1
|
Mu Y, Liu J, Wu Q, Wang B, Hu T, Li Y, Yan X, Ma L, Tan Z. A dual αvβ1/αvβ6 integrin inhibitor Bexotegrast (PLN-74809) ameliorates organ injury and fibrogenesis in fibrotic kidney disease. Eur J Pharmacol 2024; 983:176983. [PMID: 39243926 DOI: 10.1016/j.ejphar.2024.176983] [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: 06/24/2024] [Revised: 09/05/2024] [Accepted: 09/05/2024] [Indexed: 09/09/2024]
Abstract
Chronic kidney disease (CKD) is a global public health problem, involving about 10% of the global population. Unfortunately, there are currently no effective drugs. Kidney fibrosis is the main pathology of CKD, where integrins play crucial roles in renal fibrogenesis. Recently, Bexotegrast (PLN-74809) as a dual integrin αvβ1/αvβ6 inhibitor could reduce the degree of lung fibrosis in patients with idiopathic pulmonary fibrosis. However, the role of PLN-74809 remains unclear in fibrotic kidney disease. Here, we have revealed that PLN-74809 administration dose-dependently delayed the progression of renal fibrosis in both adenine diet- and unilateral ureteral obstruction (UUO)-induced mice. Mechanistically, PLN-74809 targeted integrin αvβ1/αvβ6 to inhibit FAK/Src/Akt/β-catenin cascade in fibrotic kidneys. In summary, our results for the first time highlighted the αvβ1/αvβ6 inhibitor PLN-74809 exerted potential therapeutic against kidney fibrosis.
Collapse
Affiliation(s)
- Yingsong Mu
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China
| | - Jing Liu
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Qimei Wu
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China
| | - Bo Wang
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - TingTing Hu
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China
| | - Yiman Li
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China
| | - Xiaoyong Yan
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China.
| | - Liang Ma
- Department of Nephrology, Institute of Kidney Diseases, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Zhouke Tan
- Department of Nephrology, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China; Organ Transplant Center, Affiliated Hospital of ZunYi Medical University, ZunYi, 563000, China; Guizhou Province Key Laboratory of Cell Engineering, Affiliated Hospital of ZunYi Medical University, ZunYi, 563003, China.
| |
Collapse
|
2
|
Liu DD, Liu XL, Zheng TF, Li X, Zhao YC, Pan JC, Yuan C, Wang QQ, Zhang M. Dapagliflozin alleviates right heart failure by promoting collagen degradation by reducing ROS levels. Eur J Pharmacol 2024; 981:176875. [PMID: 39121982 DOI: 10.1016/j.ejphar.2024.176875] [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: 05/14/2024] [Revised: 08/05/2024] [Accepted: 08/07/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND Right ventricular (RV) fibrosis is an important pathological change that occurs during the development of right heart failure (RHF) induced by pulmonary hypertension (PH). Dapagliflozin (DAPA), a sodium-glucose cotransporter 2 (SGLT2) inhibitor, has been shown to play a major role in left heart failure, but it is unclear whether it has a positive effect on RHF. This study aimed to clarify the effect of DAPA on PH-induced RHF and investigate the underlying mechanisms. METHODS We conducted experiments on two rat models with PH-induced RHF and cardiac fibroblasts (CFs) exposed to pathological mechanical stretch or transforming growth factor-beta (TGF-β) to investigate the effect of DAPA. RESULTS In vivo, DAPA could improve pulmonary hemodynamics and RV function. It also attenuated right heart hypertrophy and RV fibrosis. In vitro, DAPA reduced collagen expression by increasing the production of matrix metalloproteinase 2 (MMP2) and matrix metalloproteinase 9 (MMP9). Additionally, DAPA was found to reduce reactive oxygen species (ROS) levels in CFs and the right heart in rats. Similar to DAPA, the ROS scavenger N-acetylcysteine (NAC) exerted antifibrotic effects on CFs. Therefore, we further investigated the mechanism by which DAPA promoted collagen degradation by reducing ROS levels. CONCLUSIONS In summary, we concluded that DAPA ameliorated PH-induced structural and functional changes in the right heart by increasing collagen degradation. Our study provides new ideas for the possibility of using DAPA to treat RHF.
Collapse
Affiliation(s)
- Dong-Dong Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao-Lin Liu
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Teng-Fei Zheng
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Li
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ya-Chao Zhao
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Ji-Chen Pan
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Chong Yuan
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China
| | - Qian-Qian Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, No.1677 Wutai Mountain Road, Qingdao, 266000, China.
| | - Mei Zhang
- National Key Laboratory for Innovation and Transformation of Luobing Theory, The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education, Chinese National Health Commission and Chinese Academy of Medical Sciences, Department of Cardiology, Qilu Hospital of Shandong University, Jinan, China.
| |
Collapse
|
3
|
Ewing LE, Harpenau RJ, Skinner CM, Clement K, Quick CM, Yee EU, Williams DK, Walker LA, ElSohly MA, Gurley BJ, Koturbash I. Inter-strain variability in responses to a single administration of the cannabidiol-rich cannabis extract in mice. Food Chem Toxicol 2024; 192:114909. [PMID: 39128689 PMCID: PMC11381146 DOI: 10.1016/j.fct.2024.114909] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 07/30/2024] [Accepted: 08/05/2024] [Indexed: 08/13/2024]
Abstract
Cannabidiol (CBD) has gained widespread popularity; however, its pharmacological and toxicological profiles in the context of human genetic diversity remain largely unexplored. Here, we investigated the variability in metabolism and toxicity of CBD-rich cannabis extract (CRCE) in genetically diverse mouse models: C57BL/6J, B6C3F1/J, and NZO/HlLtJ strains. Mice received a single dose of CRCE containing 57.9% CBD at dosages of 0, 246, 738, and 2460 mg/kg of CBD. At 24 h after treatment, no appreciable histomorphological changes were detected in the liver. Plasma bilirubin levels increased markedly in all strains at the highest CBD dose. Mice in all treatment groups displayed significant but distinct increases in ALT and AST levels. While B6C3F1/J and NZO/HlLtJ mice had negligible plasma CBD levels at 738 mg/kg, C57BL/6J mice exhibited levels exceeding 7000 ng/mL. At 2460 mg/kg, high CBD concentrations were found in B6C3F1/J and C57BL/6J mice, but markedly lower levels were seen in NZO/HlLtJ mice. Gene expression profiling showed significant increases in Cyp2b10 across all strains but varying responses in Cyp1a1 expression, indicating strain-specific CYP dysregulation. Genetically diverse mice exhibited differential pharmacological and toxicological responses to CRCE, suggesting a high potential for inter-individual variability in the pharmacology and toxicology of CBD in humans.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Ryan J Harpenau
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Charles M Skinner
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Arkansas Children's Research Institute, Little Rock, AR, USA
| | - Kirsten Clement
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Charles M Quick
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Eric U Yee
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - D Keith Williams
- Department of Biostatistics, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA
| | - Larry A Walker
- National Center for Natural Products Research, University of Mississippi University, MS, 38677, USA
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, University of Mississippi University, MS, 38677, USA; ElSohly Laboratories, Inc. (ELI), Oxford, MS, 38677, USA
| | - Bill J Gurley
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; National Center for Natural Products Research, University of Mississippi University, MS, 38677, USA
| | - Igor Koturbash
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
4
|
Tian L, Yang M, Tu S, Chang K, Jiang H, Jiang Y, Ding L, Weng Z, Wang Y, Tan X, Zong C, Chen B, Dou X, Wang X, Qi X. Xiaoke Bitong capsule alleviates inflammatory impairment via inhibition of the TNF signaling pathway to against diabetic peripheral neuropathy. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 132:155867. [PMID: 39047415 DOI: 10.1016/j.phymed.2024.155867] [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: 02/23/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Xiaoke Bitong capsule (XBC) is a crude herbal compound believed to tonify qi, improve blood circulation, and alleviate blood stasis. It has been used as an herbal formula for the prevention and treatment of diabetic peripheral neuropathy (DPN) under the guidance of traditional Chinese medicine (TCM). However, the pharmacological mechanisms by which XBC ameliorates DPN remain poorly understood. The interaction between pro-inflammatory factors and the activation of tumor necrosis factor (TNF) plays a critical role in the underlying mechanisms of DPN. XBC may protect against DPN through the regulation of the TNF pathway. PURPOSE Many studies show the association between DPN and nerve dysfunction, however, treatment options are limited. To identify specific therapeutic targets and active components of XBC that contribute to its anti-DPN effects, our study aimed to investigate the potential mechanism of action of XBC during the progression of DPN using a system pharmacology approach. METHODS An approach involving UPLC-Q-TOF/MS and network pharmacology was used to analyze the compositions, potential targets, and active pathways of XBC. Further, models of streptozocin (STZ) induced mouse and glucose induced RSC96 cells were established to explore the therapeutic effects of XBC. High glucose induced RSC96 cells were pretreated with small interfering RNA (siRNA) to identify potential therapeutic targets of DPN. RESULTS Seventy-one active compositions of XBC and five potential targets, including mitogen-activated protein kinase 8 (MAPK), interleukin-6 (IL-6), poly-ADP-ribose polymerase-1 (PARP1), vascular endothelial growth factor A (VEGFA), and transcription factor p65 (NF-κB), were considered as the potential regulators of DPN. In addition, the results revealed that the TNF signaling pathway was closely related to DPN. Moreover, DPN contributed to the decreased expressions of PI3K and AKT, increased TNF-α and IL-1β in RSC96 cells, which were both reversed by XBC or TNF-α siRNA. CONCLUSION XBC could protect against DPN by inhibiting the release of pro-inflammatory cytokines and regulating the activation of the TNF signaling pathway, further accelerating neurogenesis, and alleviating peripheral nerve lesions. Therefore, this study highlights the therapeutic value of XBC for DPN.
Collapse
Affiliation(s)
- Lulu Tian
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Meiqi Yang
- Liaoning University of Traditional Chinese Medicine Xinglin College, Shenyang, Liaoning, PR China
| | - Shanjie Tu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Kaixin Chang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Huanyu Jiang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yuwei Jiang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Lu Ding
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, PR China
| | - Zhiwei Weng
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Yueqiang Wang
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xiaolong Tan
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Chunxiao Zong
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Buyang Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China
| | - Xiaobing Dou
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, PR China.
| | - Xiuge Wang
- College of Chinese Medicine, Changchun University of Chinese Medicine, Changchun, Jilin, PR China.
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Neurosurgery, Shaoxing People's Hospital, Shaoxing, Zhejiang, PR China.
| |
Collapse
|
5
|
Ewing LE, Skinner CM, McGill MR, Kennon-McGill S, Clement K, Quick CM, Yee EU, Williams DK, Walker LA, ElSohly MA, Gurley BJ, Koturbash I. Human quad liver-on-chip system as a tool toward bridging the gap between animals and humans regarding toxicology and pharmacology of a cannabidiol-rich cannabis extract. Drug Chem Toxicol 2024:1-8. [PMID: 39155655 DOI: 10.1080/01480545.2024.2388292] [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: 03/29/2024] [Revised: 07/11/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
Cannabidiol (CBD) is a major phytocannabinoid from Cannabis sativa. It is currently widely available and widely used in the USA, but despite its rapid progress to market, the pharmacology and toxicology of both CBD and cannabidiol-rich cannabis extracts (CRCE) remain largely unknown. The goals of this study were to investigate the potential of a novel human microphysiological system to emulate CRCE-induced hepatotoxicity and pharmacological properties demonstrated in animal models. For this purpose, C57BL6/J male mice were subjected to dosing with either 0, 61.5, 184.5, or 615 mg/kg of CRCE for 10 days. The liver-on-chip system, incorporating human primary hepatocytes, sinusoidal endothelial cells, as well as Kupffer and stellate cells was subjected to 0, 300, 1,200, or 4,400 ng/mL of CRCE (8 h exposure followed by 16 h washout) for 5 days. Administration of CRCE in mice resulted in nearly 4-fold elevations of plasma ALT at 615 mg/kg (p < 0.01) and a dose-dependent decrease in intrahepatic miR-122. Elevated levels of ALT, paralleled by decreased intrahepatic and increased effluent levels of miR-122, were also observed in the liver-on-chip, although these results were not statistically significant. Exposure to CRCE resulted in a robust and dose-dependent induction of key cytochrome P450 enzymes, namely Cyp1a2, Cyp2b6 (CYP2B10), Cyp2e1, and Cyp2c9 (CYP2C19) in both mouse livers and liver-on-chip. The results of this study demonstrate the congruence between the responses observed in mouse and human liver-on-chip experimental systems and provide evidence of the potential microphysiological systems hold for translating animal data into clinical practice.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charles M Skinner
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mitchell R McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Department of Pharmacology and Toxicology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Stefanie Kennon-McGill
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Kirsten Clement
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charles M Quick
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eric U Yee
- Department of Pathology, College of Medicine, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - D Keith Williams
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Larry A Walker
- National Center for Natural Products Research, University of Mississippi, University, MS, USA
- ElSohly Laboratories, Inc. (ELI), Oxford, MS, USA
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, University of Mississippi, University, MS, USA
- ElSohly Laboratories, Inc. (ELI), Oxford, MS, USA
| | - Bill J Gurley
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Arkansas Children's Nutrition Center, Little Rock, AR, USA
| | - Igor Koturbash
- Department of Environmental Health Sciences, Fay W. Boozman College of Public Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
6
|
Sun N, Zhang Y, Ding L, An X, Bai F, Yang Y, Yu K, Fan J, Liu L, Yang H, Yang X. Blockade of aryl hydrocarbon receptor restricts omeprazole-induced chronic kidney disease. J Mol Med (Berl) 2024; 102:679-692. [PMID: 38453697 DOI: 10.1007/s00109-024-02429-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 12/02/2023] [Accepted: 02/06/2024] [Indexed: 03/09/2024]
Abstract
Chronic kidney disease (CKD) is the 16th leading cause of mortality worldwide. Clinical studies have raised that long-term use of omeprazole (OME) is associated with the morbidity of CKD. OME is commonly used in clinical practice to treat peptic ulcers and gastroesophageal reflux disease. However, the mechanism underlying renal failure following OME treatment remains mostly unknown and the rodent model of OME-induced CKD is yet to be established. We described the process of renal injury after exposure to OME in mice; the early renal injury markers were increased in renal tubular epithelial cells (RTECs). And after long-term OME treatment, the OME-induced CKD mice model was established. Herein, aryl hydrocarbon receptor (AHR) translocation appeared after exposure to OME in HK-2 cells. Then for both in vivo and in vitro, we found that Ahr-knockout (KO) and AHR small interfering RNA (siRNA) substantially alleviated the OME-induced renal function impairment and tubular cell damage. Furthermore, our data demonstrate that antagonists of AHR and CYP1A1 could attenuate OME-induced tubular cell impairment in HK-2 cells. Taken together, these data indicate that OME induces CKD through the activation of the AHR-CYP axis in RTECs. Our findings suggest that blocking the AHR-CYP1A1 pathway acts as a potential strategy for the treatment of CKD caused by OME. KEY MESSAGES: We provide an omeprazole-induced chronic kidney disease (CKD) mice model. AHR activation and translocation process was involved in renal tubular damage and promoted the occurrence of CKD. The process of omeprazole nephrotoxicity can be ameliorated by blockade of the AHR-CYP1A1 axis.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Basic Helix-Loop-Helix Transcription Factors/metabolism
- Basic Helix-Loop-Helix Transcription Factors/genetics
- Cell Line
- Cytochrome P-450 CYP1A1/metabolism
- Cytochrome P-450 CYP1A1/genetics
- Disease Models, Animal
- Epithelial Cells/metabolism
- Epithelial Cells/drug effects
- Kidney Tubules/pathology
- Kidney Tubules/metabolism
- Kidney Tubules/drug effects
- Mice, Inbred C57BL
- Mice, Knockout
- Omeprazole/pharmacology
- Receptors, Aryl Hydrocarbon/metabolism
- Receptors, Aryl Hydrocarbon/genetics
- Renal Insufficiency, Chronic/metabolism
- Renal Insufficiency, Chronic/drug therapy
- Renal Insufficiency, Chronic/etiology
- Renal Insufficiency, Chronic/chemically induced
- RNA, Small Interfering/metabolism
- RNA, Small Interfering/genetics
Collapse
Affiliation(s)
- Nan Sun
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yimeng Zhang
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lin Ding
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xin An
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Fang Bai
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yanjiang Yang
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Kuipeng Yu
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jiahui Fan
- Laboratory of Basic Medical Sciences, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Lei Liu
- Key Laboratory for Experimental Teratology of Ministry of Education and Department of Immunology, Shandong University School of Basic Medical Science, Jinan, Shandong, China
| | - Huimin Yang
- Department of General Practice, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Xiangdong Yang
- Department of Nephrology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
| |
Collapse
|
7
|
Xue Y, Zhang YN, Wang M, Fu HY, Mao YC, Hu M, Sun MT, Guo HG, Cao L, Feng CZ. Prolonged oral intake of green tea polyphenols attenuates delirium-like behaviors in mice induced by anesthesia/surgery. Heliyon 2024; 10:e26200. [PMID: 38495146 PMCID: PMC10943306 DOI: 10.1016/j.heliyon.2024.e26200] [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: 06/15/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 03/19/2024] Open
Abstract
Postoperative delirium (POD) is a severe postoperative complication characterized by delirium-like symptoms. So far, no effective preventable strategy for POD prevention has been identified. Reports show that the consumption of green tea polyphenols (GTP) is associated with better cognitive function by modulating the composition of gut microbiota. Whether GTP also play a role in alleviating POD through gut microbiota is unknown. Herein, we studied the effect of prolonged (eight weeks) GTP intake on postoperative delirium in C57BL/6 mice with laparotomies under isoflurane anesthesia (anesthesia/surgery). We subsequently investigated anesthesia/surgery caused behavioral changes and increased the expression of malondialdehyde (MAD), an oxidative stress marker, and the activities of superoxide dismutase (SOD), an antioxidant marker, in the mice at 6 h after anesthesia/surgery. However, GTP administration reversed these changes and alleviated anesthesia/surgery-induced decrease in the abundance of gut bacterial genera, Roseburia. Further, fecal microbiota transplant demonstrated that compared with mice in the control group, treatment of C57BL/6 mice with feces from GTP-treated mice had a slight effect on the behavioral changes of mice. These data suggest that daily consumption of GTP could protect against anesthesia/surgery-induced behavioral changes, which is closely associated with gut microbiota modification by GTP.
Collapse
Affiliation(s)
- Yao Xue
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Yan-Na Zhang
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Man Wang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Hui-Yuan Fu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Ying-Chao Mao
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Min Hu
- School of Pharmacy, Hangzhou Medical College, Hangzhou, China
| | - Mei-Tao Sun
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| | - Hong-Gang Guo
- Center of Animal Research, Hangzhou Medical College, Hangzhou, China
| | - Lin Cao
- Department of Anesthesiology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chen-Zhuo Feng
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
8
|
Sun D, Xing D, Wang D, Liu Y, Cai B, Deng W, Hu Q, Ma W, Jin B. The Protective Effects of Bushen Daozhuo Granule on Chronic Non-bacterial Prostatitis. Front Pharmacol 2024; 14:1281002. [PMID: 38239203 PMCID: PMC10794918 DOI: 10.3389/fphar.2023.1281002] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 11/27/2023] [Indexed: 01/22/2024] Open
Abstract
Background: Chronic non-bacterial prostatitis (CNP), one of the most common chronic diseases in urology, leads to pain in the prostate and dysuria, critically affecting the physical or mental health of patients. However, there are no standard treatment approaches for the treatment of CNP in the clinic. Although the clinical application of Bushen Daozhuo granule (BSDZG) offers hope to CNP patients in China, the mechanisms of BSDZG in treating CNP are still not entirely clear. Hence, we aimed to investigate the novel therapeutic mechanisms of BSDZG on CNP. Methods: In this study, we first assayed the prostate index of rats and then determined the anti-inflammatory and anti-apoptotic effects of BSDZG on CNP in vivo and in vitro by employing ELISA kits and TUNEL staining. Next, we investigated whether the anti-inflammatory and anti-apoptotic mechanisms of BSDZG on prostate protein-induced rats and lipopolysaccharide (LPS) induced RWPE-1 cells were related to the AKT, p38 MAPK, and NF-κB pathways with the help of Western blot. Finally, the influence of BSDZG on the interaction between the p38 MAPK and NF-κB pathway in LPS-induced RWPE-1 cells was explored by adopting dehydrocorydaline (DHC, p38 MAPK activator) with the help of ELISA kits and Western blot. Results: In vivo, BSDZG effectively reduced the prostate index. In vivo and in vitro, BSDZG dramatically declined the level of two pro-inflammatory cytokines, TNF-α and IL-1β, as well as the apoptosis rate. Moreover, in vivo and in vitro, BSDZG memorably upregulated the expression level of p-AKT, and substantially downregulated the expression level of p-p38 MAPK and NF-κB2. The activation of p38 MAPK significantly reversed the moderation effects of BSDZG on the level of TNF-α and IL-1β, as well as the expression level of p-p38 MAPK and NF-κB2 in vitro. Conclusion: To sum up, the in vivo and in vitro therapeutic mechanisms of BSDZG on CNP were reflected as the anti-inflammation and anti-apoptosis that was formed by inhibiting the level of pro-inflammatory cytokines, TNF-α and IL-1β, to regulate the AKT, p38 MAPK, and NF-κB pathways, and the anti-inflammatory effect of BSDZG was realized by suppressing the p38 MAPK pathway to inhibit the downstream NF-κB pathway.
Collapse
Affiliation(s)
- Dalin Sun
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Dong Xing
- School of Medicine, Southeast University, Nanjing, China
| | - Dandan Wang
- School of Medicine, Southeast University, Nanjing, China
| | - Yuanyuan Liu
- School of Medicine, Southeast University, Nanjing, China
| | - Bin Cai
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Weimin Deng
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Qinglin Hu
- Department of Urology, Chuzhou Integrated Hospital of Chinese and Western Medicine, Affiliated to Anhui University of Traditional Chinese Medicine, Chuzhou, China
| | - Wenjun Ma
- Department of Urology, Chuzhou Integrated Hospital of Chinese and Western Medicine, Affiliated to Anhui University of Traditional Chinese Medicine, Chuzhou, China
| | - Baofang Jin
- Andrology Department of Integrative Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
9
|
Wu F, Cui M, Wang S, Yu C, Yin W, Li J, Yan X. Effect of berberine on pharmacokinetics and pharmacodynamics of atorvastatin in hyperlipidemia rats. Xenobiotica 2023; 53:644-652. [PMID: 38054840 DOI: 10.1080/00498254.2023.2290648] [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: 10/05/2023] [Accepted: 11/29/2023] [Indexed: 12/07/2023]
Abstract
Atorvastatin, an effective lipid-lowering drug, could reduce the risks of morbidity and mortality of cardiovascular diseases. Patients with cardiovascular diseases often use atorvastatin along with berberine. Atorvastatin is the substrate of CYP3A4 and P-gp. However, berberine is the inhibitor. The combination might lead to DDIs. The aim of this study was to assess the effect of berberine on pharmacokinetics and pharmacodynamics of atorvastatin in rats.Plasma concentrations of atorvastatin with or without berberine were determined by HPLC. Pharmacokinetics parameters were calculated and used to evaluate pharmacokinetics interactions. The effect of berberine on pharmacodynamics of atorvastatin was investigated by detecting blood lipid, SOD, MDA, GSH-Px, AST, ALT, and liver histopathology.Cmax, tmax, and AUC0-t of atorvastatin in combination group significantly increased both in normal and model rats (p < 0.01). The increase of t1/2, AUC0-t in model rats was more significant than that in normal rats (p < 0.05). Pharmacodynamics indexes in treatment groups were significantly improved, especially combination group (p < 0.05). Moreover, it could be found that there is a significant recovery in liver histopathology.In conclusion, berberine could affect pharmacokinetics of atorvastatin, enhance lipid-lowering effect and improve liver injury in rats. More attention should be paid to plasma exposure in clinical to avoid adverse reactions.
Collapse
Affiliation(s)
- Fan Wu
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Mingyu Cui
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Siwen Wang
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Chao Yu
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Weihong Yin
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Jiao Li
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| | - Xueying Yan
- Department of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, China
| |
Collapse
|
10
|
Zhang X, Hou X, Xu C, Cheng S, Ni X, Shi Y, Yao Y, Chen L, Hu MG, Xia D. Kaempferol regulates the thermogenic function of adipocytes in high-fat-diet-induced obesity via the CDK6/RUNX1/UCP1 signaling pathway. Food Funct 2023; 14:8201-8216. [PMID: 37551935 DOI: 10.1039/d3fo00613a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/09/2023]
Abstract
Activation of adipose tissue thermogenesis is a promising strategy in the treatment of obesity and obesity-related metabolic disorders. Kaempferol (KPF) is a predominant dietary flavonoid with multiple pharmacological properties, such as anti-inflammatory and antioxidant activities. In this study, we sought to characterize the role of KPF in adipocyte thermogenesis. We demonstrated that KPF-treated mice were protected from diet-induced obesity, glucose tolerance, and insulin resistance, accompanied by markedly increased energy expenditure, ex vivo oxygen consumption of white fat, and increased expression of proteins related to adaptive thermogenesis. KPF-promoted beige cell formation is a cell-autonomous effect, since the overexpression of cyclin-dependent kinase 6 (CDK6) in preadipocytes partially reversed browning phenotypes observed in KPF-treated cells. Overall, these data implicate that KPF is involved in promoting beige cell formation by suppressing CDK6 protein expression. This study provides evidence that KPF is a promising natural product for obesity treatment by boosting energy expenditure.
Collapse
Affiliation(s)
- Xiaoxi Zhang
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xiaoli Hou
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
- Academy of Chinese Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Changyu Xu
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Siyao Cheng
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Xintao Ni
- School of Life Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Yueyue Shi
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Yanjing Yao
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Liangxin Chen
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| | - Miaofen G Hu
- Department of Medicine, Division of Hematology Oncology, Tufts Medical Center, Boston, MA, 02111, USA.
| | - Daozong Xia
- Department of Food Science and Nutrition, School of Pharmaceutical Sciences, Zhejiang Chinese Medical University, Hangzhou, 310053, China.
| |
Collapse
|
11
|
Souza MR, Brito ECB, Furtado LS, Barco VS, Cruz LLD, Moraes-Souza RQ, Monteiro GC, Lima GPP, Damasceno DC, Volpato GT. Maternal-fetal toxicity of Strychnos pseudoquina extract treatment during pregnancy. JOURNAL OF ETHNOPHARMACOLOGY 2023; 311:116459. [PMID: 37023837 DOI: 10.1016/j.jep.2023.116459] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 03/31/2023] [Accepted: 04/03/2023] [Indexed: 06/19/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Plants and herbs have been used by women throughout history for therapeutic purposes. Strychnos pseudoquina, a plant used in the treatment of various diseases, can also function as an abortive herb. There is no scientific confirmation of its effects during pregnancy, and the activity of this plant needs to be substantiated or refuted with experimental evidence. AIM OF THE STUDY Evaluating the effect of the S. pseudoquina aqueous extract on maternal reproductive toxicity and fetal development. MATERIALS AND METHODS The aqueous extract of S. pseudoquina bark was evaluated in Wistar rats. Pregnant rats were distributed into four experimental groups (n = 12 rats/group): Control = treated with water (vehicle); Treated 75, Treated 150, and Treated 300 = treated with S. pseudoquina at dose 75, 150 and 300 mg/kg, respectively. The rats were treated by an intragastric route (gavage) from day 0 to day 21 of pregnancy. At the end of pregnancy, maternal reproductive outcomes, organs, biochemical and hematological profiles, fetuses, and placentas were analyzed. Maternal toxicity was evaluated through body weight gain, water, and food intake. With knowledge of the harmful dosage of the plant, other rats were used on gestational day 4 for the evaluation of morphological analyses before embryo implantation. P < 0.05 was considered as statistically significant. RESULTS The S. pseudoquina treatment showed elevated liver enzymatic activities. The Treated 300 group presented toxicity with reduced maternal body weight, water and food intake, and increased kidney relative weight compared to those of the Control group. At a high dosage, the plant presents an abortifacient activity, confirmed by embryo losses before and after implantation and degenerated blastocysts. In addition, the treatment contributed to an increased percentage of fetal visceral anomalies, decreased ossification sites, and intrauterine growth restriction (300 mg/kg dose). CONCLUSION In general, our study showed that an aqueous extract of S. pseudoquina bark caused significant abortifacient activity that testified to its traditional use. Furthermore, the S. pseudoquina extract caused maternal toxicity that contributed to impaired embryofetal development. Therefore, the use of this plant should be completely avoided during pregnancy to prevent unintended abortion and risks to maternal-fetal health.
Collapse
Affiliation(s)
- Maysa Rocha Souza
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso - Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista (Unesp), Botucatu Medical School, Botucatu, São Paulo State, Brazil
| | - Evelyn Caroline Barbosa Brito
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso - Barra do Garças, Mato Grosso State, Brazil
| | - Linne Stephane Furtado
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso - Barra do Garças, Mato Grosso State, Brazil
| | - Vinícius Soares Barco
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista (Unesp), Botucatu Medical School, Botucatu, São Paulo State, Brazil
| | - Larissa Lopes da Cruz
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso - Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista (Unesp), Botucatu Medical School, Botucatu, São Paulo State, Brazil
| | - Rafaianne Queiroz Moraes-Souza
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso - Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista (Unesp), Botucatu Medical School, Botucatu, São Paulo State, Brazil
| | - Gean Charles Monteiro
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University, Botucatu, São Paulo State, Brazil
| | - Giuseppina Pace Pereira Lima
- Department of Chemical and Biological Sciences, Institute of Bioscience, São Paulo State University, Botucatu, São Paulo State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista (Unesp), Botucatu Medical School, Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso - Barra do Garças, Mato Grosso State, Brazil.
| |
Collapse
|
12
|
Huang N, Wei Y, Liu M, Yang Z, Yuan K, Chen J, Wu Z, Zheng F, Lei K, He M. Dachaihu decoction ameliorates septic intestinal injury via modulating the gut microbiota and glutathione metabolism as revealed by multi-omics. JOURNAL OF ETHNOPHARMACOLOGY 2023; 312:116505. [PMID: 37080366 DOI: 10.1016/j.jep.2023.116505] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 04/05/2023] [Accepted: 04/16/2023] [Indexed: 05/03/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dachaihu decoction (DCH), a classic formula for Yangming and Shaoyang Syndrome Complex recorded in "Treatise on Cold Damage", has been widely used in treating intestinal disorders and inflammatory diseases with few side effects in China. However, the mechanism of DCH on septic intestinal injury (SII) remains to be explored. AIM OF THE STUDY This study aimed to clarify the mechanism of DCH on SII. MATERIALS AND METHODS SII model of rat, established by cecal ligation and puncture (CLP), was used to study the effect of DCH on SII. 24 h mortality was recorded. Histological changes were observed by H&E staining. The expression of tight junction protein ZO-1 (ZO-1) and mucin2 (MUC2) was determined by immunohistochemical analysis. Secretory IgA (sIgA), diamine oxidase (DAO) and intestinal fatty acid binding protein (iFABP) were determined by enzyme-linked immunosorbent assay (ELISA). IL-1β, IL-6 and TNF-α were measured by ELISA and quantitative Real-time PCR (RT-qPCR). The gut microbiota was analyzed by 16S rRNA sequencing. The potential targets and pathways of DCH in treating SII were analyzed by integrative analysis of transcriptomic and metabolomic methods. Total glutathione (T-GSH), GSH, GSSG (reduced form of GSH), GSH peroxidase (GPX), superoxide dismutase (SOD), malonaldehyde (MDA) and indicators of hepatic and renal function were measured by biochemical kits. RESULTS Medium dose of DCH improved 24 h mortality of SII rats, reduced the pathological changes of ileum, and increased the expression levels of ZO-1, MUC2 and sIgA. DCH decreased DAO, iFABP of serum and IL-1β, IL-6, TNF-α of ileum. DCH improved α- and β-diversity and modulated the structure of gut microbiota, with Escherichia_Shigella decreased and Bacteroides and Ruminococcus increased. GSH metabolism was identified as the key pathway of DCH on SII by integrative analysis of transcriptome and metabolome. GSH/GSSG and the most common indicators of oxidative stress, were validated. Antioxidative T-GSH, GSH, GPX and SOD were increased, while MDA, the mark of lipid peroxidation was downregulated by DCH. Eventually, DCH was proved to be safe and hepato- and nephro-protective. CONCLUSION DCH ameliorated septic intestinal injury possibly by modulating the gut microbiota and enhancing glutathione metabolism of SII rats, without hepatotoxicity and nephrotoxicity.
Collapse
Affiliation(s)
- Na Huang
- The Eighth School of Clinical Medicine (Foshan Hospital of Traditional Chinese Medicine), Guangzhou University of Chinese Medicine, Foshan, 528000, China
| | - Yu Wei
- Basic Medical College Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Meng Liu
- Basic Medical College Guangzhou University of Chinese Medicine, Guangzhou, 510000, China
| | - Zhen Yang
- The Eighth School of Clinical Medicine (Foshan Hospital of Traditional Chinese Medicine), Guangzhou University of Chinese Medicine, Foshan, 528000, China
| | - Kang Yuan
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, China
| | - Jingli Chen
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, China
| | - Zhixin Wu
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, China
| | - Fanghao Zheng
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, China
| | - Kaijun Lei
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, China.
| | - Mingfeng He
- Foshan Hospital of Traditional Chinese Medicine, Foshan, 528000, China.
| |
Collapse
|
13
|
Mirahmad M, Mohseni S, Tabatabaei-Malazy O, Esmaeili F, Alatab S, Bahramsoltani R, Ejtahed HS, Qulami H, Bitarafan Z, Arjmand B, Nazeri E. Antioxidative hypoglycemic herbal medicines with in vivo and in vitro activity against C-reactive protein; a systematic review. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 109:154615. [PMID: 36610136 DOI: 10.1016/j.phymed.2022.154615] [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: 06/01/2022] [Revised: 11/16/2022] [Accepted: 12/17/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Inflammation is a double-edged sword in the pathophysiology of chronic diseases, such as type 2 diabetes mellitus (T2DM). The global rise in the prevalence of T2DM in one hand, and poor disease control with currently-available treatments on the other hand, along with an increased tendency towards the use of natural products make scientists seek herbal medicines for the management of diabetes and its complications by reducing C-reactive protein (CRP) as an inflammatory marker. PURPOSE To systematically review the literature to identify the efficacy of various medicinal plants with antioxidative and anti-inflammatory properties considering their effect on CRP in animal models of T2DM. STUDY DESIGN systematic review. METHODS Electronic databases including PubMed, Scopus, Web of Science and Cochran Library were searched using the search terms "herbal medicine", "diabetes", "c-reactive protein", "antioxidants" till August 2021. The quality of evidence was assessed using the Systematic Review Centre for Laboratory animal Experimentation (SYRCLE's) tool. The study protocol was registered in PROSPERO with an ID number CRD42020207190. A manual search to detect any articles not found in the databases was also made. The identified studies were then critically reviewed and relevant data were extracted and summarized. RESULTS Among total of 9904 primarily-retrieved articles, twenty-three experimental studies were finally included. Our data indicated that numerous herbal medicines, compared to placebo or hypoglycemic medications, are effective in treatment of diabetes and its complications through decreasing CRP concentrations and oxidative stresses levels. Medicinal plants including Psidium guajava L., Punica granatum L., Ginkgo biloba L., Punica granatum L., Dianthus superbusn L.. Moreover, Eichhornia crassipes (Mart.) Solms, Curcuma longa L., Azadirachta indica A. Juss., Morus alba L., and Ficus racemosa L. demonstrated potential neuroprotective effects in animal models of diabetes. CONCLUSION Hypoglycemic medicinal plants discussed in this review seem to be promising regulators of CRP, and oxidative stress. Thus, these plants are suitable candidates for management of diabetes' complications. Nevertheless, further high-quality in vivo studies and clinical trials are required to confirm these effects.
Collapse
Affiliation(s)
- Maryam Mirahmad
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Shahrzad Mohseni
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ozra Tabatabaei-Malazy
- Non-Communicable Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fataneh Esmaeili
- Department of Clinical Biochemistry, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sudabeh Alatab
- Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Roodabeh Bahramsoltani
- Department of Traditional Pharmacy, School of Persian Medicine, Tehran University of Medical Sciences, Tehran, Iran; PhytoPharmacology Interest Group (PPIG), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Hanieh-Sadat Ejtahed
- Obesity and Eating Habits Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Husseyn Qulami
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Bitarafan
- Division of Biotechnology and Plant Health, Norwegian Institute of Bioeconomy Research, Høgskoleveien 7, As 1433, Norway
| | - Babak Arjmand
- Cell Therapy and Regenerative Medicine Research Center, Endocrinology and Metabolism Molecular-Cellular Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Elahe Nazeri
- Genetics Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran
| |
Collapse
|
14
|
Aljobaily N, Krutsinger K, Viereckl MJ, Joly R, Menlove B, Cone B, Suppes A, Han Y. Low-Dose Administration of Cannabigerol Attenuates Inflammation and Fibrosis Associated with Methionine/Choline Deficient Diet-Induced NASH Model via Modulation of Cannabinoid Receptor. Nutrients 2022; 15:nu15010178. [PMID: 36615835 PMCID: PMC9823433 DOI: 10.3390/nu15010178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/25/2022] [Accepted: 12/28/2022] [Indexed: 01/03/2023] Open
Abstract
Non-Alcoholic Steatohepatitis (NASH) is the progressive form of Non-Alcoholic Fatty Liver Disease (NAFLD). NASH is distinguished by severe hepatic fibrosis and inflammation. The plant-derived, non-psychotropic compound cannabigerol (CBG) has potential anti-inflammatory effects similar to other cannabinoids. However, the impact of CBG on NASH pathology is still unknown. This study demonstrated the therapeutic potential of CBG in reducing hepatic steatosis, fibrosis, and inflammation. METHODS 8-week-old C57BL/6 male mice were fed with methionine/choline deficient (MCD) diet or control (CTR) diets for five weeks. At the beginning of week 4, mice were divided into three sub-groups and injected with either a vehicle, a low or high dose of CBG for two weeks. Overall health of the mice, Hepatic steatosis, fibrosis, and inflammation were evaluated. RESULTS Increased liver-to-body weight ratio was observed in mice fed with MCD diet, while a low dose of CBG treatment rescued the liver-to-body weight ratio. Hepatic ballooning and leukocyte infiltration were decreased in MCD mice with a low dose of CBG treatment, whereas the CBG treatment did not change the hepatic steatosis. The high dose CBG administration increased inflammation and fibrosis. Similarly, the expression of cannabinoid receptor (CB)1 and CB2 showed decreased expression with the low CBG dose but not with the high CBG dose intervention in the MCD group and were co-localized with mast cells. Additionally, the decreased mast cells were accompanied by decreased expression of transforming growth factor (TGF)-β1. CONCLUSIONS Collectively, the low dose of CBG alleviated hepatic fibrosis and inflammation in MCD-induced NASH, however, the high dose of CBG treatment showed enhanced liver damage when compared to MCD only group. These results will provide pre-clinical data to guide future intervention studies in humans addressing the potential uses of CBG for inflammatory liver pathologies, as well as open the door for further investigation into systemic inflammatory pathologies.
Collapse
Affiliation(s)
- Nouf Aljobaily
- Division of Microbiology and Immunology, Department of Pathology, University of Utah, Salt Lake City, UT 84112, USA
| | - Kelsey Krutsinger
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
| | - Michael J. Viereckl
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
| | - Raznin Joly
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
| | - Bridger Menlove
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
| | - Brexton Cone
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
| | - Ailaina Suppes
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
| | - Yuyan Han
- Department of Biological Sciences, University of Northern Colorado, Greeley, CO 80639, USA
- Correspondence: ; Tel.: +1-970-351-2004
| |
Collapse
|
15
|
Qingda granule prevents obesity-induced hypertension and cardiac dysfunction by inhibiting adverse Akt signaling activation. Heliyon 2022; 8:e12099. [PMID: 36578425 PMCID: PMC9791312 DOI: 10.1016/j.heliyon.2022.e12099] [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: 09/27/2022] [Revised: 10/29/2022] [Accepted: 11/28/2022] [Indexed: 12/12/2022] Open
Abstract
Obesity rates have rapidly increased worldwide and obesity-related diseases such as hypertension and cardiovascular diseases have become leading factors for global morbidity and mortality. Currently, there are no effective treatments that can prevent or reverse obesity long-term, and hence the prevention of obesity-related adverse effects such as hypertension is critical. Qingda granule (QDG) is a condensed Traditional Chinese Medicine (TCM) formula that has been used clinically for treating hypertension, however, its effectiveness in obesity-induced hypertension and cardiac dysfunction remains explored. Mouse models of obesity via long-term feeding of high-fat high-fructose diet (HFFD) were established to examine the effect and mechanism of QDG in protecting against obesity-induced hypertension and cardiac dysfunction. C57BL/6 mice were fed with either normal diet or HFFD over a period of 16 weeks and administered with either saline or QDG for assessment of obesity-induced blood pressure and cardiac function. QDG administration demonstrated robust anti-hypertensive effects and significantly attenuated HFFD-induced elevations in blood pressures. Moreover, QDG treatment also demonstrated robust cardioprotective effects during obesity-induced hypertension by markedly improving cardiac function and preventing cardiac hypertrophy. QDG protected against obesity-induced hypertension and cardiac dysfunction was due to its ability to prevent adverse chronic activation of Akt signaling pathway during long-term feeding of HFFD. Long-term usage of QDG treatments exhibited no observable side effects and also completely prevented obesity-induced organ damage, demonstrating the feasibility and safety of prolonged use. Our findings thus elucidated the role of QDG in preventing obesity-induced hypertension and cardiac hypertrophy via inhibiting adverse activation of Akt signaling activation. Therefore, our study provides the theoretical basis for the utilization of QDG as both a safe and effective drug in the therapeutic treatment of metabolic diseases such as obesity-induced hypertension.
Collapse
|
16
|
Triiodothyronine enhances cardiac contractility in septic rats and probably through Akt-Caspase9 pathway to reduce septic-induced cardiomyocyte apoptosis. Mol Cell Probes 2022; 66:101852. [PMID: 36084907 DOI: 10.1016/j.mcp.2022.101852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/27/2022] [Accepted: 08/27/2022] [Indexed: 12/30/2022]
|
17
|
Wang L, Bao Y, Tong H, Zhang K, Cheng Y, Jin H, Shi J, Wang T, Wang H, Chen G, Wang C. Traditional Mongolian medicine (HHQG) attenuates CCl 4-induced acute liver injury through inhibiting monocyte/macrophage infiltration via the p-P38/p-JNK pathway. JOURNAL OF ETHNOPHARMACOLOGY 2022; 293:115152. [PMID: 35240240 DOI: 10.1016/j.jep.2022.115152] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 02/09/2022] [Accepted: 02/24/2022] [Indexed: 06/14/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Honghua Qinggan 13 Flavor Pills (HHQG), whose Mongolian name is Guri Gumu-13, is a traditional Mongolian medicine, that was stated in the "Diagnosis and Treatment of Ming Medical Code". The HHQG has been included in the Mongolian Medicine Division of the Ministry of Health Drug Standards (1998 edition). Based on our clinical expertise, HHQG demonstrated satisfactory therapeutic effects in hepatitis and liver failure. However, the pharmacological effects and potential mechanisms of HHQG have not been investigated. AIM OF THE STUDY In this study, we combined network pharmacology, transcriptomics, and molecular biology to detect the underlying mechanism for the effect of HHQG on acute liver injury in mice. MATERIALS AND METHODS Network pharmacology was used to explore the pathways involved in the protective effect HHQG in acute liver injury. This effect was further verified by injecting carbon tetrachloride (CCl4; 10 mL/kg, i.p.) to induce acute liver injury in mice. Serum markers of liver injury, morphology, histology, and monocyte/macrophage infiltration in the liver tissue were investigated. Transcriptomics further defined the HHQG targets. Transwell analysis was performed to confirm that HHQG inhibited monocyte/macrophage RAW.264.7 infiltration. qPCR and Western blot were performed to explore the mechanism of action of HHQG. RESULTS Network pharmacology showed that HHQG exerted anti-oxidative and anti-inflammatory effects and promoted metabolic effects against acute liver injury. Pretreatment of mice with HHQG significantly maintained their body weight and decreased serum tumor necrosis factor-alpha (TNF-α) levels induced by CCl4 treatment in vivo. Histopathological examination further confirmed that HHQG protected the liver cells from CCl4-induced damage. Importantly, HHQG significantly inhibited CCl4-induced monocyte/macrophage infiltration. Transcriptomic analysis revealed that HHQG significantly reduced the expression of chemokines and cell adhesion molecules. We determined that HHQG significantly downregulated the expression of the key chemokine (monocyte chemokine protein-1, CCL2) at the gene and protein levels. Further research showed that HHQG inhibited chemokine production in hepatocytes by inhibiting the p-P38 and p-JNK pathways, thereby reducing monocyte/macrophage infiltration. CONCLUSIONS These combined data showed that HHQG alleviated acute liver injury in mice, and further verified that HHQG exerted protective effects by inhibiting the production of CCL2 and reducing the infiltration of monocyte/macrophage by inhibiting the p-P38 and p-JNK pathways.
Collapse
Affiliation(s)
- Li Wang
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China; School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China.
| | - Yulong Bao
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - He Tong
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China
| | - Kefan Zhang
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China
| | - Yipeng Cheng
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China
| | - Haowei Jin
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China
| | - Jing Shi
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China
| | - Tegexibaiyin Wang
- Pharmacy Laboratory, Inner Mongolia International Mongolian Hospital, Hohhot, Inner Mongolia, 010065, China
| | - Haisheng Wang
- School of Basic Medicine, Inner Mongolia Medical University, Hohhot, Inner Mongolia, 010110, China
| | - Guilin Chen
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China.
| | - Changshan Wang
- School of Life Science, Inner Mongolia University, Xin Lin Guo Le Nan Road 49, Yu Quan District, Hohhot, Inner Mongolia, 010020, China; Affiliated Hospital, Inner Mongolia University for the Nationalities, Tongliao, Inner Mongolia, 028007, China.
| |
Collapse
|
18
|
Ding X, Han C, Hu W, Fu C, Zhou Y, Wang Z, Xu Q, Lv R, He C, Zuo Z, Huang J. Acute and Subacute Safety Evaluation of Black Tea Extract (Herbt Tea Essences) in Mice. TOXICS 2022; 10:toxics10060286. [PMID: 35736895 PMCID: PMC9228953 DOI: 10.3390/toxics10060286] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 05/22/2022] [Accepted: 05/24/2022] [Indexed: 12/10/2022]
Abstract
Theabrownin (TB) is a heterogeneous biomacromolecule, extracted from tea, with many functional groups. Importantly, TB possesses diverse health benefits, such as antitumor activity and blood lipid-lowering effects. Presently, the content of TB in tea extract is relatively low. Here, we obtained a deep-processed black tea extract with a high content of TB (close to 80%), which was named Herbt Tea Essences (HTE). Currently, this study was designed to evaluate the biosafety of high-content TB products on mice. We implemented acute and subacute toxic experiments to assess its safety on organs, the serum biochemical and molecular levels. In the acute exposure study, we found that the median lethal dose (LD50) value of HTE was 21.68 g/kg (21.06–24.70 g/kg, greater than 5 g/kg), suggesting that HTE had a low acute toxicity. In the 28-day subacute exposure study, our results showed that no abnormal effects were observed in the 40 and 400 mg/kg/day HTE-treated groups. However, we observed slight nephrotoxicity in the 4000 mg/kg/day HTE-treated group. The HTE-induced nephrotoxic effect might involve the inflammatory response activation mediated by the nuclear transcription factor kappa-B (NF-κB) signaling pathway. This study would provide valuable data for the TB safety assessment and promote this natural biomacromolecule application in daily drinking.
Collapse
Affiliation(s)
- Xiaoyan Ding
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Changshun Han
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Weiping Hu
- First Affiliated Hospital of Xiamen University, Xiamen 361003, China;
| | - Chengqing Fu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Yixi Zhou
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Zheng Wang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Qingyan Xu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Rongfu Lv
- Xiamen Herbt Biotechnology Company Limited, Xiamen 361005, China;
| | - Chengyong He
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
| | - Zhenghong Zuo
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
- Correspondence: (Z.Z.); (J.H.)
| | - Jiyi Huang
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, The Fifth Hospital of Xiamen, Xiang’an Branch of the First Affiliated Hospital, Xiamen University, Xiamen 361102, China; (X.D.); (C.H.); (C.F.); (Y.Z.); (Z.W.); (Q.X.); (C.H.)
- Correspondence: (Z.Z.); (J.H.)
| |
Collapse
|
19
|
Damiano S, Longobardi C, Salzano A, D’Angelo L, Amenta M, Maggiolino A, De Palo P, Claps S, Rufrano D, Iannaccone F, Matera R, Ciarcia R. Red orange and lemon extract preserve from oxidative stress, DNA damage and inflammatory status in lambs. ITALIAN JOURNAL OF ANIMAL SCIENCE 2022. [DOI: 10.1080/1828051x.2022.2056527] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Sara Damiano
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Consiglia Longobardi
- Dipartimento di Salute Mentale, Fisica e Medicina Preventiva, Università della Campania "Luigi Vanvitelli", Napoli Italia
| | - Angela Salzano
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Livia D’Angelo
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Margherita Amenta
- Consiglio per la Ricerca e l’Economia Agraria (CREA)— Centro di Ricerca per l’Olivo, Frutticultura e Agrumi, Acireale, Italia
| | - Aristide Maggiolino
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari“Aldo Moro”, Valenzano (BA), Italy
| | - Pasquale De Palo
- Dipartimento di Medicina Veterinaria, Università degli Studi di Bari“Aldo Moro”, Valenzano (BA), Italy
| | - Salvatore Claps
- Consiglio per la Ricerca e l’Economia Agraria (CREA) – Centro di Ricerca per le Produzioni Animali e l’Acquacoltura, Bella Muro (PZ), Italia
| | - Domenico Rufrano
- Consiglio per la Ricerca e l’Economia Agraria (CREA) – Centro di Ricerca per le Produzioni Animali e l’Acquacoltura, Bella Muro (PZ), Italia
| | - Francesco Iannaccone
- Dipartimento di Scienze Agro-Ambientali e Territoriali (DISAAT), Università degli Studi di Bari “Aldo Moro”, Bari, Italia
| | - Roberta Matera
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| | - Roberto Ciarcia
- Dipartimento di Medicina Veterinaria e Produzioni Animali, Università degli Studi di Napoli Federico II, Napoli, Italia
| |
Collapse
|
20
|
Brookes A, Ji L, Bradshaw TD, Stocks M, Gray D, Butler J, Gershkovich P. Is Oral Lipid-Based Delivery for Drug Targeting to the Brain Feasible? Eur J Pharm Biopharm 2022; 172:112-122. [PMID: 35149190 DOI: 10.1016/j.ejpb.2022.02.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/21/2022] [Accepted: 02/05/2022] [Indexed: 12/12/2022]
Abstract
This review outlines the feasibility of oral lipid-based targeted delivery of drugs to the brain, including permeation of the central nervous system's (CNS) protective blood-brain barrier (BBB). The structure of the BBB and disruption caused by varying disease states highlights the need for disease-specific approaches to alter permeation. Disruption during disease state, and the effects of certain molecules on the barrier, demonstrate the possibility of exploiting such BBB disruption for drug delivery. Many administration methods can be used to target the brain, but oral administration is considered ideal for chronic, long-term illnesses. Several lipids that have been shown to facilitate drug delivery into the brain after systemic administration, but could also be delivered orally are discussed, including oleic acid, triolein, alkylglycerol, and conjugates of linoleic and myristic acids. Current data reveal the potential for the use of such lipids as part of oral formulations for delivery to the brain by reaching sufficient plasma levels after administration to increase the permeability of the BBB. However, gaps in the literature remain regarding the concentrations and form of most lipids required to produce the desired effects. The use of lipids via oral delivery for brain targeting has not been investigated thoroughly enough to determine with certainty if similar permeability-enhancing effects would be observed as for parenteral administration. In conclusion, further research to fill research gaps is needed, but the limited evidence suggests that oral lipid-based drug delivery for brain targeting is potentially feasible.
Collapse
Affiliation(s)
- Alice Brookes
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Liuhang Ji
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Tracey D Bradshaw
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - Michael Stocks
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD
| | - David Gray
- Division of Food, Nutrition and Dietetics, University of Nottingham, Sutton Bonington Campus, Loughborough, Leicestershire, UK, LE12 5RD
| | - James Butler
- GlaxoSmithKline Research and Development, Park Road, Ware, Hertfordshire, UK, SG12 0DP
| | - Pavel Gershkovich
- School of Pharmacy, University of Nottingham, Nottingham, Nottinghamshire, UK, NG7 2RD.
| |
Collapse
|
21
|
Floyd ZE, Ribnicky DM, Raskin I, Hsia DS, Rood JC, Gurley BJ. Designing a Clinical Study With Dietary Supplements: It's All in the Details. Front Nutr 2022; 8:779486. [PMID: 35118104 PMCID: PMC8804374 DOI: 10.3389/fnut.2021.779486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Accepted: 12/15/2021] [Indexed: 11/13/2022] Open
Abstract
A successful randomized clinical trial of the effect of dietary supplements on a chosen endpoint begins with developing supporting data in preclinical studies while paying attention to easily overlooked details when planning the related clinical trial. In this perspective, we draw on our experience studying the effect of an ethanolic extract from Artemisia dracunculus L. (termed PMI-5011) on glucose homeostasis as a potential therapeutic option in providing resilience to metabolic syndrome (MetS). Decisions on experimental design related to issues ranging from choice of mouse model to dosing levels and route of administration in the preclinical studies will be discussed in terms of translation to the eventual human studies. The more complex considerations in planning the clinical studies present different challenges as these studies progress from testing the safety of the dietary supplement to assessing the effect of the dietary supplement on a predetermined clinical outcome. From the vantage point of hindsight, we will outline potential pitfalls when translating preclinical studies to clinical studies and point out details to address when designing clinical studies of dietary supplements.
Collapse
Affiliation(s)
- Z. Elizabeth Floyd
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
- *Correspondence: Z. Elizabeth Floyd
| | - David M. Ribnicky
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, United States
| | - Ilya Raskin
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, United States
| | - Daniel S. Hsia
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Jennifer C. Rood
- Pennington Biomedical Research Center, Louisiana State University System, Baton Rouge, LA, United States
| | - Bill J. Gurley
- National Center for Natural Products Research, University of Mississippi, University, MS, United States
- Bill J. Gurley
| |
Collapse
|
22
|
Qian D, Zhou H, Fan P, Yu T, Patel A, O’Brien M, Wang Z, Lu S, Tong G, Shan Y, Wang L, Gao Y, Xiong Y, Zhang L, Wang X, Liu Y, Zhou S. A Traditional Chinese Medicine Plant Extract Prevents Alcohol-Induced Osteopenia. Front Pharmacol 2021; 12:754088. [PMID: 35002697 PMCID: PMC8730326 DOI: 10.3389/fphar.2021.754088] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 11/11/2021] [Indexed: 12/12/2022] Open
Abstract
Traditional Chinese medicine (TCM) has been practiced in the treatment of bone diseases and alcoholism. Chronic excessive alcohol use results in alcohol-induced bone diseases, including osteopenia and osteoporosis, which increases fracture risk, deficient bone repair, and osteonecrosis. This preclinical study investigated the therapeutic effects of TCM herbal extracts in animal models of chronic excessive alcohol consumption-induced osteopenia. TCM herbal extracts (Jing extracts) were prepared from nine Chinese herbal medicines, a combinative herbal formula for antifatigue and immune regulation, including Astragalus, Cistanche deserticola, Dioscorea polystachya, Lycium barbarum, Epimedium, Cinnamomum cassia, Syzygium aromaticum, Angelica sinensis, and Curculigo orchioides. In this study, Balb/c male mice were orally administrated alcohol (3.2 g/kg/day) with/without TCM herbal extracts (0.125 g/kg, 0.25 g/kg, or 0.5 g/kg) by gavage. Our results showed that after 50 days of oral administration, TCM herbal extracts prevented alcohol-induced osteopenia demonstrated by μ-CT bone morphological analysis in young adults and middle-aged/old Balb/c male mice. Biochemical analysis demonstrated that chronic alcohol consumption inhibits bone formation and has a neutral impact on bone resorption, suggesting that TCM herbal extracts (Jing extracts) mitigate the alcohol-induced abnormal bone metabolism in middle-aged/old male mice. Protocatechuic acid, a natural phenolic acid in Jing extracts, mitigates in vivo alcohol-induced decline of alkaline phosphatase (ALP) gene expression in the bone marrow of Balb/c male mice and in vitro ALP activity in pre-osteoblast MC3T3-E1 cells. Our study suggests that TCM herbal extracts prevent chronic excessive alcohol consumption-induced osteopenia in male mice, implying that traditional medicinal plants have the therapeutic potential of preventing alcohol-induced bone diseases.
Collapse
Affiliation(s)
- Dongyang Qian
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
- Department of Orthopedics, The First Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Hui Zhou
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
| | - Pan Fan
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
- Department of Spine Center, Zhongda Hospital, Southeast University Medical School, Nanjing, China
| | - Tao Yu
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
- Department of Orthopedic Surgery, Tongji Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Anish Patel
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Morgan O’Brien
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Zhe Wang
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
| | - Shiguang Lu
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
| | - Guoqiang Tong
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
| | - Yimin Shan
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
| | - Lei Wang
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
| | - Yuan Gao
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
- Department of Orthopaedics, Qilu Hospital, Shandong University, Jinan, China
| | - Yuan Xiong
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Lily Zhang
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
| | - Xin Wang
- Department of Neurosurgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Yuancai Liu
- Jing Brand Research Institute, Jing Brand Co., Ltd., Daye, China
- *Correspondence: Shuanhu Zhou, , ; Yuancai Liu,
| | - Shuanhu Zhou
- Department of Orthopedic Surgery, Brigham and Women’s Hospital, Harvard Medical School, Harvard University, Boston, MA, United States
- Harvard Stem Cell Institute, Harvard University, Cambridge, MA, United States
- *Correspondence: Shuanhu Zhou, , ; Yuancai Liu,
| |
Collapse
|
23
|
Rodriguez S, Skeet K, Mehmetoglu-Gurbuz T, Goldfarb M, Karri S, Rocha J, Shahinian M, Yazadi A, Poudel S, Subramani R. Phytochemicals as an Alternative or Integrative Option, in Conjunction with Conventional Treatments for Hepatocellular Carcinoma. Cancers (Basel) 2021; 13:cancers13225753. [PMID: 34830907 PMCID: PMC8616323 DOI: 10.3390/cancers13225753] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/15/2021] [Indexed: 12/12/2022] Open
Abstract
Simple Summary Hepatocellular carcinoma (HCC) is globally ranked as the sixth most diagnosed cancer, and the second most deadly cancer. To worsen matters, there are only limited therapeutic options currently available; therefore, it is necessary to find a reservoir from which new HCC treatments may be acquired. The field of phytomedicine may be the solution to this problem, as it offers an abundance of plant-derived molecules, which show capabilities of being effective against HCC proliferation, invasion, migration, and metastasis. In our review, we collect and analyze current evidence regarding these promising phytochemical effects on HCC, and delve into their potential as future chemotherapies. Additionally, information on the signaling behind these numerous phytochemicals is provided, in an attempt to understand their mechanisms. This review makes accessible the current body of knowledge pertaining to phytochemicals as HCC treatments, in order to serve as a reference and inspiration for further research into this subject. Abstract Hepatocellular carcinoma (HCC) is the most abundant form of liver cancer. It accounts for 75–85% of liver cancer cases and, though it ranks globally as the sixth most common cancer, it ranks second in cancer-related mortality. Deaths from HCC are usually due to metastatic spread of the cancer. Unfortunately, there are many challenges and limitations with the latest HCC therapies and medications, making it difficult for patients to receive life-prolonging care. As there is clearly a high demand for alternative therapy options for HCC, it is prudent to turn to plants for the solution, as their phytochemicals have long been used and revered for their many medicinal purposes. This review explores the promising phytochemical compounds identified from pre-clinical and clinical trials being used either independently or in conjunction with already existing cancer therapy treatments. The phytochemicals discussed in this review were classified into several categories: lipids, polyphenols, alkaloids, polysaccharides, whole extracts, and phytochemical combinations. Almost 80% of the compounds failed to progress into clinical studies due to lack of information regarding the toxicity to normal cells and bioavailability. Although large obstacles remain, phytochemicals can be used either as an alternative or integrative therapy in conjunction with existing HCC chemotherapies. In conclusion, phytochemicals have great potential as treatment options for hepatocellular carcinoma.
Collapse
Affiliation(s)
- Sheryl Rodriguez
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
| | - Kristy Skeet
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Tugba Mehmetoglu-Gurbuz
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
| | - Madeline Goldfarb
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (M.G.); (S.K.)
| | - Shri Karri
- Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (M.G.); (S.K.)
| | - Jackelyn Rocha
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Mark Shahinian
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Abdallah Yazadi
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
| | - Seeta Poudel
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
| | - Ramadevi Subramani
- Center of Emphasis in Cancer Research, Department of Molecular and Translational Medicine, Paul L. Foster School of Medicine, Texas Tech University Health Sciences Center El Paso, El Paso, TX 79905, USA; (S.R.); (T.M.-G.); (S.P.)
- Graduate School of Biomedical Sciences, Texas Tech University Health Sciences Center, El Paso, TX 79905, USA; (K.S.); (J.R.); (M.S.); (A.Y.)
- Correspondence: ; Tel.: +1-915-215-6851
| |
Collapse
|
24
|
Li C, Wang M, Sui J, Zhou Y, Chen W. Protective mechanisms of Agrimonia pilosa Ledeb in dextran sodium sulfate-induced colitis as determined by a network pharmacology approach. Acta Biochim Biophys Sin (Shanghai) 2021; 53:1342-1353. [PMID: 34523667 DOI: 10.1093/abbs/gmab116] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Indexed: 12/27/2022] Open
Abstract
Previous studies reported that Agrimonia pilosa (AP) Ledeb possessed diverse biological activities, including anti-inflammatory, antioxidant, and anti-tumor activities. However, the effect of AP on ulcerative colitis (UC) remains unclear. In this study, we investigated the therapeutic effect and mechanisms of AP on dextran sodium sulfate (DSS)-induced colitis. The potential constituents of AP were investigated by ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-Q-TOF/MS). A total of 13 compounds were recognized by UPLC-Q-TOF/MS chromatogram. Furthermore, a network pharmacology approach revealed that there are 297 candidate targets of UC and 549 common targets for the 13 active ingredients of AP. GO enrichment and KEGG pathway analysis indicated that AP might have a protective effect on UC through the nuclear factor κB (NF-κB) and nucleotide-binding oligomerization domain (NOD)-like receptor signaling pathways. Subsequent experimental validation in a DSS-induced colitis model revealed that AP alleviated the severity of DSS-induced colitis, reduced the production of proinflammatory factors, and protected against the loss of intestinal integrity. Moreover, AP inhibited the phosphorylation of NF-κB p65 and the activation of the NLRP3 inflammasome. In conclusion, AP ameliorated DSS-induced colitis through suppressing the activation of the NLRP3 inflammasome and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Chen Li
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Meng Wang
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Junkang Sui
- College of Agriculture, Liaocheng University, Liaocheng 252000, China
| | - Yongkun Zhou
- Department of General Surgery, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan 250014, China
| | - Wei Chen
- Department of Gastroenterology, Shuguang Hospital, Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| |
Collapse
|
25
|
Xu H, Zhou Q, Liu B, Cheng KW, Chen F, Wang M. Neuroprotective Potential of Mung Bean ( Vigna radiata L.) Polyphenols in Alzheimer's Disease: A Review. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:11554-11571. [PMID: 34551518 DOI: 10.1021/acs.jafc.1c04049] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Mung bean contains various neuroprotective polyphenols, so it might be a healthy food for Alzheimer's disease (AD) prevention. Totally, 19 major phenolic compounds were quantified in mung bean, including 10 phenolic acids and 9 flavonoids. After summarizing their contents and effective doses in rodent AD models, it was speculated that vitexin, isovitexin, sinapic acid, and ferulic acid might be the major bioactive compounds for mung bean-mediated neuroprotection. The mechanisms involved inhibition of β-amyloidogenesis, tau hyperphosphorylation, oxidative stress, and neuroinflammation, and promotion of autophagy and acetylcholinesterase enzyme activity. Notably, the neuroprotective phenolic profile in mung bean changed after germination, with decreased vitexin and isovitexin, and increased rutin, isoquercitrin, isorhamnetin, and caffeic acid detected. However, only studies of individual phenolic compounds in mung bean are published at present. Hence, further studies are needed to elucidate the neuroprotective activities and mechanisms of extractions of mung bean seeds and sprouts, and the synergism between different phenolic compounds.
Collapse
Affiliation(s)
- Hui Xu
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Qian Zhou
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Bin Liu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Ka-Wing Cheng
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Feng Chen
- College of Civil and Transportation Engineering, Shenzhen University, Shenzhen 518060, China
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| | - Mingfu Wang
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Institute for Advanced Study, Shenzhen University, Shenzhen 518060, China
- Institute for Innovative Development of Food Industry, Shenzhen University, Shenzhen 518060, China
| |
Collapse
|
26
|
Pharmacological properties of durva swaras (Cynodon dactylon L. Pers.) in an ovariectomised rat model mimicking chronic menopausal syndrome. Biomed Pharmacother 2021; 142:111976. [PMID: 34352715 DOI: 10.1016/j.biopha.2021.111976] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2021] [Revised: 07/22/2021] [Accepted: 07/26/2021] [Indexed: 11/22/2022] Open
Abstract
Hormonal replacement therapy (HRT), as the first-line management of chronic menopausal syndrome (CMS) in women, has limited application due to adverse effects. We aimed to evaluate the therapeutic potential of a herbal alternative (HALT), durva swaras (DS) of Cynodon dactylon L. Pers., in a CMS rat model. Female Sprague-Dawley rats were subjected to Sham and ovariectomy (OVX) surgery. OVX rats received either 0.11 mg/kg oestrogen as a positive treatment control or 1 (DS1), 2 (DS2), and 4 (DS3) g/kg DS for 160 days. Vaginal smear tests indicated the menopausal status. Routine clinical examinations, weekly body weights (BW), serum calcium, proinflammatory cytokines, and reproductive hormones levels were monitored. Clinical chemistry, body composition, bone mineral density (BMD), uterotrophic response, bone morphometry, and histopathology of major organs were evaluated. BW of OVX rats increased by 18-25% compared to Sham. Total fat and fat percentage were significantly elevated in the oestrogen group compared to DS2, DS3, and OVX group. DS treatment groups showed the levels of TNF- α was slightly reduced, while IL-1β and IL-6 levels were significantly reduced (P < 0.05) compared to the oestrogen treated group. DS treatment restored serum calcium levels, while BMD, bone quality, osteoblast/osteoclast ratio, and collagen levels improved in both DS and oestrogen treatment groups. The uterotrophic assay demonstrated non-oestrogenic activity of DS. Endometrial hyperplastic change was observed in oestrogen-treated rats. The preclinical non-oestrogenic activity of DS has therapeutic potential in CMS through anti-inflammatory and osteo-protective effects. Further clinical research into DS, as a viable HALT to HRT, is required.
Collapse
|
27
|
de Abreu MS, Costa F, Giacomini ACVV, Demin KA, Petersen EV, Rosemberg DB, Kalueff AV. Exploring CNS effects of American traditional medicines using zebrafish models. Curr Neuropharmacol 2021; 20:550-559. [PMID: 34254921 DOI: 10.2174/1570159x19666210712153329] [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: 03/06/2021] [Revised: 05/23/2021] [Accepted: 05/28/2021] [Indexed: 11/22/2022] Open
Abstract
Although American traditional medicine (ATM) has been practiced for millennia, its complex multi-target mechanisms of therapeutic action remain poorly understood. Animal models are widely used to elucidate the therapeutic effects of various ATMs, including their modulation of brain and behavior. Complementing rodent models, the zebrafish (Danio rerio) is a promising novel organism in translational neuroscience and neuropharmacology research. Here, we emphasize the growing value of zebrafish for testing neurotropic effects of ATMs and outline future directions of research in this field. We also demonstrate the developing utility of zebrafish as complementary models for probing CNS mechanisms of ATM action and their potential to treat brain disorders.
Collapse
Affiliation(s)
- Murilo S de Abreu
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Fabiano Costa
- Toxicological Biochemistry, Natural and Exact Sciences Center, Federal University of Santa Maria, Brazil
| | - Ana C V V Giacomini
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | | | - Elena V Petersen
- Laboratory of Cell and Molecular Biology and Neurobiology, Moscow Institute of Physics and Technology, Moscow, Russian Federation
| | - Denis B Rosemberg
- The International Zebrafish Neuroscience Research Consortium (ZNRC), Slidell, LA, United States
| | | |
Collapse
|
28
|
Ye Q, Zhang Q, Yao H, Xu A, Liu Y, Qi J, Zhang H, Zhang J. Active-Ingredient Screening and Synergistic Action Mechanism of Shegan Mixture for Anti-Asthma Effects Based on Network Pharmacology in a Mouse Model of Asthma. DRUG DESIGN DEVELOPMENT AND THERAPY 2021; 15:1765-1777. [PMID: 33953545 PMCID: PMC8092947 DOI: 10.2147/dddt.s288829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/08/2021] [Indexed: 12/02/2022]
Abstract
Background Shegan Mixture (SGM) is a traditional Chinese medicine that has anti-inflammatory and therapeutic effects on asthma. However, its active ingredients and combined action mechanism have not been fully elucidated so far. The purpose of this study was to screen the effective ingredients and targets and elucidate the synergistic action mechanism of SGM in asthma mice using the network pharmacological approach. Methods A mouse model of asthma model was used in this study. Mice were orally administered SGM at three doses for 4 weeks and the effect of SGM on asthma was evaluated. The active ingredients and their targets of SGM were identified by searching databases, such as Traditional Chinese Medicine Systems Pharmacology Database (TCMSP). The main active ingredients were selected with parameters OB and DL. The synergistic action mechanisms of SGM in asthma were studied through key active ingredient-target interaction network and verified using surface plasmon resonance assay (SPR). Results SGM exerts anti-asthmatic effects by reducing lung tissue damage and inflammatory factors (IFN-γ, IL-4, IL-5, and IL-13) in asthmatic mice. Twenty ingredients and 45 related proteins were selected as potential nodes using enrichment analysis and network analysis. Inflammation and smooth muscle regulation-related pathways were considered to be the main pharmacological mechanisms of SGM in the treatment of asthma. Especially, 5 molecule-target pairs (including 3 ingredients and 4 proteins) were well docked with each other and the SPR assay revealed that glabridin-PTGS2 had good binding with 44.5 μM Kd value. Conclusion SGM exerts the synergistic anti-asthma effects by virtue of reducing lung-tissue damage and inflammatory factors in asthmatic mice, which explains the theoretical basis for the traditional Chinese medicine, SGM, to treat asthma. Our study thus sheds light on a variety of options including Chinese medicine that could potentially be used in the clinical treatment of asthma.
Collapse
Affiliation(s)
- Qing Ye
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| | - Qiqiang Zhang
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| | - Huijuan Yao
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| | - Ajing Xu
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| | - Yan Liu
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| | - Jia Qi
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| | - Hai Zhang
- Department of Pharmacy, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, 201204, People's Republic of China
| | - Jian Zhang
- Department of Pharmacy, Xinhua Hospital, Affiliated to Shanghai Jiao Tong University, School of Medicine, Shanghai, 200092, People's Republic of China
| |
Collapse
|
29
|
Chen CY, Yuen HM, Lin CC, Hsu CC, Bernard JR, Chen LN, Liao YH, Tsai SC. Anti-fatigue Effects of Santé Premium Silver Perch Essence on Exhaustive Swimming Exercise Performance in Rats. Front Physiol 2021; 12:651972. [PMID: 33828489 PMCID: PMC8019704 DOI: 10.3389/fphys.2021.651972] [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: 01/11/2021] [Accepted: 02/26/2021] [Indexed: 11/18/2022] Open
Abstract
Aim: Fish soup is a traditional Chinese food usually offered as a healthy supplement to elders, pregnant women and persons who just had surgery. Silver perch (Santé premium silver perch essence, SPSPE) extract contains various quality proteins, collagen, minerals, trace elements, and branch chain amino acids (BCAA) that could help individuals recover from exhaustion and control body weight. However, there are very limited studies exploring the effects of fish extracts on exercise performance and fatigue, and relevant physiological mechanisms. Therefore, the purpose of this study was to investigate the effects of chronic SPSPE administration on exhaustive exercise performance. Method: Male Wistar rats weighing around 250 g were divided into 4 groups: Control, 1X SPSPE (6.2 ml/kg), 2X SPSPE (12.4 ml/kg) and 5X SPSPE (31.0 ml/kg). Rats were administrated SPSPE by oral gavage feeding every day for 33 days. Their body weight were measured every week. Before and after the exhaustive swimming test, the blood was collected for circulating lactate, glucose, ammonia, hormones, and myoglobin analysis. Rats were sacrificed after performing an exhaustive swimming exercise test. The liver tissues were collected for glycogen content and H&E staining. Results: After the administration of 1X and 5X SPSPE, swimming fatigue was significantly delayed (p = 0.024). There was no difference in the hormone plasma level between the control and SPSPE groups. The induction of plasma corticosterone and TBARS by exhaustive swimming exercise could be decreased by SPSPE administration. The increased plasma myoglobin concentration from exhaustive swimming exercise was weakened by SPSPE supplementation. The higher glycogen sparing contained in liver tissue was observed in SPSPE-treated groups (p < 0.05). Conclusion: SPSPE could efficiently delay swimming fatigue through sparing of liver glycogen and attenuation of plasma TBARS, myoglobin induction by exhaustive exercise. Our findings provide a scientific-based fundamental information and better understanding for developing a fish extract-based anti-fatigue supplement.
Collapse
Affiliation(s)
- Chung-Yu Chen
- Department of Exercise and Health Sciences, University of Taipei, Taipei City, Taiwan
| | - Hei-Man Yuen
- Institute of Sports Sciences, University of Taipei, Taipei City, Taiwan
| | - Chung-Chi Lin
- Healthcare and Service Center, Taipei Veterans General Hospital, Taipei City, Taiwan.,Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Chi-Chieh Hsu
- Department of Aquatic Sports, University of Taipei, Taipei City, Taiwan
| | - Jeffrey R Bernard
- Department of Kinesiology, California State University-Stanislaus, Turlock, CA, United States
| | - Ling-Ni Chen
- Anyong Biotechnology, Inc., Kaohsiung City, Taiwan
| | - Yi-Hung Liao
- Department of Exercise and Health Science, National Taipei University of Nursing and Health Sciences, Taipei City, Taiwan
| | - Shiow-Chwen Tsai
- Institute of Sports Sciences, University of Taipei, Taipei City, Taiwan
| |
Collapse
|
30
|
Sha J, Cao D, Cui R, Xia L, Hua X, Lu Y, Han S. Mannose Impairs Lung Adenocarcinoma Growth and Enhances the Sensitivity of A549 Cells to Carboplatin. Cancer Manag Res 2020; 12:11077-11083. [PMID: 33173340 PMCID: PMC7648561 DOI: 10.2147/cmar.s278673] [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: 08/26/2020] [Accepted: 10/08/2020] [Indexed: 12/02/2022] Open
Abstract
Introduction Mannose, a major monosaccharide component of N-glycans, involves in the glycometabolism of human body. Recently, mannose has been shown to suppress tumor growth through enhancing chemosensitivity and reducing the activity of mannose phosphate isomerase (MPI). However, it is largely unknown whether mannose exerts effects on non-small cell lung cancer (NSCLC). Materials and Methods First, a mannose IC50 assay was conducted to find a suitable concentration of mannose for cell experiments. Then, vitro studies including CCK-8 assay, scratch wound healing assay, and TUNEL assay were performed to evaluate the effects of mannose on A549 cells, and an animal model was established to evaluate the antitumoural effect of mannose on NSCLC in vivo. Finally, immunohistochemistry was done to detect the expression of MPI by Rabbit Anti-MPI. Results In this study, a concentration of mannose, 15mM, was used to explore the suppressive effect of mannose on A549 cells. CCK-8 assay demonstrated that mannose significantly inhibited the proliferation of A549 cells and enhanced the anti-tumor efficacy of carboplatin. Wound healing assay showed that mannose inhibits the migration of A549 cells, and mannose-induced migration inhibition was more efficient in A549 cells treated with carboplatin. TUNEL assay demonstrated that mannose significantly enhanced the efficacy of carboplatin to promote apoptosis treated by mannose (15mM) or carboplatin. The results of animal experiments revealed that the size and weight of tumors derived from A549 cells treated with mannose were smaller than those derived from control cells, and co-treatment with mannose and carboplatin had most efficient inhibition on tumor growth. MPI expression detection showed that the expression level of MPI in the stage Tis (tumor in situ) was the highest, while the stage IV has the lowest. Discussion Collectively, our findings suggest that mannose inhibited cell proliferation and migration, promoted cell apoptosis and enhanced the efficacy of carboplatin in lung adenocarcinoma. Preliminary results showed that mannose had less side effect on health. In the future, mannose may be a potential candidate drug for adjuvant therapy of lung adenocarcinoma.
Collapse
Affiliation(s)
- Jun Sha
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China.,Medicine Department of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Dandan Cao
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China.,Medicine Department of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Rui Cui
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China.,Medicine Department of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Lu Xia
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China.,Medicine Department of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Xin Hua
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China.,Medicine Department of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Yuan Lu
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China.,Medicine Department of Southeast University, Nanjing, Jiangsu, People's Republic of China
| | - Shuhua Han
- Department of Respiratory Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing City, Jiangsu Province, People's Republic of China
| |
Collapse
|
31
|
Yang M, Yan T, Yu M, Kang J, Gao R, Wang P, Zhang Y, Zhang H, Shi L. Advances in understanding of health‐promoting benefits of medicine and food homology using analysis of gut microbiota and metabolomics. FOOD FRONTIERS 2020. [DOI: 10.1002/fft2.49] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Affiliation(s)
- Minmin Yang
- College of Life Sciences Shaanxi Normal University Xi'an China
| | - Tao Yan
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Meng Yu
- The Institute of Medicinal Plant Development Chinese Academy of Medical Sciences and Peking Union Medical College Beijing China
| | - Jie Kang
- Physical Education Institute Shaanxi Normal University Xi'an China
| | - Ruoxi Gao
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Peng Wang
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Yuhuan Zhang
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
| | - Huafeng Zhang
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
- Internatinal Joint Research Center of Shaanxi Province for Food and Health Science Shaanxi Normal University Xi'an China
| | - Lin Shi
- School of Food Engineering and Nutritional Science Shaanxi Normal University Xi'an China
- Internatinal Joint Research Center of Shaanxi Province for Food and Health Science Shaanxi Normal University Xi'an China
- Department of Biology and Biological Engineering Chalmers University of Technology Gothenburg Sweden
| |
Collapse
|
32
|
Safety and Molecular-Toxicological Implications of Cannabidiol-Rich Cannabis Extract and Methylsulfonylmethane Co-Administration. Int J Mol Sci 2020; 21:ijms21207808. [PMID: 33096940 PMCID: PMC7656295 DOI: 10.3390/ijms21207808] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/15/2020] [Accepted: 10/18/2020] [Indexed: 12/16/2022] Open
Abstract
Cannabidiol (CBD) is a biologically active, non-psychotropic component of Cannabis sativa whose popularity has grown exponentially in recent years. Besides a wealth of potential health benefits, ingestion of CBD poses risks for a number of side effects, of which hepatotoxicity and CBD/herb-drug interactions are of particular concern. Here, we investigated the interaction potential between the cannabidiol-rich cannabis extract (CRCE) and methylsulfonylmethane (MSM), a popular dietary supplement, in the mouse model. For this purpose, 8-week-old male C57BL6/J mice received MSM-containing water (80 mg/100 mL) ad libitum for 17 days. During the last three days of treatment, mice received three doses of CRCE administered in sesame oil via oral gavage (123 mg/kg/day). Administration of MSM alone did not result in any evidence of liver toxicity and did not induce expression of mouse cytochrome P450 (CYP) enzymes. Administration of CRCE did produce significant (p < 0.05) increases in Cyp1a2, Cyp2b10, Cyp2c29, Cyp3a4, Cyp3a11, Cyp2c65, and Cyp2c66 messenger RNA, however, this effect was not amplified by MSM/CRCE co-treatment. Similarly, no evidence of liver toxicity was observed in MSM/CRCE dosed mice. In conclusion, short-term MSM/CRCE co-administration did not demonstrate any evidence of hepatotoxicity in the mouse model.
Collapse
|
33
|
Li Y, Xie M, Zhou J, Lin H, Xiao T, Wu L, Ding H, Fang B. Increased Antimicrobial Activity of Colistin in Combination With Gamithromycin Against Pasteurella multocida in a Neutropenic Murine Lung Infection Model. Front Microbiol 2020; 11:511356. [PMID: 33072002 PMCID: PMC7536268 DOI: 10.3389/fmicb.2020.511356] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2019] [Accepted: 08/21/2020] [Indexed: 01/09/2023] Open
Abstract
We investigate the antimicrobial activity of combined colistin and gamithromycin against nine Pasteurella multocida strains by testing in vitro susceptibility. Two high-colistin minimal inhibitory concentration (MIC) isolates (D18 and T5) and one low-colistin MIC isolate (WJ11) were used in time-kill tests and therapeutic effect experiments using a neutropenic murine pneumonia model over 24 h. Pharmacokinetics (PK) in plasma was calculated along with pharmacodynamics (PD) to determine the PK/PD index. Synergy between colistin and gamithromycin was observed using high-colistin MIC isolates, equating to a 128- or 256-fold and 4- or 8-fold reduction in colistin and gamithromycin concentration, respectively. Interestingly, no synergistic effect of the combination on low-colistin MIC isolates was observed. However, regardless of the MIC difference among isolates, each drug tended to reach the same concentration in all isolates subjected to combined treatments, which was verified by the time-kill tests presenting similar rates and extent of killing for isolates D18, T5, and WJ11. The AUC( 0 - 24 h)/MIC index was used to evaluate the relationship between PK and PD, and the correlation was >0.89. The relevant gamithromycin doses for combined therapy were determined, and the value decreased from 6- to 35-fold compared with monotherapy. Combined colistin and gamithromycin therapy provides a more potent therapeutic regimen than monotherapy against P. multocida strains.
Collapse
Affiliation(s)
- Yanqin Li
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Mengjuan Xie
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Junwen Zhou
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Hao Lin
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Tianan Xiao
- Guangdong Center for Agricultural Products Quality and Safety, Guangzhou, China
| | - Liqin Wu
- Guangdong Center for Agricultural Products Quality and Safety, Guangzhou, China
| | - Huanzhong Ding
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| | - Binghu Fang
- National Risk Assessment Laboratory for Antimicrobial Resistance of Animal Original Bacteria, South China Agricultural University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Veterinary Pharmaceutics Development and Safety Evaluation, South China Agricultural University, Guangzhou, China
| |
Collapse
|
34
|
Zhao W, Song F, Hu D, Chen H, Zhai Q, Lu W, Zhao J, Zhang H, Chen W, Gu Z, Wang G. The Protective Effect of Myristica fragrans Houtt. Extracts Against Obesity and Inflammation by Regulating Free Fatty Acids Metabolism in Nonalcoholic Fatty Liver Disease. Nutrients 2020; 12:E2507. [PMID: 32825154 PMCID: PMC7551042 DOI: 10.3390/nu12092507] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2020] [Revised: 08/05/2020] [Accepted: 08/17/2020] [Indexed: 02/06/2023] Open
Abstract
Nonalcoholic fatty liver disease (NAFLD) is a disorder characterized by the excess accumulation of fat in the hepatocytes. It is commonly associated with severe obesity and inflammation. Free fatty acids (FFAs) are the key to regulate lipid metabolism and immune response in hepatocyte cells. This study examined the effects of AEN (alcohol extract of nutmeg, the seed of Myristica fragrans Houtt.) on the inhibition of lipid synthesis and inflammation in vitro and in vivo and on high-fat diet-induced obesity in NAFLD mice. Our results showed that AEN treatment could downregulate the expression of lipid synthesis-related genes fatty acid synthase (FASN) and sterol regulatory element-binding protein 1c (SREBP-1c) and lower the lipid content of cells. AEN also inhibited FFAs-mediated inflammation-related cytokines interleukin-6 (IL-6) and tumor necrosis factor α (TNFα) expression in cells. In a mouse model, AEN reduced the bodyweight of obese mice and improved NAFLD without affecting food intake. Further analysis revealed that AEN significantly reduced inflammation level, cholesterol and lipid accumulation, blood glucose, and other liver function indexes in mice fed with a high-fat diet. In conclusion, AEN inhibited the aggravation of obesity and inflammation by downregulating lipid-gene expression in the liver to ameliorate NAFLD.
Collapse
Affiliation(s)
- Wenyu Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Fanfen Song
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Diangeng Hu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Haiqin Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| | - Hao Zhang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Wuxi Translational Medicine Research Center and Jiangsu Translational Medicine Research Institute Wuxi Branch, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
- Beijing Innovation Centre of Food Nutrition and Human Health, Beijing Technology and Business University (BTBU), Beijing 100048, China
| | - Zhennan Gu
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
| | - Gang Wang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China; (W.Z.); (F.S.); (D.H.); (H.C.); (Q.Z.); (W.L.); (J.Z.); (H.Z.); (W.C.); (Z.G.)
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Probiotics, Jiangnan University, Wuxi 214122, China
- (Yangzhou) Institute of Food Biotechnology, Jiangnan University, Yangzhou 225004, China
| |
Collapse
|
35
|
Pharmacokinetics and tissue distribution of eighteen major alkaloids of Aconitum carmichaelii in rats by UHPLC-QQQ-MS. J Pharm Biomed Anal 2020; 185:113226. [DOI: 10.1016/j.jpba.2020.113226] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 03/01/2020] [Accepted: 03/02/2020] [Indexed: 12/18/2022]
|
36
|
Paula VG, Cruz LL, Sene LB, Gratão TB, Soares TS, Moraes-Souza RQ, Damasceno DC, Volpato GT. Maternal-fetal repercussions of Phyllanthus niruri L. treatment during rat pregnancy. JOURNAL OF ETHNOPHARMACOLOGY 2020; 254:112728. [PMID: 32145330 DOI: 10.1016/j.jep.2020.112728] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 02/11/2020] [Accepted: 02/29/2020] [Indexed: 06/10/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Phyllanthus niruri is a well-known plant for its therapeutic purposes to treat various diseases, being widely used by the population, mainly by women. However, there is no scientific confirmation of the effects of use during pregnancy. AIM OF THE STUDY Evaluating the effect of Phyllanthus niruri aqueous extract on the maternal toxicity, reproductive outcomes and fetal anomaly incidence in rats. MATERIALS AND METHODS Pregnant rats were distributed into four experimental groups: Control = treated with water (vehicle); Treated 150 = treated with P. niruri at dose 150 mg/kg and; Treated 300 = treated with P. niruri at dose 300 mg/kg; and Treated 600 = treated with P. niruri at dose 600 mg/kg. The rats were treated by intragastric route (gavage) with P. niruri or vehicle (water) from gestational day 0 to 21. At day 21 of pregnancy, maternal reproductive outcomes, biochemical profile and maternal renal tissue were evaluated. The fetuses and placentas were collected and analyzed. RESULTS Treatment with P. niruri did not alter the reproductive performance outcomes of rats. However, treated 600 group presented with changes in maternal kidney weight and morphology. The plant did not present teratogenic effect, but caused fetal macrosomia and increased ossification sites. CONCLUSION Treatment with aqueous extract of P. niruri administered during gestation did not cause reproductive toxicity, but led to changes in maternal kidneys and in offspring weight, showing that the leaf extract of this plant can produce detrimental effects during pregnancy.
Collapse
Affiliation(s)
- Verônyca Gonçalves Paula
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Larissa Lopes Cruz
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Letícia Barros Sene
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Thamires Ballarini Gratão
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Mato Grosso State, Brazil
| | - Thaigra Sousa Soares
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Rafaianne Queiroz Moraes-Souza
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Débora Cristina Damasceno
- Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil
| | - Gustavo Tadeu Volpato
- Laboratory of System Physiology and Reproductive Toxicology, Institute of Biological and Health Sciences, Federal University of Mato Grosso, Barra do Garças, Mato Grosso State, Brazil; Laboratory of Experimental Research on Gynecology and Obstetrics, Postgraduate Course on Tocogynecology, Univ Estadual Paulista_Unesp, Botucatu, São Paulo State, Brazil.
| |
Collapse
|
37
|
Skinner CM, Nookaew I, Ewing LE, Wongsurawat T, Jenjaroenpun P, Quick CM, Yee EU, Piccolo BD, ElSohly M, Walker LA, Gurley B, Koturbash I. Potential Probiotic or Trigger of Gut Inflammation - The Janus-Faced Nature of Cannabidiol-Rich Cannabis Extract. J Diet Suppl 2020; 17:543-560. [PMID: 32400224 DOI: 10.1080/19390211.2020.1761506] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Cannabidiol (CBD) is the major non-psychotropic phytocannabinoid present in Cannabis sativa. In 2018, Congress designated certain C. sativa plant material as "hemp," thus removing it from the DEA's list of controlled substances. As a result, CBD-containing hemp extracts and other CBD products are now widely available and heavily marketed, yet their FDA regulatory status is still hotly debated. The goal of this study was to investigate the effects of a cannabidiol-rich cannabis extract (CRCE) on the gut microbiome and associated histomorphological and molecular changes in the mouse gut mucosa. Male C57BL6/J mice were gavaged with either 0, 61.5, 184.5, or 615 mg/kg/bw of CRCE in sesame oil for 2 weeks (Mon-Fri). Substantial CRCE-induced increases in the relative abundance of A. muciniphila, a bacterial species currently accepted as probiotic, was observed in fecal samples at all doses. This was paralleled by decreases in the relative abundance of other gut bacterial species. Coincident with the observed changes in gut ecology were multiple pro-inflammatory responses, including increased expression of cytokines and chemokines-Il1ß, Cxcl1, and Cxcl2 in the colon tissue. Furthermore, dramatic increases in the relative abundance of A. muciniphila significantly decreased expression of Muc2-a gene intimately associated with gut integrity. Taken together, these findings raise concerns about the safety of long-term CBD usage and underline the need for additional well-designed studies into its tolerability and efficacy.
Collapse
Affiliation(s)
- Charles M Skinner
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Intawat Nookaew
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Thidathip Wongsurawat
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Piroon Jenjaroenpun
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Charles M Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Eric U Yee
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Brian D Piccolo
- United States Department of Agriculture(USDA), Arkansas Children's Nutrition Center, Little Rock, AR, USA.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| | - Mahmoud ElSohly
- National Center for Natural Products Research, University of Mississippi, MS, USA.,Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, MS, USA.,ElSohly Laboratories, Inc. (ELI), Oxford, MS, USA
| | - Larry A Walker
- National Center for Natural Products Research, University of Mississippi, MS, USA.,ElSohly Laboratories, Inc. (ELI), Oxford, MS, USA
| | - Bill Gurley
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,National Center for Natural Products Research, University of Mississippi, MS, USA
| | - Igor Koturbash
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR, USA.,Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, USA
| |
Collapse
|
38
|
Choi S, Kim C, Son H, Hwang JK, Kang W. Estimation of an Appropriate Human Dose of Boesenbergia pandurata Extracts Based on Allometric Scaling Data of Panduratin A in Mice, Rats, and Dogs. J Med Food 2020; 23:453-458. [PMID: 32176564 DOI: 10.1089/jmf.2019.4564] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
It can be difficult to identify health/functional foods that exert therapeutic benefits for alleviating gingivitis and periodontitis. Recently, extracts of Boesenbergia pandurata (Roxb.), which is a tropical plant, have shown promising inhibitory activity against lipopolysaccharide-induced periodontitis. As a result, a clinical trial is being planned to assess utility of B. pandurata (Roxb.) extracts for promoting oral health; this study was designed to determine an appropriate human dose of the extracts for the trial. Pharmacokinetic studies of panduratin A, which is an active substance in fingerroot, were carried out in mice, rats, and dogs after oral administration of the extracts. The clearance data for each species were used to estimate clearance in humans through allometric scaling based on the maximum lifespan potential, and a daily dose providing sufficient anti-periodontitis activity was estimated for use in the clinical trial. The findings indicated that allometric scaling is a reasonable approach that is relatively free of safety issues and can be used to determine doses of substances for incorporation into health/functional foods appropriate for humans.
Collapse
Affiliation(s)
- Seungmok Choi
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Changhee Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Heebin Son
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| | - Jae-Kwan Hwang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul, Korea
| | - Wonku Kang
- College of Pharmacy, Chung-Ang University, Seoul, Korea
| |
Collapse
|
39
|
Cangiano LR, Zenobi MG, Nelson CD, Ipharraguerre IR, Dilorenzo N. A bioactive extract from Olea europaea protects newly weaned beef heifers against experimentally induced chronic inflammation1. J Anim Sci 2020; 97:4349-4361. [PMID: 31581301 DOI: 10.1093/jas/skz285] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2019] [Accepted: 09/13/2019] [Indexed: 01/17/2023] Open
Abstract
Weaning is one of the most stressful periods in the life of a ruminant. Several factors entrenched within typical management practices pose challenges to the calf gastrointestinal health. Weaning is associated with losses in BW and feed intake. In addition, increasing highly fermentable carbohydrates in the diet at the expense of physically effective fiber after weaning predisposes the development of rumen acidosis and increases the concentration of endotoxin in rumen fluid and the permeability of the lower gut to luminal contents. Endotoxin translocation can elicit immune activation, shifting the metabolic priorities toward the immune system, which if sustained over time can hinder animal health and performance. Strategic supplementation of additives with anti-inflammatory capacity could represent a suitable approach to decrease systemic inflammation, restoring barrier function to luminal contents. Bioactive extracts from Olea europaea have anti-inflammatory activity and have been shown to reduce systemic inflammation in other animal models. A generalized randomized block design was used to evaluate the impact of feeding an olive oil bioactive extract (OBE) to newly weaned heifers injected intravenously with sequentially increasing doses of lipopolysaccharide (LPS). A total of 36 heifers, distributed across 3 experimental periods, were randomly assigned to 1 of 4 treatments that consisted of intravenous injection of either saline (CTL-) or with 6 sequentially increasing doses of LPS (0.10, 0.25, 0.50, 0.75, 1.00, and 1.25 µg/kg of BW) over a 10-d period (CTL+), and CTL+ plus dietary supplementation with a low (OBE-L; 0.04% of diet DM) or a high (OBE-H; 0.16% of diet DM) dose of OBE. Feeding OBE reduced some of the negative effects of prolonged immune activation with LPS, such as improved DMI and decreased intravaginal temperature in some, but not all of the days of LPS challenge (P < 0.05). In addition, feeding OBE reduced circulating concentration of inflammatory markers such as IL-6 and haptoglobin (P < 0.05). Heifers supplemented with OBE had reduced cell surface expression of the cluster of differentiation 14 (CD14) in monocyte cells (P < 0.01), a key receptor for LPS recognition, which was correlated with a faster recovery of immune cell counts in plasma. In conclusion, dietary supplementation with OBE was successful in mitigating the negative effects of sustained immune activation in newly weaned heifers.
Collapse
Affiliation(s)
- Lautaro R Cangiano
- North Florida Research and Education Center, Department of Animal Sciences, University of Florida, Marianna, FL
| | - Marcos G Zenobi
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | - Corwin D Nelson
- Department of Animal Sciences, University of Florida, Gainesville, FL
| | | | - Nicolas Dilorenzo
- North Florida Research and Education Center, Department of Animal Sciences, University of Florida, Marianna, FL
| |
Collapse
|
40
|
Zhu B, Luo X, Gao Z, Feng Q, Hu L, Weng Q. The complete chloroplast genome sequence of Dendrobium thyrsiflorum (Orchidaceae) and its phylogenetic analysis. Mitochondrial DNA B Resour 2019; 4:4085-4086. [PMID: 33366330 PMCID: PMC7707769 DOI: 10.1080/23802359.2019.1691947] [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: 09/11/2019] [Accepted: 10/26/2019] [Indexed: 11/21/2022] Open
Abstract
Dendrobium thyrsiflorum Rchb.f., a native species to China, is widely used as an important garden flower and a traditional Chinese medicine. Herein, the complete chloroplast (cp) genome of D. thyrsiflorum was deciphered by high-throughput sequencing. The cp genome exhibited a typical quadripartite cycle of 151,686 bp in length, comprising of a pair of inverted repeats (IRa and IRb) of 26,293 bp which were intersected by a large single copy (LSC) region of 84,749 bp and a small single copy (SSC) region of 14,351 bp. A total of 126 genes were de novo assembled in this cp genome, including 78 protein genes, 40 tRNA genes and 8 rRNA genes. Among these genes, 86 genes (22 tRNAs and 64 coding genes) were single copy, the rest were two-copy genes, and the average of GC content of the whole genome is 37.55%. Phylogenetic trees showed that the D. thyrsiflorum was closely related to D. devonianum. This study provides molecular information for future evolution, genetic and molecular biology studies of Dendrobium.
Collapse
Affiliation(s)
- Bin Zhu
- School of Life Sciences, Guizhou Normal University, Guiyang, People’s Republic of China
| | - Xi Luo
- School of Life Sciences, Guizhou Normal University, Guiyang, People’s Republic of China
| | - Zuomin Gao
- School of Life Sciences, Guizhou Normal University, Guiyang, People’s Republic of China
| | - Qun Feng
- School of Life Sciences, Guizhou Normal University, Guiyang, People’s Republic of China
| | - Lijuan Hu
- School of Life Sciences, Guizhou Normal University, Guiyang, People’s Republic of China
| | - Qingbei Weng
- School of Life Sciences, Guizhou Normal University, Guiyang, People’s Republic of China
| |
Collapse
|
41
|
Ewing LE, McGill MR, Yee EU, Quick CM, Skinner CM, Kennon-McGill S, Clemens M, Vazquez JH, McCullough SS, Williams DK, Kutanzi KR, Walker LA, ElSohly MA, James LP, Gurley BJ, Koturbash I. Paradoxical Patterns of Sinusoidal Obstruction Syndrome-Like Liver Injury in Aged Female CD-1 Mice Triggered by Cannabidiol-Rich Cannabis Extract and Acetaminophen Co-Administration. Molecules 2019; 24:molecules24122256. [PMID: 31212965 PMCID: PMC6630875 DOI: 10.3390/molecules24122256] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 06/06/2019] [Accepted: 06/13/2019] [Indexed: 12/18/2022] Open
Abstract
The goal of this study was to investigate the potential for a cannabidiol-rich cannabis extract (CRCE) to interact with the most common over-the-counter drug and the major known cause of drug-induced liver injury–acetaminophen (APAP)–in aged female CD-1 mice. Gavaging mice with 116 mg/kg of cannabidiol (CBD) [mouse equivalent dose (MED) of 10 mg/kg of CBD] in CRCE delivered with sesame oil for three consecutive days followed by intraperitoneally (i.p.) acetaminophen (APAP) administration (400 mg/kg) on day 4 resulted in overt toxicity with 37.5% mortality. No mortality was observed in mice treated with 290 mg/kg of CBD+APAP (MED of 25 mg/kg of CBD) or APAP alone. Following CRCE/APAP co-administration, microscopic examination revealed a sinusoidal obstruction syndrome-like liver injury–the severity of which correlated with the degree of alterations in physiological and clinical biochemistry end points. Mechanistically, glutathione depletion and oxidative stress were observed between the APAP-only and co-administration groups, but co-administration resulted in much greater activation of c-Jun N-terminal kinase (JNK). Strikingly, these effects were not observed in mice gavaged with 290 mg/kg CBD in CRCE followed by APAP administration. These findings highlight the potential for CBD/drug interactions, and reveal an interesting paradoxical effect of CBD/APAP-induced hepatotoxicity.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Mitchell R McGill
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Eric U Yee
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Charles M Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Charles M Skinner
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Stefanie Kennon-McGill
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Melissa Clemens
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Joel H Vazquez
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Sandra S McCullough
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - D Keith Williams
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Kristy R Kutanzi
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Larry A Walker
- National Center for Natural Products Research, University of Mississippi, University, MS 38677, USA.
- ElSohly Laboratories, Inc. (ELI), Oxford, MS 38677, USA.
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, University of Mississippi, University, MS 38677, USA.
- ElSohly Laboratories, Inc. (ELI), Oxford, MS 38677, USA.
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Laura P James
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Bill J Gurley
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
42
|
Ewing LE, Skinner CM, Quick CM, Kennon-McGill S, McGill MR, Walker LA, ElSohly MA, Gurley BJ, Koturbash I. Hepatotoxicity of a Cannabidiol-Rich Cannabis Extract in the Mouse Model. Molecules 2019; 24:molecules24091694. [PMID: 31052254 PMCID: PMC6539990 DOI: 10.3390/molecules24091694] [Citation(s) in RCA: 86] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2019] [Revised: 04/29/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023] Open
Abstract
The goal of this study was to investigate Cannabidiol (CBD) hepatotoxicity in 8-week-old male B6C3F1 mice. Animals were gavaged with either 0, 246, 738, or 2460 mg/kg of CBD (acute toxicity, 24 h) or with daily doses of 0, 61.5, 184.5, or 615 mg/kg for 10 days (sub-acute toxicity). These doses were the allometrically scaled mouse equivalent doses (MED) of the maximum recommended human maintenance dose of CBD in EPIDIOLEX® (20 mg/kg). In the acute study, significant increases in liver-to-body weight (LBW) ratios, plasma ALT, AST, and total bilirubin were observed for the 2460 mg/kg dose. In the sub-acute study, 75% of mice gavaged with 615 mg/kg developed a moribund condition between days three and four. As in the acute phase, 615 mg/kg CBD increased LBW ratios, ALT, AST, and total bilirubin. Hepatotoxicity gene expression arrays revealed that CBD differentially regulated more than 50 genes, many of which were linked to oxidative stress responses, lipid metabolism pathways and drug metabolizing enzymes. In conclusion, CBD exhibited clear signs of hepatotoxicity, possibly of a cholestatic nature. The involvement of numerous pathways associated with lipid and xenobiotic metabolism raises serious concerns about potential drug interactions as well as the safety of CBD.
Collapse
Affiliation(s)
- Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Charles M Skinner
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Charles M Quick
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Stefanie Kennon-McGill
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Mitchell R McGill
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| | - Larry A Walker
- National Center for Natural Products Research, University of Mississippi, University, MS 38677, USA.
- ElSohly Laboratories, Inc. (ELI), Oxford, MS 38655, USA.
| | - Mahmoud A ElSohly
- National Center for Natural Products Research, University of Mississippi, University, MS 38677, USA.
- ElSohly Laboratories, Inc. (ELI), Oxford, MS 38655, USA.
- Department of Pharmaceutics and Drug Delivery, School of Pharmacy, University of Mississippi, University, MS 38677, USA.
| | - Bill J Gurley
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72223, USA.
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA.
| |
Collapse
|
43
|
Gurley BJ, Miousse IR, Nookaew I, Ewing LE, Skinner CM, Jenjaroenpun P, Wongsurawat T, Kennon-McGill S, Avula B, Bae JY, McGill MR, Ussery D, Khan IA, Koturbash I. Decaffeinated Green Tea Extract Does Not Elicit Hepatotoxic Effects and Modulates the Gut Microbiome in Lean B6C3F₁ Mice. Nutrients 2019; 11:nu11040776. [PMID: 30987244 PMCID: PMC6521095 DOI: 10.3390/nu11040776] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 03/25/2019] [Accepted: 03/29/2019] [Indexed: 02/07/2023] Open
Abstract
The main purpose of this study was to investigate the hepatotoxic potential and effects on the gut microbiome of decaffeinated green tea extract (dGTE) in lean B6C3F1 mice. Gavaging dGTE over a range of 1X–10X mouse equivalent doses (MED) for up to two weeks did not elicit significant histomorphological, physiological, biochemical or molecular alterations in mouse livers. At the same time, administration of dGTE at MED comparable to those consumed by humans resulted in significant modulation of gut microflora, with increases in Akkermansia sp. being most pronounced. Results of this study demonstrate that administration of relevant-to-human-consumption MED of dGTE to non-fasting mice does not lead to hepatotoxicity. Furthermore, dGTE administered to lean mice, caused changes in gut microflora comparable to those observed in obese mice. This study provides further insight into the previously reported weight management properties of dGTE; however, future studies are needed to fully evaluate and understand this effect.
Collapse
Affiliation(s)
- Bill J Gurley
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Department of Biochemistry and Molecular Biology, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Intawat Nookaew
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Charles M Skinner
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Piroon Jenjaroenpun
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Thidathip Wongsurawat
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Stefanie Kennon-McGill
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Bharathi Avula
- National Center for Natural Product Research, School of Pharmacy, The University of Mississippi, University, MS 38677, USA.
| | - Ji-Yeong Bae
- National Center for Natural Product Research, School of Pharmacy, The University of Mississippi, University, MS 38677, USA.
| | - Mitchell R McGill
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - David Ussery
- Department of Biomedical Informatics, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | - Ikhlas A Khan
- National Center for Natural Product Research, School of Pharmacy, The University of Mississippi, University, MS 38677, USA.
| | - Igor Koturbash
- Center for Dietary Supplements Research, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| |
Collapse
|
44
|
Ranneh Y, Akim AM, Hamid HA, Khazaai H, Fadel A, Mahmoud AM. Stingless bee honey protects against lipopolysaccharide induced-chronic subclinical systemic inflammation and oxidative stress by modulating Nrf2, NF-κB and p38 MAPK. Nutr Metab (Lond) 2019; 16:15. [PMID: 30858869 PMCID: PMC6391794 DOI: 10.1186/s12986-019-0341-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 02/13/2019] [Indexed: 01/21/2023] Open
Abstract
Background Epidemiological and experimental studies have extensively indicated that chronic subclinical systemic inflammation (CSSI) and oxidative stress are risk factors for several chronic diseases, including cancer, arthritis, type 2 diabetes, and cardiovascular and neurodegenerative diseases. This study examined the protective effect of stingless bee honey (SBH) supplementation against lipopolysaccharide (LPS)-induced CSSI, pointing to the possible involvement of NF-κB, p38 MAPK and Nrf2 signaling. Methods CSSI was induced in male Sprague Dawley rats by intraperitoneal injection of LPS three times per week for 28 days, and SBH (4.6 and 9.3 g/kg/day) was supplemented for 30 days. Results LPS-induced rats showed significant leukocytosis, and elevated serum levels of CRP, TNF-α, IL-1β, IL-6, IL-8, MCP-1, malondialdehyde (MDA) and 8-hydroxy-2'-deoxyguanosine (8-OHdG), accompanied with diminished antioxidants. Treatment with SBH significantly ameliorated inflammatory markers, MDA and 8-OHdG, and enhanced antioxidants in LPS-induced rats. In addition, SBH decreased NF-κB p65 and p38 MAPK, and increased Nrf2 expression in the liver, kidney, heart and lung of LPS-induced rats. Furthermore, SBH prevented LPS-induced histological and functional alterations in the liver, kidney, heart and lung of rats. Conclusion SBH has a substantial protective role against LPS-induced CSSI in rats mediated via amelioration of inflammation, oxidative stress and NF-κB, p38 MAPK and Nrf2 signaling.
Collapse
Affiliation(s)
- Yazan Ranneh
- 1Department of Nutrition and Dietetics, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
| | - Abdah Md Akim
- 2Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
| | - Hasiah Ab Hamid
- 2Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
| | - Huzwah Khazaai
- 2Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM, 43400 Serdang, Selangor Malaysia
| | - Abdulmannan Fadel
- 3School of Food Science and Nutrition, University of Leeds, Leeds, UK
| | - Ayman M Mahmoud
- 4Physiology Division, Zoology Department, Faculty of Science, Beni-Suef University, Beni-Suef, Egypt
| |
Collapse
|
45
|
Flemingia philippinensis Flavonoids Relieve Bone Erosion and Inflammatory Mediators in CIA Mice by Downregulating NF- κB and MAPK Pathways. Mediators Inflamm 2019; 2019:5790291. [PMID: 30906224 PMCID: PMC6397971 DOI: 10.1155/2019/5790291] [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: 07/25/2018] [Revised: 11/29/2018] [Accepted: 12/30/2018] [Indexed: 12/19/2022] Open
Abstract
Background The dry root of Flemingia philippinensis has been widely used in the treatment of rheumatism, arthropathy, and osteoporosis in traditional Chinese medicine; the therapeutic effects of Flemingia philippinensis are associated with antiarthritis in traditional Chinese medicine theory. This study was undertaken to investigate the mechanism of bone erosion protection and anti-inflammatory effect of Flemingia philippinensis flavonoids (FPF) in collagen-induced arthritis (CIA) in mice. Methods Flavonoids were extracted from the dry root of Flemingia philippinensis. Collagen-induced arthritis in C57BL/6 mice was used as a rheumatoid arthritis model, and the mice were orally fed with FPF prior to induction to mimic clinical prophylactic therapy for a total of 39 days. After treatment, histology and immunohistochemistry staining were performed, and the levels of anti-collagen type II (CII) antibody and inflammatory mediators, as well as the key proteins of nuclear factor kappa-B (NF-κB) and mitogen-activated protein kinase (MAPK) pathways, were detected in the samples taken from ankle joints, plasma, and paws. Results FPF administration significantly suppressed the paw swelling and arthritic score in CIA mice. FPF reduced inflammatory infiltration and pannus formation, articular cartilage destruction and osteoclast infiltration, and the expression of MMP-9 and cathepsin K in the ankle joint. FPF inhibited plasma anti-CII antibody levels and the production of inflammatory cytokines and chemokines in CIA paws. FPF treatment suppressed the activation of NF-κB as indicated by downregulating the phosphorylation of NF-κB p65 and mitogen-activated protein kinases in CIA paws. Additionally, FPF significantly inhibited inflammation signaling by suppressing the activation of activator protein-1 subset and signal transducers and activators of transcription 3 (STAT3). Conclusions Our data suggest that FPF might be an active therapeutic agent for rheumatoid arthritis and the preventive effect of FPF on arthritis is attributable to an anti-inflammatory effect on CIA by preventing bone destruction, regulating inflammatory mediators, and suppressing NF-κB and MAPK signaling pathways.
Collapse
|
46
|
Alipour F, Vakili A, Danesh Mesgaran M, Ebrahimi H. The effect of adding ethanolic saffron petal extract (SPE) and vitamin E on growth performance, blood metabolites and antioxidant status in Baluchi male lambs. ASIAN-AUSTRALASIAN JOURNAL OF ANIMAL SCIENCES 2019; 32:1695-1704. [PMID: 31011009 PMCID: PMC6817774 DOI: 10.5713/ajas.18.0615] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/15/2018] [Accepted: 01/28/2019] [Indexed: 12/21/2022]
Abstract
Objective This study investigated the effects of the administration of ethanolic saffron petal extract (SPE) and vitamin E on growth performance, blood metabolites and antioxidant status in Baluchi lambs. Methods Thirty-two Baluchi male lambs (35.22±5.75 kg) were randomly divided into 4 groups. The 1st (control), 2nd (ISPE) and 3rd (Vit. E) groups were respectively injected subcutaneously with either physiological saline (5 ml), SPE (25 mg/kg BW) or DL-α-tocopheryl acetate (225 IU) once a week. An oral dose of SPE (500 mg/kg BW) was also administered to the 4th group (OSPE). Feed intake and body weight were measured for 42 days and blood samples were taken on days 1, 14, 28 and 42. The lambs were slaughtered and tissue samples were taken. Results Growth performance and many blood metabolites were not affected (p>0.05) by the treatments. Cholesterol of plasma in the ISPE and Vit. E groups was similar and less (p<0.01) than both the OSPE and control groups. Although there was no significant difference between the control and other groups for plasma triglyceride, the ISPE group showed lower (p<0.05) triglyceride than the OSPE and Vit. E groups. The highest (p<0.01) plasma glutathione peroxidase (GPx) was detected in the OSPE group, while the ISPE and Vit. E groups showed higher (p<0.01) superoxide dismutase (SOD) of plasma than the control. Malondialdehyde of plasma in the ISPE group was lower (p<0.05) than the OSPE. No differences (p>0.05) were observed among the groups for antioxidant status of both of the longissimus dorsi muscle and liver. However, the activity of GPx in the kidney and heart, as well as SOD activity in the kidney, were influenced (p≤0.01) by the treatments. Conclusion Adding ethanolic SPE improved antioxidant status and lowered lipids oxidation in lambs. The SPE and vitamin E demonstrated similar effects on antioxidant status in lambs.
Collapse
Affiliation(s)
- Fereshteh Alipour
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Alireza Vakili
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Mohsen Danesh Mesgaran
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| | - Hadi Ebrahimi
- Department of Animal Science, Faculty of Agriculture, Ferdowsi University of Mashhad, Mashhad 9177948974, Iran
| |
Collapse
|
47
|
Wang ZY, Qin LH, Zhang WG, Zhang PX, Jiang BG. Qian-Zheng-San promotes regeneration after sciatic nerve crush injury in rats. Neural Regen Res 2019; 14:683-691. [PMID: 30632509 PMCID: PMC6352607 DOI: 10.4103/1673-5374.247472] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Qian-Zheng-San, a traditional Chinese prescription consisting of Typhonii Rhizoma, Bombyx Batryticatus, Scorpio, has been found to play an active therapeutic role in central nervous system diseases. However, it is unclear whether Qian-Zheng-San has therapeutic value for peripheral nerve injury. Therefore, we used Sprague-Dawley rats to investigate this. A sciatic nerve crush injury model was induced by clamping the right sciatic nerve. Subsequently, rats in the treatment group were administered 2 mL Qian-Zheng-San (1.75 g/mL) daily as systemic therapy for 1, 2, 4, or 8 weeks. Rats in the control group were not administered Qian-Zheng-San. Rats in sham group did not undergo surgery and systemic therapy. Footprint analysis was used to assess nerve motor function. Electrophysiological experiments were used to detect nerve conduction function. Immunofluorescence staining was used to assess axon counts and morphological analysis. Immunohistochemical staining was used to observe myelin regeneration of the sciatic nerve and the number of motoneurons in the anterior horn of the spinal cord. At 2 and 4 weeks postoperatively, the sciatic nerve function index, nerve conduction velocity, the number of distant regenerated axons and the axon diameter of the sciatic nerve increased in the Qian-Zheng-San treatment group compared with the control group. At 2 weeks postoperatively, nerve fiber diameter, myelin thickness, and the number of motor neurons in the lumbar spinal cord anterior horn increased in the Qian-Zheng-San treatment group compared with the control group. These results indicate that Qian-Zheng-San has a positive effect on peripheral nerve regeneration.
Collapse
Affiliation(s)
- Zhi-Yong Wang
- Department of Anatomy and Histo-embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Li-Hua Qin
- Department of Anatomy and Histo-embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Wei-Guang Zhang
- Department of Anatomy and Histo-embryology, School of Basic Medical Sciences, Peking University, Beijing, China
| | - Pei-Xun Zhang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| | - Bao-Guo Jiang
- Department of Trauma and Orthopedics, Peking University People's Hospital, Beijing, China
| |
Collapse
|
48
|
Skinner CM, Miousse IR, Ewing LE, Sridharan V, Cao M, Lin H, Williams DK, Avula B, Haider S, Chittiboyina AG, Khan IA, ElSohly MA, Boerma M, Gurley BJ, Koturbash I. Impact of obesity on the toxicity of a multi-ingredient dietary supplement, OxyELITE Pro™ (New Formula), using the novel NZO/HILtJ obese mouse model: Physiological and mechanistic assessments. Food Chem Toxicol 2018; 122:21-32. [PMID: 30282009 DOI: 10.1016/j.fct.2018.09.067] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2018] [Revised: 09/18/2018] [Accepted: 09/28/2018] [Indexed: 02/07/2023]
Abstract
Herbal dietary supplement (HDS)-induced hepato- and cardiotoxicity is an emerging clinical problem. In this study, we investigated the liver and heart toxicity of HDS OxyELITE-PRO™ New Formula (OEP-NF), a dietary supplement marketed for weight loss and performance enhancement that was recently withdrawn from the market. Using a novel NZO/HlLtJ obese mouse model, we demonstrated that administration of clinically relevant mouse equivalent doses (MED) of OEP-NF produced cardio- and hepatotoxic risks following both short- and long-term administration schedules. Specifically, gavaging female NZO/HlLtJ with up to 2X MED of OEP-NF resulted in 40% mortality within two weeks. Feeding mice with either 1X or 3X MED of OEP-NF for eight weeks, while not exhibiting significant effects on body weights, significantly altered hepatic gene expression, increased the number of apoptotic and mast cells in the heart and affected cardiac function. The degree of toxicity in NZO/HlLtJ mice was higher than that observed previously in non-obese CD-1 and B6C3F1 strains, suggesting that an overweight/obese condition can sensitize mice to OEP-NF. Adverse health effects linked to OEP-NF, together with a number of other hepato- and cardiotoxicity cases associated with HDS ingestion, argue strongly for introduction of quality standards and pre-marketing safety assessments for multi-ingredient HDS.
Collapse
Affiliation(s)
- Charles M Skinner
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Center for Dietary Supplement Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Isabelle R Miousse
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Laura E Ewing
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Department of Pharmacology and Toxicology, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Vijayalakshmi Sridharan
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72223, USA.
| | - Maohua Cao
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72223, USA.
| | - Haixia Lin
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - D Keith Williams
- Department of Biostatistics, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Bharathi Avula
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| | - Saqlain Haider
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| | - Amar G Chittiboyina
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| | - Ikhlas A Khan
- National Center for Natural Product Research, School of Pharmacy, University of Mississippi, University, MS, 38677, USA.
| | - Mahmoud A ElSohly
- ElSohly Laboratories, Inc. (ELI), Phyto Chemical Services, Inc. (PSI), 5 Industrial Park Drive, Oxford, MS 38655, USA.
| | - Marjan Boerma
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72223, USA; Center for Dietary Supplement Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Bill J Gurley
- Department of Pharmaceutical Sciences, University of Arkansas for Medical Sciences, Little Rock, AR, 72223, USA; Center for Dietary Supplement Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| | - Igor Koturbash
- Department of Environmental and Occupational Health, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA; Center for Dietary Supplement Research, University of Arkansas for Medical Sciences, Little Rock, AR, 72205, USA.
| |
Collapse
|
49
|
Gao YX, Zhou YY, Xie Y, Feng L, Shen SG. The complete chloroplast genome sequence of an endangered Orchidaceae species Dendrobium monilforme and its phylogenetic implications. CONSERV GENET RESOUR 2018. [DOI: 10.1007/s12686-017-0834-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
50
|
Mabou Tagne A, Marino F, Cosentino M. Tithonia diversifolia (Hemsl.) A. Gray as a medicinal plant: A comprehensive review of its ethnopharmacology, phytochemistry, pharmacotoxicology and clinical relevance. JOURNAL OF ETHNOPHARMACOLOGY 2018; 220:94-116. [PMID: 29596999 DOI: 10.1016/j.jep.2018.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 03/22/2018] [Accepted: 03/22/2018] [Indexed: 05/28/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Tithonia diversifolia (TD) is widely valued in several cultures for its medicinal properties. A comprehensive review of the current understanding of this plant species is required due to emerging concerns over its efficacy, toxicity and allergenic potential. AIM OF THE REVIEW We critically summarized the current evidence on the botany, traditional use, phytochemistry, pharmacology and safety of TD, with the view to provide perspectives for developing more attractive pharmaceuticals of plant origin, but also to lay a new foundation for further investigations on this plant. MATERIALS AND METHODS A preliminary consultation of search engines such as Web of Science, PubMed, ScienceDirect and other published/unpublished resources provided an overview of extant literature on TD. Then, we meticulously screened all titles, abstracts and full-texts to establish consistency in the application of inclusion criteria. Studies were considered for inclusion if they dealt with taxonomy, global distribution, local and traditional knowledge, phytochemistry, toxicity and biological effects. RESULTS 1856 articles were retrieved among which 168 were revised and included. Several studies conducted on cell lines and animals provided supporting evidence for some ethnomedicinal claims of extracts from TD. Short-term use of Tithonia extracts were effective and well-tolerated in animals when taken at lower doses. Both the toxic and therapeutic effects were attributed to bioactive principles naturally occurring in this species including sesquiterpene lactones, chlorogenic acid and flavonoids. CONCLUSIONS T. diversifolia is a valuable source of bioactive compounds with significant therapeutic implications and favourable safety index. However, more rigorously designed investigations are needed to recommend the whole plant or its active ingredients as a medication, and should focus on understanding the multi-target network pharmacology of the plant, clarifying the effective doses as well as identifying the potential interactions with prescribed drugs or other chemicals.
Collapse
Affiliation(s)
- Alex Mabou Tagne
- Centre for Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy.
| | - Franca Marino
- Centre for Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
| | - Marco Cosentino
- Centre for Research in Medical Pharmacology, University of Insubria, 21100 Varese, Italy
| |
Collapse
|