1
|
Karimi H, Rabbani S, Babadi D, Dadashzadeh S, Haeri A. Piperine Liposome-Embedded in Hyaluronan Hydrogel as an Effective Platform for Prevention of Postoperative Peritoneal Adhesion. J Microencapsul 2023; 40:279-301. [PMID: 36948888 DOI: 10.1080/02652048.2023.2194415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/24/2023]
Abstract
This study aimed to prepare piperine (PIP) loaded liposomes in hyaluronic acid (HA) hydrogel to provide a hybrid superstructure for postoperative adhesion prevention. Liposomes were prepared using thin-film hydration method. The optimised formulation was characterised by size, SEM, TEM, FTIR, encapsulation efficiency (EE)% (w/w), and release pattern. Liposome-in-hydrogel formulation was investigated by rheology, SEM, and release studies. The efficacy was evaluated in a rat peritoneal abrasion model. EE% (w/w) increased with increasing lipid concentration from 10 to 30; however, a higher percentage of Chol reduced EE% (w/w). The optimised liposome (EE: 68.10 ± 4.18% (w/w), average diameter: 513 ± 14.67 nm, PDI: 0.15 ± 0.04) was used for hydrogel embedding. No sign of adhesion in 5/8 rats and no collagen deposition confirmed the in vivo effectiveness of the optimised formulation. Overall, providing a sustained delivery of PIP, the developed liposome-in-hydrogel formulation can be a promising carrier to prevent postoperative adhesion.
Collapse
Affiliation(s)
- Hanieh Karimi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Shahram Rabbani
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Delaram Babadi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Simin Dadashzadeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Azadeh Haeri
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Protein Technology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Shang LY, Zhou MH, Cao SY, Zhang M, Wang PJ, Zhang S, Meng XX, Yang QM, Gao XL. Effect of polyethylene glycol 400 on the pharmacokinetics and tissue distribution of baicalin by intravenous injection based on the enzyme activity of UGT1A8/1A9. Eur J Pharm Sci 2023; 180:106328. [PMID: 36379359 DOI: 10.1016/j.ejps.2022.106328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 10/11/2022] [Accepted: 11/09/2022] [Indexed: 11/13/2022]
Abstract
Baicalin (BG) is a bioactive flavonoid extracted from the dried root of the medicinal plant, Scutellaria radix (SR) (dicotyledonous family, Labiatae), and has several biological activities. Polyethylene glycol 400 (PEG400) has been used as a suitable solvent for several traditional Chinese medicines (TCM) and is often used as an excipient for the compound preparation of SR. However, the drug-excipient interactions between BG and PEG400 are still unknown. Herein, we evaluated the effect of a single intravenous PEG400 administration on the BG levels of rats using pharmacokinetic and tissue distribution studies. A liver microsome and recombinant enzyme incubation system were used to further confirm the interaction mechanism between PEG400 and UDP-glucuronosyltransferases (UGTs) (UGT1A8 and UGT1A9). The pharmacokinetic study demonstrated that following the co-intravenous administration of PEG400 and BG, the total clearance (CLz) of BG in the rat plasma decreased by 101.60% (p < 0.05), whereas the area under the plasma concentration-time curve (AUC)0-t and AUC0-inf increased by 144.59% (p < 0.05) and 140.05% (p < 0.05), respectively. Additionally, the tissue distribution study showed that the concentration of BG and baicalein-6-O-β-D-glucuronide (B6G) in the tissues increased, whereas baicalein (B) in the tissues decreased, and the total amount of BG and its metabolites in tissues altered following the intravenous administration of PEG400. We further found that PEG400 induced the UGT1A8 and UGT1A9 enzyme activities by affecting the maximum enzymatic velocity (Vmax) and Michaelis-Menten constant (Km) values of UGT1A8 and UGT1A9. In conclusion, our results demonstrated that PEG400 interaction with UGTs altered the pharmacokinetic behaviors and tissue distribution characteristics of BG and its metabolites in rats.
Collapse
Affiliation(s)
- Le-Yuan Shang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Ming-Hao Zhou
- Inspection Center of Guizhou Drug Administration, Guiyang 550025, China
| | - Si-Yuan Cao
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Min Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Peng-Jiao Wang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Shuo Zhang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Xiao-Xia Meng
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Qi-Mei Yang
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China
| | - Xiu-Li Gao
- State Key Laboratory of Functions and Applications of Medicinal Plants and School of Pharmacy, Guizhou Medical University, Guiyang 550025, China; Microbiology and Biochemical Pharmaceutical Engineering Research Center of Guizhou Provincial Department of Education, Guizhou Medical University, Guiyang 550004, China; Guizhou Medical University Experimental Animal Center, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
3
|
Bao M, Ma Y, Liang M, Sun X, Ju X, Yong Y, Liu X. Research progress on pharmacological effects and new dosage forms of baicalin. Vet Med Sci 2022; 8:2773-2784. [DOI: 10.1002/vms3.960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Affiliation(s)
- Minglong Bao
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences Guangdong Ocean University Zhanjiang P. R. China
| | - Yunfei Ma
- College of Veterinary Medicine, China Agricultural University Beijing P. R. China
| | - Mei Liang
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences Guangdong Ocean University Zhanjiang P. R. China
| | - Xinyi Sun
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences Guangdong Ocean University Zhanjiang P. R. China
| | - Xianghong Ju
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences Guangdong Ocean University Zhanjiang P. R. China
| | - Yanhong Yong
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences Guangdong Ocean University Zhanjiang P. R. China
| | - Xiaoxi Liu
- Department of Veterinary Medicine, College of Coastal Agricultural Sciences Guangdong Ocean University Zhanjiang P. R. China
| |
Collapse
|
4
|
Liu Z, Xiang H, Xiang D, Xiao S, Xiang H, Xiao J, Ren H, Hu P, Liu H, Peng M. Revealing potential anti-fibrotic mechanism of Ganxianfang formula based on RNA sequence. Chin Med 2022; 17:23. [PMID: 35180857 PMCID: PMC8855591 DOI: 10.1186/s13020-022-00579-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Accepted: 02/01/2022] [Indexed: 11/18/2022] Open
Abstract
Background Ganxianfang (GXF) formula as a traditional Chinese medicine (TCM) is used for liver fibrosis in clinical practice while its mechanism is unclear. The aim of this study is to explore the molecular mechanism of GXF against CCl4-induced liver fibrosis rats. Methods Detected the main compounds of GXF by UPLC-MS/MS. Evaluated the efficacy of GXF (1.58, 3.15, 4.73 g/kg/day) and Fuzheng Huayu (FZHY, positive control, 0.47 g/kg/day) through serum alanine aminotransferase (ALT), aspartate aminotransferase (AST) levels and histopathological changes. Explored the underlying mechanisms by integrating our total liver RNA sequencing (RNA-seq) data with recent liver single-cell sequencing (scRNA-seq) studies. Verified potential pharmacodynamic substances of GXF by hepatic stellate cell (HSC)-T6 line. Results Main compounds were identified in GXF by UPLC-MS/MS, including baicalin, wogonoside and matrine etc. With GXF-high dose treatment, the elevation of ALT and AST induced by CCl4 were significantly reduced, and the protective effect of GXF-high dose treatment was better than FZHY. Liver histopathological changes were alleviated by GXF-high dose treatment, the ISHAK scoring showed the incidence of liver cirrhosis (F5/F6) decreased from 76.5 to 55.6%. The results of liver hydroxyproline content were consistent with the histopathological changes. RNA-seq analysis revealed the differential genes (DEGs) were mainly enriched in ECM-receptor interaction and chemokine signaling pathway. GXF effectively inhibited collagen deposition and significantly downregulated CCL2 to inhibit the recruitment of macrophages in liver tissue. Integrating scRNA-seq data revealed that GXF effectively inhibited the expansion of scar-associated Trem2+CD9+ macrophages subpopulation and PDGFRα+PDGFRβ+ scar-producing myofibroblasts in the damaged liver, and remodeled the fibrotic niche via regulation of ligand-receptor interactions including TGFβ/EGFR, PDGFB/PDGFRα, and TNFSF12/TNFRSF12a signaling. In vitro experiments demonstrated that baicalin, matrine and hesperidin in GXF inhibited the activation of hepatic stellate cells. Conclusions This study clarified the potential anti-fibrotic effects and molecular mechanism of GXF in CCl4-induced liver fibrosis rats, which deserves further promotion and application. Supplementary Information The online version contains supplementary material available at 10.1186/s13020-022-00579-7.
Collapse
Affiliation(s)
- Zongyi Liu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Huanyu Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Dejuan Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Shuang Xiao
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Hongyan Xiang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Jing Xiao
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Hong Ren
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Peng Hu
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China
| | - Huabao Liu
- Department of Liver Diseases, Chongqing Traditional Chinese Medicine Hospital, Chongqing, 400021, China.
| | - Mingli Peng
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
5
|
Abd-Alla HI, Souguir D, Radwan MO. Genus Sophora: a comprehensive review on secondary chemical metabolites and their biological aspects from past achievements to future perspectives. Arch Pharm Res 2021; 44:903-986. [PMID: 34907492 PMCID: PMC8671057 DOI: 10.1007/s12272-021-01354-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 10/29/2021] [Indexed: 12/13/2022]
Abstract
Sophora is deemed as one of the most remarkable genera of Fabaceae, and the third largest family of flowering plants. The genus Sophora comprises approximately 52 species, 19 varieties, and 7 forms that are widely distributed in Asia and mildly in Africa. Sophora species are recognized to be substantial sources of broad spectrum biopertinent secondary metabolites namely flavonoids, isoflavonoids, chalcones, chromones, pterocarpans, coumarins, benzofuran derivatives, sterols, saponins (mainly triterpene glycosides), oligostilbenes, and mainly alkaloids. Meanwhile, extracts and isolated compounds from Sophora have been identified to possess several health-promising effects including anti-inflammatory, anti-arthritic, antiplatelets, antipyretic, anticancer, antiviral, antimicrobial, antioxidant, anti-osteoporosis, anti-ulcerative colitis, antidiabetic, anti-obesity, antidiarrheal, and insecticidal activities. Herein, the present review aims to provide comprehensive details about the phytochemicals and biological effects of Sophora species. The review spotlighted on the promising phytonutrients extracted from Sophora and their plethora of bioactivities. The review also clarifies the remaining gaps and thus qualifies and supplies a platform for further investigations of these compounds.
Collapse
Affiliation(s)
- Howaida I Abd-Alla
- Chemistry of Natural Compounds Department, National Research Centre, El-Bohouth Street, Giza-Dokki, 12622, Egypt.
| | - Dalila Souguir
- Institut National de Recherches en Génie Rural, Eaux et Forêts (INRGREF), Université de Carthage, 10 Rue Hédi Karray, Manzeh IV, 2080, Ariana, Tunisia
| | - Mohamed O Radwan
- Chemistry of Natural Compounds Department, National Research Centre, El-Bohouth Street, Giza-Dokki, 12622, Egypt.
- Medicinal and Biological Chemistry Science Farm Joint Research Laboratory, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto, 862-0973, Japan.
| |
Collapse
|
6
|
Wang J, Zhu J, Meng J, Qiu T, Wang W, Wang R, Liu J. Baicalin inhibits biofilm formation by influencing primary adhesion and aggregation phases in Staphylococcus saprophyticus. Vet Microbiol 2021; 262:109242. [PMID: 34562786 DOI: 10.1016/j.vetmic.2021.109242] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 09/19/2021] [Indexed: 12/20/2022]
Abstract
The ability to form biofilms on surfaces makes Staphylococcus saprophyticus (S. saprophyticus) becomes the main pathogenic factor in nosocomial infections. Previously, we demonstrated that baicalin (Bac) inhibited azithromycin-resistant S. saprophyticus (ARSS) biofilm formation. This investigation aims to explore the influence of baicalin on primary adhesion and aggregation phases of biofilm formation, and the treatment effect of baicalin and azithromycin on ARSS biofilm-associated infection. Crystal violet (CV) staining and scanning electron microscope (SEM) observations clearly showed that sub-inhibitory concentration baicalin inhibited ARSS biofilm formation when baicalin was added before the adhesion and aggregation phases. Baicalin significantly increased the relative adhesion inhibition rate and decreased the rate of bacteria aggregation in a dose-dependent manner. Moreover, CLSM and cell lysis assays revealed that baicalin inhibited the production of surface proteins and cell autolysis in bacteria adhesion and aggregation phases of biofilm formation. Meanwhile, the relative expressions of adhesion-related and autolysis-related genes were down-regulated by baicalin. In vivo, the combination of baicalin and azithromycin succeeded in eradicating ARSS from the mouse cutaneous infection model and decreasing the pathological injuries, the expressions of cytokines in infected tissue, and the number of inflammatory cells in the blood. Simultaneously, baicalin decreased the bacterial burdens in tubes, the level of TNF-α, and the number of monocytes and neutrophils compared with that of the SS and azithromycin groups. Based on these results, baicalin inhibited the adhesion and aggregation phases of biofilm formation by influenced the production of surface proteins and cell autolysis. Baicalin and azithromycin synergetically treated ARSS biofilm-associated infection.
Collapse
Affiliation(s)
- Jinli Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinyue Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jinwu Meng
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Tianxin Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Wenjia Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Rui Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Jiaguo Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Traditional Chinese Veterinary Medicine Research Center, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
7
|
Abstract
Hinokitiol is a natural bioactive compound found in several aromatic and medicinal plants. It is a terpenoid synthetized and secreted by different species as secondary metabolites. This volatile compound was tested and explored for its different biological properties. In this review, we report the pharmacological properties of hinokitiol by focusing mainly on its anticancer mechanisms. Indeed, it can block cell transformation at different levels by its action on the cell cycle, apoptosis, autophagy via inhibiting gene expression and dysregulating cellular signaling pathways. Moreover, hinokitiol also exhibits other pharmacological properties, including antidiabetic, anti-inflammatory, and antimicrobial effects. It showed multiple and several effects through its inhibition, interaction and/or activation of the main cellular targets inducing these pathologies.
Collapse
|
8
|
Baicalin decreases somatic cell count in mastitis of dairy cows. ANNALS OF ANIMAL SCIENCE 2021. [DOI: 10.2478/aoas-2021-0019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Abstract
Baicalin is a flavonoid that has an influence on molecular processes. It possesses anticancer, anti-inflammatory, antiviral, antioxidative, and antithrombotic properties. It was found that baicalein treatment attenuated the damage of the mammary gland induced by LPS, suppressed the activity of myeloperoxidase, TNFα, and IL-1β in mice with mastitis. The aim of the study was a pilot analysis of baicalin tolerability after intramammary (IMM) administration and its impact on somatic cell count (SCC) after multiple IMM treatment on dairy cows with clinical mastitis. Moreover, the determination of baicalin in milk was performed by the sensitive ultra-high performance liquid chromatography with tandem mass spectrometry. The pharmacokinetic analyses were performed using Phoenix® WinNonlin® 6.4 and ThothPro v 4.1 software. Twelve dairy cows with clinical mastitis were selected for this study. The pharmacodynamic endpoint was SCC level and the clinical investigation was also carried out. Baseline SCC analysis was performed every 24 h among all cows three days before the first dose (B1–B3). After the baseline monitoring, 8 days of treatment (T1–T8) was performed and 8 days within recovery period SCC level was observed (R1–R8). Starting from T1 to T8, a decrease of SCC in relation to baseline was characterized by a declining trend. The presented results confirm the effect of baicalin on the reduction of SCC in mastitis in dairy cows after this therapy. The current study has shown that baicalin accumulation was not confirmed.
Collapse
|
9
|
Su L, Wang R, Qiu T, Wang J, Meng J, Zhu J, Wang D, Wu Y, Liu J. The protective effect of baicalin on duck hepatitis A virus type 1-induced duck hepatic mitochondria dysfunction by activating nuclear erythroid 2-related factor 2/antioxidant responsive element signaling pathway. Poult Sci 2021; 100:101032. [PMID: 33744612 PMCID: PMC8010464 DOI: 10.1016/j.psj.2021.101032] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 01/09/2021] [Accepted: 01/21/2021] [Indexed: 01/21/2023] Open
Abstract
Duck hepatitis A virus type 1 (DHAV-1) is the main pathogen of duck viral hepatitis, but the efficacy of the licensed commercial vaccine needs to be further improved. Therapeutic measures of specific drugs for DHAV-1-infected ducklings need to be urgently developed. Baicalin possesses good antiviral effects. This study aims to investigate the mechanism of baicalin in protecting hepatic mitochondrial function from DHAV-1. The ELISA method was used to detect changes of hepatic and mitochondrial catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPX), inducible nitric oxide synthase (iNOS), adenosine triphosphate (ATP), and malondialdehyde (MDA) levels in vivo and vitro. Hematoxylin and eosin sections and transmission electron microscopy were used to observe liver pathological changes and mitochondrial structural changes. The changes in mitochondrial membrane potential were detected by JC-1 staining method. Western blot and quantitative real-time PCR were employed to analyze the gene and protein expressions in the nuclear erythroid 2-related factor 2 (Nrf2)/antioxidant responsive element (ARE) pathway in duck embryonic hepatocytes infected with DHAV-1. Results showed the administration of baicalin increased the survival rate of ducklings, and alleviated hepatic damage caused by DHAV-1 by enhancing the antioxidant enzyme activities of the liver and mitochondria, including SOD, GPX, CAT, and reducing lipid peroxidative damage (MDA content) and iNOS activities. The mitochondrial ultrastructure changed and the significant increase of ATP content showed that baicalin maintained the structural integrity and ameliorated mitochondrial dysfunction after DHAV-1 infection. In vitro, DHAV-1 infection led to loss of mitochondrial membrane potential and lipid peroxidation and decreased antioxidative enzyme activities (SOD, GPX) and mitochondrial respiratory chain complex activities (succinate dehydrogenase, cytochrome c oxidase). Baicalin relieved the above changes caused by DHAV-1 and activated the gene and protein expressions of Nrf2, which activated ARE-dependent genes including heme oxygenase-1 (HO-1), nicotinamide adenine dinucleotide phosphate quinone oxidoreductase 1 (NQO1), SOD-1, and GPX-1. In addition, baicalin increased the protein expressions of antioxidative enzymes (SOD, GPX). Hence, baicalin protects the liver against oxidative stress in hepatic mitochondria caused by DHAV-1 via activating the Nrf2/ARE signaling pathway.
Collapse
Affiliation(s)
- Linglin Su
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Rui Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Tianxin Qiu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Jinli Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Jinwu Meng
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Jinyue Zhu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Deyun Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Yi Wu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China
| | - Jiaguo Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety and Institute of Traditional Chinese Veterinary Medicine, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing 210095, P R China.
| |
Collapse
|
10
|
The anti-rotavirus effect of baicalin via the gluconeogenesis-related p-JNK-PDK1-AKT-SIK2 signaling pathway. Eur J Pharmacol 2021; 897:173927. [PMID: 33567320 DOI: 10.1016/j.ejphar.2021.173927] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 01/24/2021] [Accepted: 01/29/2021] [Indexed: 02/08/2023]
Abstract
Rotavirus (RV) infection is a leading cause of severe, dehydrating gastroenteritis in children < 5 years of age, and by now, the prevention and treatment of RV are still the major public health problems due to a lack of specific clinical drugs. Thus, the aims of this study are to explore the anti-RV effect of baicalin and its influence on glucose metabolism. Here, we demonstrated for the first time that baicalin had an anti-RV attachment effect with the strongest effect at a concentration of 100 μM, and also inhibited the replication of RV at concentrations of 100, 125, 150, 175, and 200 μM. Moreover, baicalin helped to overcome the weight loss and reduced the diarrhea rate and score with the best therapeutic effect at a concentration of 0.3 mg/g in RV-infected neonatal mice. Interestingly, baicalin decreased glucose consumption in RV-infected Caco-2 cells with the optimal concentration of 125 μM. Next, metabolomic analysis indicated that there were 68 differentially expressed metabolites, including an increase in pyruvic acid, asparagine, histidine and serine, and a decrease in dihydroxyacetone phosphate, which suggested that the underlying signaling pathway was gluconeogenesis. Further studies demonstrated that baicalin inhibited gluconeogenesis via improving glucose 6-phosphatase (G-6-Pase) and phosphoenolpyruvate carboxylase (PEPCK). Moreover, baicalin upregulated the potential gluconeogenesis proteins named salt inducible kinase 2, pyruvate dehydrogenase kinase 1, AKT serine/threonine kinase 1 and down-regulated phosphorylated c-Jun NH2-terminal kinase, which are associated with G-6-Pase and PEPCK expressions. Therefore, baicalin improved the gluconeogenesis disruption caused by RV.
Collapse
|
11
|
Chen H, Feng R, Muhammad I, Abbas G, Zhang Y, Ren Y, Huang X, Zhang R, Diao L, Wang X, Li G. Protective effects of hypericin against infectious bronchitis virus induced apoptosis and reactive oxygen species in chicken embryo kidney cells. Poult Sci 2020; 98:6367-6377. [PMID: 31399732 PMCID: PMC7107269 DOI: 10.3382/ps/pez465] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/26/2019] [Indexed: 12/17/2022] Open
Abstract
Avian infectious bronchitis virus (IBV), a coronavirus, causes infectious bronchitis leading to enormous economic loss in the poultry industry worldwide. Hypericin (HY) is an excellent compound that has been investigated in antiviral, antineoplastic, and antidepressant. To investigate the inhibition effect of HY on IBV infection in chicken embryo kidney (CEK) cells, 3 different experimental designs: pre-treatment of cells prior to IBV infection, direct treatment of IBV-infected cells, and pre-treatment of IBV prior to cell infection were used. Quantitative real-time PCR (qRT-PCR), immunofluorescence assay (IFA), flow cytometry, and fluorescence microscopy were performed and virus titer was determined by TCID50. The results revealed that HY had a good anti-IBV effect when HY directly treated the IBV-infected cells, and virus infectivity decreased in a dose-dependent manner. Furthermore, HY inhibited IBV-induced apoptosis in CEK cells, and significantly reduced the mRNA expression levels of Fas, FasL, JNK, Bax, Caspase 3, and Caspase 8, and significantly increased Bcl-2 mRNA expression level in CEK cells. In addition, HY treatment could decrease IBV-induced reactive oxygen species (ROS) generation in CEK cells. These results suggested that HY showed potential antiviral activities against IBV infection involving the inhibition of apoptosis and ROS generation in CEK cells.
Collapse
Affiliation(s)
- Huijie Chen
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China.,College of Biological and Pharmaceutical Engineering, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Rui Feng
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ishfaq Muhammad
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ghulam Abbas
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yue Zhang
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Yudong Ren
- College of Electrical and Information, Northeast Agricultural University, Harbin 150030, China
| | - Xiaodan Huang
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Ruili Zhang
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| | - Lei Diao
- College of Biological and Pharmaceutical Engineering, Jilin Agriculture Science and Technology College, Jilin 132101, China
| | - Xiurong Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150001, China
| | - Guangxing Li
- Key Laboratory for Laboratory Animals and Comparative Medicine of Heilongjiang Province, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, China
| |
Collapse
|
12
|
Lv W, Liu C, Zeng Y, Li Y, Chen W, Shi D, Guo S. Explore the potential effect of natural herbals to resist Newcastle Disease Virus. Poult Sci 2019; 98:1993-1999. [PMID: 30566670 DOI: 10.3382/ps/pey557] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/21/2018] [Indexed: 01/10/2023] Open
Abstract
In this study, we examined the dose-dependent effects of the formula on Newcastle disease virus (NDV). In in-vitro test, the formula within safety concentration scope and NDV were added into cultured chick embryo fibroblast in 3 modes, and the cellular A570 values were determined by MTT (3-(4, 5-dimethyithiazol-2-yl)-2, 5-diphenyltetrazolium bromide) method. In in-vivo test, we examined the expression of interferon-induced transmembrane protein 3 (IFITM3) and Interferons (IFNs) in NDV-infected chickens. The results showed that the highest virus inhibitory rates of the formula at optimal concentration group were the highest (15.625 mg/mL) in post-adding and simultaneous-adding drug and virus modes, whereas medium concentration (7.813 mg/mL) showed the highest virus inhibitory rates in pre-adding drug mode. In vivo, the formula significantly upregulated the expression of IFITM3 in NDV-infected chickens at 3-D post-infection. However, the levels of IFNs were significantly downregulated. On days 5 and 7 post-infection, the levels of IFNs quickly upregulated. Moreover, the formula can significantly upregulate the antibody to resist the NDV compared with model control group on days 5 and 7 post-infection. In animals treated with the formula, the survival rate was nearly 37% higher at 7 d post-infection. We also found that the formula had a significantly stronger effect than a single herb on upregulating the expression of IFITM3. It confirmed that the formula could significantly inhibit the infectivity of NDV to chick embryo fibroblast. Also, the formula could significantly upregulated IFITM3 expression and inhibited virus replication in NDV-infected chickens. During the early stage of infection, IFNs were consumed to stimulate IFITM3 to inhibit virus replication, whereas during later stages of the infection, the formula upregulated the levels of IFNs and their antibodies to maintain a high level of immunity.
Collapse
Affiliation(s)
- Weijie Lv
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Cui Liu
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Ying Zeng
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Yuefei Li
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Wenqian Chen
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Dayou Shi
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| | - Shining Guo
- College of Veterinary Medicine, South China Agricultural University, Guangzhou 510642, PR China
| |
Collapse
|
13
|
Wang J, Qiao M, Zhou Y, Du H, Bai J, Yuan W, Liu J, Wang D, Hu Y, Wu Y. In vitro synergistic effect of baicalin with azithromycin against Staphylococcus saprophyticus isolated from francolins with ophthalmia. Poult Sci 2019; 98:373-380. [DOI: 10.3382/ps/pey356] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Accepted: 07/14/2018] [Indexed: 11/20/2022] Open
|