai 600123, India
| |
Collapse
|
12
|
Bao X, Zhang Y, Zhang H, Xia L. Molecular Mechanism of β-Sitosterol and its Derivatives in Tumor Progression. Front Oncol 2022; 12:926975. [PMID: 35756648 PMCID: PMC9213880 DOI: 10.3389/fonc.2022.926975] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
β-Sitosterol (SIT), a white powdery organic substance with a molecular formula of C29H50O, is one of the most abundant naturally occurring phytosterols in plants. With a chemical composition similar to that of cholesterol, SIT is applied in various fields such as medicine, agriculture, and chemical industries, owing to its unique biological and physicochemical properties. Modern pharmacological studies have elucidated good anti-tumor therapeutic effect activity of SIT, which mainly manifests as pro-apoptotic, anti-proliferative, anti-metastatic, anti-invasive, and chemosensitizing on tumor cells. In addition, SIT exerts an anti-tumor effect on multiple malignant tumors such as breast, gastric, lung, kidney, pancreatic, prostate, and other cancers. Further, SIT derivatives with structural modifications are promising anti-tumor drugs with significant anti-tumor effects. This review article focuses on recent studies relevant to the anti-tumor effects of SIT and summarizes its anti-tumor mechanism to provide a reference for the clinical treatment of malignant tumors and the development of novel anti-tumor drugs.
Collapse
Affiliation(s)
- Xingxun Bao
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yanan Zhang
- School of Chinese Medicine, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hairong Zhang
- Department of Obstetrics and Gynecology, Shandong Provincial Third Hospital, Jinan, China
| | - Lei Xia
- Department of Pathology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
13
|
Chen YP, Wang KX, Cai JQ, Li Y, Yu HL, Wu Q, Meng W, Wang H, Yin CH, Wu J, Huang MB, Li R, Guan DG. Detecting Key Functional Components Group and Speculating the Potential Mechanism of Xiao-Xu-Ming Decoction in Treating Stroke. Front Cell Dev Biol 2022; 10:753425. [PMID: 35646921 PMCID: PMC9136080 DOI: 10.3389/fcell.2022.753425] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 02/25/2022] [Indexed: 02/05/2023] Open
Abstract
Stroke is a cerebrovascular event with cerebral blood flow interruption which is caused by occlusion or bursting of cerebral vessels. At present, the main methods in treating stroke are surgical treatment, statins, and recombinant tissue-type plasminogen activator (rt-PA). Relatively, traditional Chinese medicine (TCM) has widely been used at clinical level in China and some countries in Asia. Xiao-Xu-Ming decoction (XXMD) is a classical and widely used prescription in treating stroke in China. However, the material basis of effect and the action principle of XXMD are still not clear. To solve this issue, we designed a new system pharmacology strategy that combined targets of XXMD and the pathogenetic genes of stroke to construct a functional response space (FRS). The effective proteins from this space were determined by using a novel node importance calculation method, and then the key functional components group (KFCG) that could mediate the effective proteins was selected based on the dynamic programming strategy. The results showed that enriched pathways of effective proteins selected from FRS could cover 99.10% of enriched pathways of reference targets, which were defined by overlapping of component targets and pathogenetic genes. Targets of optimized KFCG with 56 components can be enriched into 166 pathways that covered 80.43% of 138 pathways of 1,012 pathogenetic genes. A component potential effect score (PES) calculation model was constructed to calculate the comprehensive effective score of components in the components-targets-pathways (C-T-P) network of KFCGs, and showed that ferulic acid, zingerone, and vanillic acid had the highest PESs. Prediction and docking simulations show that these components can affect stroke synergistically through genes such as MEK, NFκB, and PI3K in PI3K-Akt, cAMP, and MAPK cascade signals. Finally, ferulic acid, zingerone, and vanillic acid were tested to be protective for PC12 cells and HT22 cells in increasing cell viabilities after oxygen and glucose deprivation (OGD). Our proposed strategy could improve the accuracy on decoding KFCGs of XXMD and provide a methodologic reference for the optimization, mechanism analysis, and secondary development of the formula in TCM.
Collapse
Affiliation(s)
- Yu-peng Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Ke-xin Wang
- Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration, Department of Neurosurgery, National Key Clinical Specialty/Engineering Technology Research Center of Education Ministry of China, Neurosurgery Institute, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jie-qi Cai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Yi Li
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Hai-lang Yu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Qi Wu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wei Meng
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Handuo Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Chuan-hui Yin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Jie Wu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China
| | - Mian-bo Huang
- Department of Histology and Embryology, Guangdong Provincial Key Laboratory of Construction and Detection in Tissue Engineering, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| | - Rong Li
- Department of Cardiovascular Disease, First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| | - Dao-gang Guan
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, China,Guangdong Provincial Key Laboratory of Single Cell Technology and Application, Southern Medical University, Guangzhou, China,*Correspondence: Mian-bo Huang, ; Rong Li, ; Dao-gang Guan,
| |
Collapse
|
14
|
Pathak K, Pathak MP, Saikia R, Gogoi U, Sahariah JJ, Zothantluanga JH, Samanta A, Das A. Cancer Chemotherapy via Natural Bioactive Compounds. Curr Drug Discov Technol 2022; 19:e310322202888. [PMID: 35362385 DOI: 10.2174/1570163819666220331095744] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 11/29/2021] [Accepted: 12/17/2021] [Indexed: 12/09/2022]
Abstract
BACKGROUND Cancer-induced mortality is increasingly prevalent globally which skyrocketed the necessity to discover new/novel safe and effective anticancer drugs. Cancer is characterized by the continuous multiplication of cells in the human which is unable to control. Scientific research is drawing its attention towards naturally-derived bioactive compounds as they have fewer side effects compared to the current synthetic drugs used for chemotherapy. OBJECTIVE Drugs isolated from natural sources and their role in the manipulation of epigenetic markers in cancer are discussed briefly in this review article. METHODS With advancing medicinal plant biotechnology and microbiology in the past century, several anticancer phytomedicines were developed. Modern pharmacopeia contains at least 25% herbal-based remedy including clinically used anticancer drugs. These drugs mainly include the podophyllotoxin derivatives vinca alkaloids, curcumin, mistletoe plant extracts, taxanes, camptothecin, combretastatin, and others including colchicine, artesunate, homoharringtonine, ellipticine, roscovitine, maytanasin, tapsigargin,andbruceantin. RESULTS Compounds (psammaplin, didemnin, dolastin, ecteinascidin,and halichondrin) isolated from marine sources and animals such as microalgae, cyanobacteria, heterotrophic bacteria, invertebrates. They have been evaluated for their anticancer activity on cells and experimental animal models and used chemotherapy.Drug induced manipulation of epigenetic markers plays an important role in the treatment of cancer. CONCLUSION The development of a new drug from isolated bioactive compounds of plant sources has been a feasible way to lower the toxicity and increase their effectiveness against cancer. Potential anticancer therapeutic leads obtained from various ethnomedicinal plants, foods, marine, and microorganisms are showing effective yet realistically safe pharmacological activity. This review will highlight important plant-based bioactive compounds like curcumin, stilbenes, terpenes, other polyphenolic phyto-compounds, and structurally related families that are used to prevent/ ameliorate cancer. However, a contribution from all possible fields of science is still a prerequisite for discovering safe and effective anticancer drugs.
Collapse
Affiliation(s)
- Kalyani Pathak
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| | - Manash Pratim Pathak
- Faculty of Pharmaceutical Sciences, Assam down town University, Panikhaiti, Guwahati-781026, Assam, India
| | - Riya Saikia
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| | - Urvashee Gogoi
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| | - Jon Jyoti Sahariah
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| | - James H Zothantluanga
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| | - Abhishek Samanta
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| | - Aparoop Das
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh - 786004, Assam, India
| |
Collapse
|
15
|
Pavani M, Singha P, Dash DR, Asaithambi N, Singh SK. Novel encapsulation approaches for phytosterols and their importance in food products: A review. J FOOD PROCESS ENG 2022. [DOI: 10.1111/jfpe.14041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Mekala Pavani
- Department of Food Process Engineering National Institute of Technology (NIT) Rourkela Rourkela India
| | - Poonam Singha
- Department of Food Process Engineering National Institute of Technology (NIT) Rourkela Rourkela India
| | - Dibya Ranjan Dash
- Department of Food Process Engineering National Institute of Technology (NIT) Rourkela Rourkela India
| | - Niveditha Asaithambi
- Department of Food Process Engineering National Institute of Technology (NIT) Rourkela Rourkela India
| | - Sushil Kumar Singh
- Department of Food Process Engineering National Institute of Technology (NIT) Rourkela Rourkela India
| |
Collapse
|
16
|
Han HS, Koo SY, Choi KY. Emerging nanoformulation strategies for phytocompounds and applications from drug delivery to phototherapy to imaging. Bioact Mater 2021; 14:182-205. [PMID: 35310344 PMCID: PMC8892098 DOI: 10.1016/j.bioactmat.2021.11.027] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/18/2022] Open
Abstract
Over thousands of years, natural bioactive compounds derived from plants (bioactive phytocompounds, BPCs) have been used worldwide to address human health issues. Today, they are a significant resource for drug discovery in the development of modern medicines. Although many BPCs have promising biological activities, most of them cannot be effectively utilized in drugs for therapeutic applications because of their inherent limitations of low solubility, structural instability, short half-life, poor bioavailability, and non-specific distribution to organs. Researchers have utilized emerging nanoformulation (NF) technologies to overcome these limitations as they have demonstrated great potential to improve the solubility, stability, and pharmacokinetic and pharmacodynamic characteristics of BPCs. This review exemplifies NF strategies for resolving the issues associated with BPCs and summarizes recent advances in their preclinical and clinical applications for imaging and therapy. This review also highlights how innovative NF technologies play a leading role in next-generation BPC-based drug development for extended therapeutic applications. Finally, this review discusses the opportunities to take BPCs with meaningful clinical impact from bench to bedside and extend the patent life of BPC-based medicines with new formulations or application to new adjacent diseases beyond the primary drug indications. Natural bioactive phytocompounds derived from plants have been used worldwide to address human health issues. However, most of them cannot be effectively utilized in drugs for therapeutic applications because of their inherent limitations. Nanoformulation approach has recently been underlined as an emerging pharmaceutical strategy to overcome the intrinsic drawbacks of bioactive phytocompounds. Various types of nanoformulation and their up-to-date applications for targeted delivery, phototherapy, and imaging are reviewed. Finally, their clinical implications for the repurposing of bioactive phytocompounds are deliberated.
Collapse
Affiliation(s)
- Hwa Seung Han
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Song Yi Koo
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
| | - Ki Young Choi
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea
- Division of Bio-Medical Science and Technology, KIST School, University of Science and Technology (UST), Seoul, 02792, Republic of Korea
- Corresponding author. Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, 25451, Republic of Korea.
| |
Collapse
|
17
|
Liu X, Shen J, Zong J, Liu J, Jin Y. Beta-Sitosterol Promotes Milk Protein and Fat Syntheses-Related Genes in Bovine Mammary Epithelial Cells. Animals (Basel) 2021; 11:ani11113238. [PMID: 34827970 PMCID: PMC8614283 DOI: 10.3390/ani11113238] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/09/2021] [Accepted: 11/10/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary The levels of milk fats and proteins are important indexes used to evaluate milk quality. Generally, feed additives are used to improve milk quality. This study aimed to investigate the effect of β-sitosterol on milk fat and protein gene expression in bovine mammary epithelial cells. β-sitosterol increased the β-casein levels in bovine mammary epithelial cells and promoted the expression of milk fat and protein synthesis-related genes, suggesting the use of β-sitosterol as a potential feed additive to improve milk quality in dairy cows. Abstract β-sitosterol, a phytosterol with multiple biological activities, has been used in the pharmaceutical industry. However, there are only a few reports on the use of β-sitosterol in improving milk synthesis in dairy cows. This study aimed to investigate the effects of β-sitosterol on milk fat and protein syntheses in bovine mammary epithelial cells (MAC-T) and its regulatory mechanism. MAC-T cells were treated with different concentrations (0.01, 0.1, 1, 5, 10, 20, 30, or 40 μM) of β-sitosterol, and the expression levels of milk protein and fat synthesis-related genes and proteins were analyzed. β-sitosterol at 0.1, 1, and 10 μM concentrations promoted the mRNA and protein expression of β-casein. β-sitosterol (0.1, 1, 10 μM) increased the mRNA and protein expression levels of signal transducer activator of transcription 5 (STAT5), mammalian target of rapamycin (mTOR), and ribosomal protein S6 kinase beta-1 (S6K1) of the JAK2/STAT5 and mTOR signaling pathways. It also stimulated the milk fat synthesis-related factors, including sterol regulatory element-binding protein 1 (SREBP1), peroxisome proliferator-activated receptor-gamma (PPARγ), acetyl-CoA carboxylase (ACC), lipoprotein lipase (LPL), and stearyl CoA desaturase (SCD). β-sitosterol (0.1, 1, 10 μM) also significantly increased the expression of growth hormone/insulin-like growth factor-1 (GH/IGF-1) axis and hypoxia-inducible factor-1α (HIF-1α)-related genes. Notably, the compound inhibited the expression of the negative regulator, the suppressor of cytokine signaling 2 (SOCS2) at the two lower concentrations (0.1, 1 μM), but significantly promoted the expression at the highest concentration (30 μM). These results highlight the role of β-sitosterol at concentrations ranging from 0.1 to 10 μM in improving milk protein and fat syntheses, regulating milk quality. Therefore, β-sitosterol can be used as a potential feed additive to improve milk quality in dairy cows.
Collapse
|
18
|
Phonghanpot S, Jarintanan F. Antiproliferative, antibacterial, and antioxidant activities of Bauhinia strychnifolia Craib aqueous extracts in gut and liver perspective. BMC Complement Med Ther 2021; 21:276. [PMID: 34736448 PMCID: PMC8567622 DOI: 10.1186/s12906-021-03448-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Accepted: 10/19/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Bauhinia strychnifolia Craib is an herb in Thai traditional medicine. Its decoction is traditionally used as an anticancer, antidiarrheal, and hangover remedy for centuries. Several studies described bioactivities of its organic solvent extracts, however, only few demonstrated the usefulness of the decoction. Here, we aimed to determine the bioactivities of Bauhinia strychnifolia Craib root and stem aqueous extracts in gut and liver perspective. METHODS To achieve the goal, we performed MTT test, microscopic analyses, disc diffusion assay, broth microdilution assay, free radicals scavenging assays, and LC-MS analysis. RESULTS We found that the extracts inhibited the growth of human hepatocellular carcinoma (HepG2) and colon adenocarcinoma (HT-29) cell lines. Moreover, they also inhibited the growth of gram-positive bacteria Staphylococcus aureus and Bacillus cereus but not inhibited the growth of gram-negative bacteria Escherichia coli and Pseudomonas aeruginosa. Furthermore, the extracts exhibited moderate antioxidant activity and increased GSH production in HepG2 cell line when compared with untreated. Our LC-MS analysis confirmed the existence of anticancer and antioxidant; 3,5,7,3',5'-pentahydroxyflavanonol-3-O-α-L-rhamnopyranoside and β-sitosterol, in the extracts. CONCLUSION The results from our study supported that the administration of Bauhinia strychnifolia Craib root and stem decoction would really aid colon or liver cancer patients and detoxify the alcoholic drunkard as it is claimed in Thai traditional medicine.
Collapse
Affiliation(s)
- Suranat Phonghanpot
- Biochemistry Unit, Department of Biomedical Science, Faculty of Sciences, Rangsit University, 52/347 Muang Ake, Phaholyothin road, Lak Hok, Muang, Pathum Thani, 12000, Thailand
| | - Faongchat Jarintanan
- Faculty of Medical Technology, Rangsit University, 52/347 Muang Ake, Phaholyothin road, Lak Hok, Muang, Pathum Thani, 12000, Thailand.
| |
Collapse
|
19
|
Karantas ID, Okur ME, Okur NÜ, Siafaka PI. Dyslipidemia Management in 2020: An Update on Diagnosis and Therapeutic Perspectives. Endocr Metab Immune Disord Drug Targets 2021; 21:815-834. [PMID: 32778041 DOI: 10.2174/1871530320666200810144004] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Revised: 06/10/2020] [Accepted: 06/19/2020] [Indexed: 11/22/2022]
Abstract
Cardiovascular diseases are the leading cause of death in the modern world and dyslipidemia is one of the major risk factors. The current therapeutic strategies for cardiovascular diseases involve the management of risk factors, especially dyslipidemia and hypertension. Recently, the updated guidelines of dyslipidemia management were presented, and the newest data were included in terms of diagnosis, imaging, and treatment. In this targeted literature review, the researchers presented the most recent evidence on dyslipidemia management by including the current therapeutic goals for it. In addition, the novel diagnostic tools based on theranostics are shown. Finally, the future perspectives on treatment based on novel drug delivery systems and their potential to be used in clinical trials were also analyzed. It should be noted that dyslipidemia management can be achieved by the strict lifestyle change, i.e., by adopting a healthy life, and choosing the most suitable medication. This review can help medical professionals as well as specialists of other sciences to update their knowledge on dyslipidemia management, which can lead to better therapeutic outcomes and newer drug developments.
Collapse
Affiliation(s)
| | - Mehmet E Okur
- University of Health Sciences, Faculty of Pharmacy, Department of Pharmacology, Istanbul, Turkey
| | - Neslihan Ü Okur
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| | - Panoraia I Siafaka
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Health Sciences, Istanbul, Turkey
| |
Collapse
|
20
|
Potential Antibacterial Activity of Bioactive β-sitosterol from Root Bark of Rhizophora apiculata from Lampung Coastal. JURNAL KIMIA SAINS DAN APLIKASI 2021. [DOI: 10.14710/jksa.24.4.114-119] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
β-sitosterol is an essential bioactive phytosterol naturally present in plant cell membranes. It has a coincident structure with animal cholesterol. This investigation reported isolation, structure analysis, and an antimicrobial assay of β-sitosterol from the root bark of Bakau Minyak (Rhizophora apiculata) from Lampung coastal. The isolation of β-sitosterol was carried out through maceration using methanol, separation by vacuum liquid chromatography (VLC), and purification by column chromatography (CC) using ethyl acetate/n-hexane (2:8) as eluent. The structure of β-sitosterol was determined using spectroscopic analysis (UV-Vis, FT-IR, 13C-NMR, 1H-NMR, DEPT, and GC-MS). The pure β-sitosterol has 107.4 mg of white needle crystalline compound, the compound melting point about 140.7-141.2oC, the molecular mass confirmed by m/z 414, and UV absorption detected at λ 203.9 nm. The β-sitosterol antimicrobial bioactivity assay has shown potential activity to be developed as a lead compound against E. coli.
Collapse
|
21
|
A Preliminary Study on the Effect of Psyllium Husk Ethanolic Extract on Hyperlipidemia, Hyperglycemia, and Oxidative Stress Induced by Triton X-100 Injection in Rats. BIOLOGY 2021; 10:biology10040335. [PMID: 33923513 PMCID: PMC8074146 DOI: 10.3390/biology10040335] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/12/2021] [Accepted: 04/13/2021] [Indexed: 02/06/2023]
Abstract
The aim of this study is to assess the efficiency of psyllium husk ethanolic extract (PHEE) on Triton X-100 induced hyperlipidemic rats by studying the changes in hepatic and pancreatic function and histopathology. Forty male albino rats (bodyweight 175-188 g) were grouped randomly into four sets with ten rats. The experimental groups included: (1) control group (CON); (2) Triton X-100 induced hyperlipidemic group-rats were intraperitoneally injected with a single dose of Triton X-100 (100 mg/kg body weight) on the 21st day of Trial onset; (3) PHEE group-PHEE was orally administered (100 mg/kg body weight dissolved in 1 mL of distilled water) by gastric tube from the first day of the experiment until the fortieth day, once daily, (PHEE); (4) PHEE +Triton group, which received PHEE orally with the induction of hyperlipidemia. Treating hyperlipidemic rats with PHEE showed a decrease in the total serum lipids, triglyceride (TG), total cholesterol (TC), atherogenic index (AI), and malondialdehyde (MDA) with an increase in superoxide dismutase (SOD) and catalase (CAT) activities. PHEE administration alleviated the negative impact of Triton on the serum levels of glucose, insulin, glycated hemoglobin (HbA1c), homeostatic model assessment for insulin resistance (HOMA IR index), leptin hormone, Alanine Aminotransferase (ALT), Aspartate Aminotransferase (AST), Gamma-Glutamyl Transferase (GGT) and proteinogram. The Triton-induced hyperlipidemic rats showed extensive histopathological changes in the liver and pancreas, which were alleviated with PHEE administration. It could be concluded that PHEE has potent effects against hyperlipidemia, hyperglycemia, and oxidative stress due to its biologically active constituents detected by GC-MS analysis. This study's findings may help develop a novel trial against the effects of hyperlipidemia in the future.
Collapse
|
22
|
Difonzo G, Troilo M, Squeo G, Pasqualone A, Caponio F. Functional compounds from olive pomace to obtain high-added value foods - a review. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2021; 101:15-26. [PMID: 32388855 DOI: 10.1002/jsfa.10478] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 04/25/2020] [Accepted: 05/09/2020] [Indexed: 06/11/2023]
Abstract
Olive pomace, the solid by-product from virgin olive oil extraction, constitutes a remarkable source of functional compounds and has been exploited by several authors to formulate high value-added foods and, consequently, to foster the sustainability of the olive-oil chain. In this framework, the aim of the present review was to summarize the results on the application of functional compounds from olive pomace in food products. Phenolic-rich extracts from olive pomace were added to vegetable oils, fish burgers, fermented milk, and in the edible coating of fruit, to take advantage of their antioxidant and antimicrobial effects. Olive pomace was also used directly in the formulation of pasta and baked goods, by exploiting polyunsaturated fatty acids, phenolic compounds, and dietary fiber to obtain high value-added healthy foods and / or to extend their shelf-life. With the same scope, olive pomace was also added to animal feeds, providing healthy, improved animal products. Different authors used olive pomace to produce biodegradable materials and / or active packaging able to increase the content of bioactive compounds and the oxidative stability of foods. Overall, the results highlighted, in most cases, the effectiveness of the addition of olive pomace-derived functional compounds in improving nutritional value, quality, and / or the shelf-life of foods. However, the direct addition of olive pomace was found to be more challenging, especially due to alterations in the sensory and textural features of food. © 2020 Society of Chemical Industry.
Collapse
Affiliation(s)
- Graziana Difonzo
- Department of Soil, Plant and Food Science (DISSPA), University of Bari Aldo Moro, Bari, Italy
| | - Marica Troilo
- Department of Soil, Plant and Food Science (DISSPA), University of Bari Aldo Moro, Bari, Italy
| | - Giacomo Squeo
- Department of Soil, Plant and Food Science (DISSPA), University of Bari Aldo Moro, Bari, Italy
| | - Antonella Pasqualone
- Department of Soil, Plant and Food Science (DISSPA), University of Bari Aldo Moro, Bari, Italy
| | - Francesco Caponio
- Department of Soil, Plant and Food Science (DISSPA), University of Bari Aldo Moro, Bari, Italy
| |
Collapse
|
23
|
An update on β-sitosterol: A potential herbal nutraceutical for diabetic management. Biomed Pharmacother 2020; 131:110702. [DOI: 10.1016/j.biopha.2020.110702] [Citation(s) in RCA: 84] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2020] [Revised: 07/26/2020] [Accepted: 08/26/2020] [Indexed: 01/12/2023] Open
|
24
|
Olorundare O, Adeneye A, Akinsola A, Kolo P, Agede O, Soyemi S, Mgbehoma A, Okoye I, Albrecht R, Mukhtar H. Irvingia gabonensis Seed Extract: An Effective Attenuator of Doxorubicin-Mediated Cardiotoxicity in Wistar Rats. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:1602816. [PMID: 33149803 PMCID: PMC7603620 DOI: 10.1155/2020/1602816] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/08/2020] [Accepted: 10/05/2020] [Indexed: 01/16/2023]
Abstract
Cardiotoxicity as an off-target effect of doxorubicin therapy is a major limiting factor for its clinical use as a choice cytotoxic agent. Seeds of Irvingia gabonensis have been reported to possess both nutritional and medicinal values which include antidiabetic, weight losing, antihyperlipidemic, and antioxidative effects. Protective effects of Irvingia gabonensis ethanol seed extract (IGESE) was investigated in doxorubicin (DOX)-mediated cardiotoxicity induced with single intraperitoneal injection of 15 mg/kg of DOX following the oral pretreatments of Wistar rats with 100-400 mg/kg/day of IGESE for 10 days, using serum cardiac enzyme markers (cardiac troponin I (cTI) and lactate dehydrogenase (LDH)), cardiac tissue oxidative stress markers (catalase (CAT), malonyldialdehyde (MDA), superoxide dismutase (SOD), glutathione-S-transferase (GST), glutathione peroxidase (GSH-Px), and reduced glutathione (GSH)), and cardiac histopathology endpoints. In addition, both qualitative and quantitative analyses to determine IGESE's secondary metabolites profile and its in vitro antioxidant activities were also conducted. Results revealed that serum cTnI and LDH were significantly elevated by the DOX treatment. Similarly, activities of tissue SOD, CAT, GST, and GSH levels were profoundly reduced, while GPx activity and MDA levels were profoundly increased by DOX treatment. These biochemical changes were associated with microthrombi formation in the DOX-treated cardiac tissues on histological examination. However, oral pretreatments with 100-400 mg/kg/day of IGESE dissolved in 5% DMSO in distilled water significantly attenuated increases in the serum cTnI and LDH, prevented significant alterations in the serum lipid profile and the tissue activities and levels of oxidative stress markers while improving cardiovascular disease risk indices and DOX-induced histopathological lesions. The in vitro antioxidant studies showed IGESE to have good antioxidant profile and contained 56 major secondary metabolites prominent among which are γ-sitosterol, Phytol, neophytadiene, stigmasterol, vitamin E, hexadecanoic acid and its ethyl ester, Phytyl palmitate, campesterol, lupeol, and squalene. Overall, both the in vitro and in vivo findings indicate that IGESE may be a promising prophylactic cardioprotective agent against DOX-induced cardiotoxicity, at least in part mediated via IGESE's antioxidant and free radical scavenging and antithrombotic mechanisms.
Collapse
Affiliation(s)
- Olufunke Olorundare
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Adejuwon Adeneye
- Department of Pharmacology, Therapeutics and Toxicology, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Akinyele Akinsola
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Phillip Kolo
- Department of Medicine, Faculty of Clinical, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Olalekan Agede
- Department of Pharmacology and Therapeutics, Faculty of Basic Medical Sciences, College of Health Sciences, University of Ilorin, Ilorin, Kwara State, Nigeria
| | - Sunday Soyemi
- Department of Pathology and Forensic Medicine, Faculty of Basic Clinical Sciences, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Alban Mgbehoma
- Department of Pathology and Forensic Medicine, Lagos State University Teaching Hospital, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Ikechukwu Okoye
- Department of Oral Pathology and Medicine, Faculty of Dentistry, Lagos State University College of Medicine, 1-5 Oba Akinjobi Way, G.R.A., Ikeja, Lagos State, Nigeria
| | - Ralph Albrecht
- Department of Animal Sciences, 1675 Observatory Drive, University of Wisconsin, Madison, WI 53706, USA
| | - Hasan Mukhtar
- Department of Dermatology, University of Wisconsin, Madison, Medical Science Center, 1300 University Avenue, Madison, WI 53706, USA
| |
Collapse
|
25
|
Li Z, Wu YY, Yu BX. Methylophiopogonanone A, an Ophiopogon homoisoflavonoid, alleviates high-fat diet-induced hyperlipidemia: assessment of its potential mechanism. ACTA ACUST UNITED AC 2020; 53:e9201. [PMID: 32130294 PMCID: PMC7057930 DOI: 10.1590/1414-431x20199201] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Accepted: 12/04/2019] [Indexed: 11/22/2022]
Abstract
Methylophiopogonanone A (MO-A), a homoisoflavonoid extracted from Ophiopogon japonicus, has been shown to attenuate myocardial apoptosis and improve cerebral ischemia/reperfusion injury. However, the hypolipidemic effects remain unknown. This study was performed to investigate a potential hypolipidemic effect of MO-A in hyperlipidemia rats, as well as its underlying mechanism of action. A rat model of hyperlipidemia was induced by a high-fat diet (HFD). Animals were randomly divided into three groups (n=8/group): normal control group (NC), HFD group, and HFD+MO-A (10 mg·kg-1·d-1) treatment group. The effects of MO-A on serum lipids, body weight, activity of lipoprotein metabolism enzyme, and gene expression of lipid metabolism were evaluated in HFD-induced rats. In HFD-induced rats, pretreatment with MO-A decreased the body weight gain and reduced serum and hepatic lipid levels. In addition, pretreatment with MO-A improved the activities of lipoprotein lipase and hepatic lipase in serum and liver, down-regulated mRNA expression of acetyl CoA carboxylase and sterol regulatory element-binding protein 1c, and up-regulated mRNA expression of low-density lipoprotein receptor and peroxisome proliferator-activated receptor α in the liver. Our results indicated that MO-A showed strong ability to ameliorate the hyperlipidemia in HFD-induced rats. MO-A might be a potential candidate for prevention of overweight and dyslipidemia induced by HFD.
Collapse
Affiliation(s)
- Zhao Li
- Center for Translational Medicine of the First Affiliated Hospital, Sun Yatsen University, Guangzhou, China
| | - Ying-Ying Wu
- Center for Translational Medicine of the First Affiliated Hospital, Sun Yatsen University, Guangzhou, China
| | - Bei-Xin Yu
- Center for Translational Medicine of the First Affiliated Hospital, Sun Yatsen University, Guangzhou, China
| |
Collapse
|
26
|
Devaraj E, Roy A, Royapuram Veeraragavan G, Magesh A, Varikalam Sleeba A, Arivarasu L, Marimuthu Parasuraman B. β-Sitosterol attenuates carbon tetrachloride-induced oxidative stress and chronic liver injury in rats. Naunyn Schmiedebergs Arch Pharmacol 2020; 393:1067-1075. [PMID: 31930431 DOI: 10.1007/s00210-020-01810-8] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Accepted: 01/04/2020] [Indexed: 02/06/2023]
Abstract
Chronic liver diseases are clinically silent and responsible for significant morbidity and mortality worldwide. β-Sitosterol (BSS), major phytosterol in plants, has a wide spectrum of protective effect against various chronic ailments. We investigated the hepatoprotective effect of BSS against carbon tetrachloride (CCl4)-induced chronic liver injury in rats. Thirty rats were divided into five groups, with six animals in each group. Group I rats served as control while groups II, III, IV, and V rats were injected intraperitoneally with CCl4 (0.2 mL/100 g b.w. in olive oil (1:1)) for 7 consecutive weeks. After 7 weeks, group II rats were left without any treatments and served as CCl4 alone group, while groups III, IV, and V rats were treated with BSS 25 and 50 mg/kg b.w. and silymarin 100 mg/kg b.w. as oral post-treatments respectively, for the next 4 weeks. At the end of the experiment, hepatotoxicity marker enzymes in serum, oxidative stress, and fibrosis marker were analyzed. CCl4 administration caused significant elevation of marker enzymes of hepatotoxicity in serum and increased lipid peroxidation and fibrosis markers such as hydroxyproline, collagen, α-smooth muscle actin, vimentin, desmin, and matrix metalloproteinases 9 in liver tissue of rats. This treatment also caused a significant diminution of intracellular enyzmic antioxidants such as SOD and CAT in the liver tissue of rats. All the above adversities were significantly mitigated by the BSS post-treatments. The results suggest that BSS could have a hepatoprotective effect against oxidative stress-mediated CLD induced by CCl4.
Collapse
Affiliation(s)
- Ezhilarasan Devaraj
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 600 077, India.
| | - Anitha Roy
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 600 077, India
| | - Geetha Royapuram Veeraragavan
- Department of Microbiology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 600 077, India
| | - Anitha Magesh
- Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai, 602 105, India
| | | | - Lakshminarayanan Arivarasu
- Department of Pharmacology, Saveetha Dental College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 600 077, India
| | - Brundha Marimuthu Parasuraman
- Department of Pathology, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Chennai, Tamil Nadu, 600 077, India
| |
Collapse
|
27
|
Qi Y, Zang R, Lu H, Wang Z, Ma Z. Network pharmacology and bioinformatics approach reveals the hypolipidemic mechanism of Dan Tian Jiang Zhi pill. Med Chem Res 2019. [DOI: 10.1007/s00044-019-02478-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|