1
|
Yenice G, Ozkanlar S, Bolat I, Yildirim S. The Potential Ability of Betulinic Acid to Prevent Experimentally Induced Acute Pancreatitis in Rats. Basic Clin Pharmacol Toxicol 2025; 136:e70052. [PMID: 40344370 PMCID: PMC12061522 DOI: 10.1111/bcpt.70052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025]
Abstract
Acute pancreatitis (AP) is a serious pancreatic inflammatory disease that results in pancreatic enzyme activation and autodegradation. Betulinic acid (BA), a pentacyclic triterpene of natural origin that was isolated from several plants, has anti-inflammatory, immunomodulatory and antioxidant effects that can help with AP. With this study, we aimed to investigate the potential positive effects of BA on l-arginine-induced AP. A total of 24 male rats were divided into four groups (control, BA, AP and BA + AP). Animals in the BA group were given BA 50 mg/kg/day for 7 days. AP was induced by administering two doses of 250-mg/100-g l-arginine to animals in the AP group. The animals in the BA + AP group were administered 50-mg/kg/day BA (gavage) for 7 days and two doses of 250-mg/100-g l-arginine on the seventh day. BA pretreatment inhibited the increased lipase activity caused by AP and showed protective activity against oxidative damage to pancreatic tissue. It decreased the severity of inflammation by suppressing the release of pro-inflammatory cytokines while increasing the level of the anti-inflammatory cytokine IL-10. It showed a protective effect on pancreatic tissue by inhibiting tumour necrosis factor (TNF-α) and Bax expression. The findings of the study show that BA exhibits multifaceted protective activity in experimental AP induced with l-arginine.
Collapse
Affiliation(s)
- Guler Yenice
- Faculty of Veterinary Medicine, Department of Animal Nutrition and Nutritional DisordersAtaturk UniversityErzurumTurkey
| | - Seckin Ozkanlar
- Faculty of Veterinary Medicine, Department of BiochemistryAtaturk UniversityErzurumTurkey
| | - Ismail Bolat
- Faculty of Veterinary Medicine, Department of PathologyAtaturk UniversityErzurumTurkey
| | - Serkan Yildirim
- Faculty of Veterinary Medicine, Department of PathologyAtaturk UniversityErzurumTurkey
| |
Collapse
|
2
|
Lao XY, Sun YL, Zhao ZJ, Liu J, Ruan XF. Pharmacological effects of betulinic acid and its protective mechanisms on the cardiovascular system. Fitoterapia 2025; 183:106561. [PMID: 40288588 DOI: 10.1016/j.fitote.2025.106561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/09/2025] [Accepted: 04/18/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND Betulinic acid (BA), a pentacyclic triterpenoid saponin widely found in plants, has attracted attention for its diverse pharmacological activities. Recent studies highlight its cardioprotective potential, promoting its relevance in cardiovascular research. AIM OF THE REVIEW This review summarizes BA's physicochemical properties, structure-activity relationships, natural sources, and synthesis strategies. It further discusses its pharmacokinetics and toxicity to evaluate its drug development potential, with emphasis on cardioprotective effects and related signaling pathways. METHODS Literature was collected from databases such as PubMed and Web of Science, focusing on studies addressing BA's chemical characteristics, biological activities, pharmacokinetics, and cardiovascular relevance. RESULTS BA exerts cardioprotective effects via multiple signaling pathways, including NRF2, NF-κB, MAPK, and NFAT. These contribute to its antioxidant, anti-inflammatory, anti-apoptotic, and anti-proliferative actions, as well as its enhancement of endothelial function through nitric oxide signaling. BA also reduces lipid accumulation. Combined with favorable physicochemical properties and synthetic accessibility, these findings support BA as a promising multifunctional lead compound in cardiovascular pharmacology. CONCLUSION BA shows strong potential as a cardioprotective natural compound. Although further research is needed to validate its clinical efficacy and safety, its multi-target actions and structural versatility provide a solid basis for development in cardiovascular drug discovery.
Collapse
Affiliation(s)
- Xu Yuan Lao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Yuan Long Sun
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| | - Zhe Jun Zhao
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Jie Liu
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Xiao Fen Ruan
- Cardiovascular Research Institute of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China; Cardiovascular Department of Traditional Chinese Medicine, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China.
| |
Collapse
|
3
|
He J, Huang C, Kong L, Huang Y, Ou Z, Yang M, Wu J, Yang Y, Yao H, Yi J, Liu S. Betulinic acid mitigates lipopolysaccharide-induced intestinal injury of weaned piglets through modulation of the mitochondrial quality control. Int Immunopharmacol 2025; 148:114097. [PMID: 39827669 DOI: 10.1016/j.intimp.2025.114097] [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: 11/16/2024] [Revised: 01/05/2025] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Intestinal injury of weaned piglets often leads to reduced immunity, diarrhea and growth retardation, resulting in significant economic losses to agriculture. Betulinic acid (BA) is a natural plant-derived active ingredient with multiple pharmacological activities including immune modulation and anti-inflammatory. This study was aimed to investigate the potential mechanism that BA as a feed additive mitigated lipopolysaccharide (LPS)-induced intestinal injury in piglets. The results indicated that BA pretreatment improved the morphology and structure of the intestine, enhanced intestinal mucosal barrier function, and activated the PPAR signaling pathway to reduce the mRNA levels of intestinal CD40 and CXCL13. Meanwhile, BA pretreatment improved the LPS-induced disruption of intestinal microbiota by increasing the abundance of the Firmicutes and decreasing the abundance of the Bacteroidota and Proteobacteria. Furthermore, BA pretreatment activated the AMPK/SIRT1/PGC-1α signaling pathway to enhance mitochondrial biogenesis, restored a balance to mitochondrial dynamics, and modulated the PINK1/Parkin, BNIP3 and FUNDC1 signaling pathways to activate mitophagy, thereby alleviating LPS-induced intestinal injury. Overall, the present study elucidated that dietary supplementation with BA could alleviate LPS-induced intestinal injury in weaned piglets by regulating mitochondrial quality control, which provided a novel approach for alleviating intestinal stress in weaned piglets.
Collapse
Affiliation(s)
- Jiayu He
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Chunlin Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - You Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zhaoping Ou
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Mingqi Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jiao Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yu Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Huan Yao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Shuiping Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
4
|
Wu J, Li J, Wu Y, Yang M, Chen Y, Wang N, Wang J, Yuan Z, Yi J, Yang C. Betulinic acid mitigates zearalenone-induced liver injury by ERS/MAPK/Nrf2 signaling pathways in mice. Food Chem Toxicol 2023; 177:113811. [PMID: 37179046 DOI: 10.1016/j.fct.2023.113811] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/29/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023]
Abstract
Zearalenone (ZEA) is a mycotoxin commonly found in cereals and feedstuffs, which can induce oxidative stress and inflammation to cause liver damage in humans and animals. Betulinic acid (BA) is extracted from pentacyclic triterpenoids of many natural plants and has anti-inflammatory, and anti-oxidation biological activities in many studies. However, the protective effect of BA on liver injury induced by ZEA has not been reported. Therefore, this study aims to explore the protective effect of BA on ZEA-induced liver injury and its possible mechanism. In the mice experiment, ZEA exposure increased the liver index and caused histopathological impairment, oxidative damage, hepatic inflammatory responses, and increased hepatocyte apoptosis. However, when combined with BA, it could inhibit the production of ROS, up-regulate the proteins expression of Nrf2 and HO-1 and down-regulate the expression of Keap1, and alleviate oxidative damage and inflammation in the liver of mice. In addition, BA could alleviate ZEA-induced apoptosis and liver injury in mice by inhibiting the endoplasmic reticulum stress (ERS) and MAPK signaling pathways. In conclusion, this study revealed the protective effect of BA on the hepatotoxicity of ZEA for the first time, providing a new perspective for the development of ZEA antidote and the application of BA.
Collapse
Affiliation(s)
- Jing Wu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - Jiayan Li
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - You Wu
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - Mengran Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - Yunqin Chen
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - Naidong Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Provincial Key Laboratory of Protein Engineering in Animal Vaccines, Laboratory of Functional Proteomics, Research Center of Reverse Vaccinology, Changsha, 410128, China
| | - Ji Wang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - Zhihang Yuan
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China
| | - Jine Yi
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China.
| | - Chenglin Yang
- College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China; Hunan Engineering Research Center of Livestock and Poultry Health Care, Hunan Agricultural University, Changsha, 410128, China.
| |
Collapse
|
5
|
Alruhaimi RS. Betulinic acid protects against cardiotoxicity of the organophosphorus pesticide chlorpyrifos by suppressing oxidative stress, inflammation, and apoptosis in rats. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2023; 30:51180-51190. [PMID: 36808036 DOI: 10.1007/s11356-023-25917-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Accepted: 02/09/2023] [Indexed: 04/16/2023]
Abstract
The widespread application of organophosphorus (OP) pesticides can affect the environment as well as the animal and human health. Chlorpyrifos (CPF) is a broad-spectrum OP pesticide used in agriculture and can cause several toxic effects in which oxidative stresses and inflammation play a key role. This study aimed to evaluate the protective activity of betulinic acid (BA), an antioxidant and anti-inflammatory pentacyclic triterpene, against CPF cardiotoxicity in rats. The rats were divided into four groups. CPF (10 mg/kg) and BA (25 mg/kg) were orally administered for 28 days, and blood and heart samples were collected. CPF-administered rats showed an increase in serum cardiac troponin I (cTnI), creatine kinase (CK)-MB, and lactate dehydrogenase (LDH), accompanied with multiple myocardial tissue alterations. Lipid peroxidation (LPO), nitric oxide (NO), nuclear factor-kappaB (NF-κB), interleukin (IL)-6, IL-1β, and tumor necrosis factor (TNF)-α were increased, and antioxidant were decrease in CPF-administered rats. BA ameliorated cardiac function markers and tissue injury, decreased LPO, NO, NF-κB, and proinflammatory cytokines, and increased antioxidants. In addition, BA decreased proapoptosis markers, and increased B-cell lymphoma (Bcl)-2, IL-10, Nrf2, and HO-1 in the heart of CPF-treated rats. In conclusion, BA protected against cardiotoxicity in CPF-administered rats by mitigating oxidative stress, inflammation, and apoptosis, and enhanced Nrf2 and antioxidants.
Collapse
Affiliation(s)
- Reem S Alruhaimi
- Department of Biology, College of Science, Princess Nourah bint Abdulrahman University, Riyadh, 11671, Saudi Arabia.
| |
Collapse
|
6
|
Transcutaneous Electrical Acupoint Stimulation Pretreatment Alleviates Cerebral Ischemia-Reperfusion Injury in Rats by Modulating Microglia Polarization and Neuroinflammation Through Nrf2/HO-1 Signaling Pathway. Neurochem Res 2023; 48:862-873. [PMID: 36357746 DOI: 10.1007/s11064-022-03797-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/29/2022] [Accepted: 10/15/2022] [Indexed: 11/12/2022]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) may lead to severe disability even death, but the strategies for prevention and treatment are still limited. Transcutaneous electrical acupoint stimulation (TEAS) has been reported to have a significant neuroprotection against CIRI, but the underlying mechanisms remain obscure. In this study, we established a focal cerebral ischemia-reperfusion model in male Sprague-Dawley rats. TEAS pretreatment was applied to Baihui (GV20), Sanyinjiao (SP6) and Zusanli (ST36) acupoints for 5 consecutive days before CIRI. After 24 h reperfusion, the brain damage was assessed using Zea-Longa score, brain water content (BWC) and infarct volume. Meanwhile, the number of activated microglia and the TNF-α were detected by immunofluorescence and ELISA respectively. Moreover, Western Blot and RT-qPCR were conducted to detect the proteins and mRNA expressions of Nrf2, HO-1, iNOS and Arg-1. We found that TEAS pretreatment significantly reduced Longa score, BWC, infarct volume and the number of activated microglia. Besides, TEAS pretreatment increased Nrf2 and HO-1 levels, while lowered the expression of TNF-α. Subsequently, we also discovered that the microglia M1 phenotype maker iNOS decreased and the M2 maker Arg-1 increased after TEAS pretreatment. However, these effects of TEAS pretreatment were markedly eliminated by brusatol. These findings clearly suggested that TEAS pretreatment exerted neuroprotection against CIRI, which might be related to modulating microglia polarization and neuroinflammation via Nrf2/HO-1 pathway.
Collapse
|
7
|
Sun N, Yang T, Tang Y, Zhao Y, Wang H, Zhao S, Tan H, Li L, Fan H. Lycopene Alleviates Chronic Stress-Induced Liver Injury by Inhibiting Oxidative Stress-Mediated Endoplasmic Reticulum Stress Pathway Apoptosis in Rats. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:14414-14426. [PMID: 36318656 DOI: 10.1021/acs.jafc.2c06650] [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: 06/16/2023]
Abstract
The liver is the major organ of metabolism and is extremely vulnerable to chronic stress. Lycopene (LYC) is a natural carotenoid with potent antioxidant and chronic disease potential. However, whether LYC protects against chronic restraint stress (CRS)-induced liver injury and the underlying mechanisms remain unclear. In this study, rats were restrained for 21 days for 6 h per day, with or without gavage of LYC (10 mg/kg). Serum ALT (85.99 ± 4.07 U/L) and AST (181.78 ± 7.35 U/L) and scores of liver injury were significantly increased in the CRS group. LYC significantly promoted the nuclear translocation of Nrf2, elevated the expression of antioxidant genes, and attenuated reactive oxygen radicals (ROS) levels within the liver. Cellular thermal shift assay (CETSA) and molecular docking results indicated that LYC competitively binds to Keap1 with the lowest molecule affinity of -9.0 kcal/mol. Moreover, LYC significantly relieved the hepatic endoplasmic reticulum swelling and decreased the expression of endoplasmic reticulum stress (ERS) hallmarks like GRP78, CHOP, and cleaved caspase-12. Meanwhile, LYC also mitigated CRS-induced hepatocyte apoptosis. Interestingly, every other day, the intraperitoneal injection of the Nrf2 inhibitor brusatol (0.4 mg/kg) significantly counteracted the protective effect of LYC. In conclusion, LYC protects against CRS-induced liver injury by activating the Nrf2 signaling pathway, scavenging ROS, and further attenuating ERS-associated apoptosis pathways.
Collapse
Affiliation(s)
- Ning Sun
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Tianyuan Yang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yulin Tang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Yuan Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Hui Wang
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Shuping Zhao
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Haoyang Tan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Lin Li
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| | - Honggang Fan
- Heilongjiang Key Laboratory for Laboratory Animals and Comparative Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China
| |
Collapse
|
8
|
Recent Advances Regarding the Molecular Mechanisms of Triterpenic Acids: A Review (Part II). Int J Mol Sci 2022; 23:ijms23168896. [PMID: 36012159 PMCID: PMC9408012 DOI: 10.3390/ijms23168896] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Revised: 08/04/2022] [Accepted: 08/08/2022] [Indexed: 12/18/2022] Open
Abstract
Triterpenic acids are a widespread class of phytocompounds which have been found to possess valuable therapeutic properties such as anticancer, anti-inflammatory, hepatoprotective, cardioprotective, antidiabetic, neuroprotective, lipolytic, antiviral, and antiparasitic effects. They are a subclass of triterpenes bearing a characteristic lipophilic structure that imprints unfavorable in vivo properties which subsequently limit their applications. The early investigation of the mechanism of action (MOA) of a drug candidate can provide valuable information regarding the possible side effects and drug interactions that may occur after administration. The current paper aimed to summarize the most recent (last 5 years) studies regarding the MOA of betulinic acid, boswellic acid, glycyrrhetinic acid, madecassic acid, moronic acid, and pomolic acid in order to provide scientists with updated and accessible material on the topic that could contribute to the development of future studies; the paper stands as the sequel of our previously published paper regarding the MOA of triterpenic acids with therapeutic value. The recent literature published on the topic has highlighted the role of triterpenic acids in several signaling pathways including PI3/AKT/mTOR, TNF-alpha/NF-kappa B, JNK-p38, HIF-α/AMPK, and Grb2/Sos/Ras/MAPK, which trigger their various biological activities.
Collapse
|
9
|
Lin X, Zhu L, Gao X, Kong L, Huang Y, Zhao H, Chen Y, Wen L, Li R, Wu J, Yuan Z, Yi J. Ameliorative effect of betulinic acid against zearalenone exposure triggers testicular dysfunction and oxidative stress in mice via p38/ERK MAPK inhibition and Nrf2-mediated antioxidant defense activation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113561. [PMID: 35489292 DOI: 10.1016/j.ecoenv.2022.113561] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 04/14/2022] [Accepted: 04/20/2022] [Indexed: 06/14/2023]
Abstract
Zearalenone (ZEA) is a nonsteroidal estrogenic mycotoxin, which mainly contaminates grains and has estrogen-like effects on the reproductive system. Betulinic acid (BA), a natural lupane-type pentacyclic triterpene, has anti-oxidative and anti-inflammatory properties. This study aimed to investigate whether BA alleviates ZEA-induced testicular damage and explore the possible mechanism. Here, BA ameliorated testicular damage by mitigating the disordered arrangement of seminiferous tubules, the exfoliation of lumen cells, and the increase of cell apoptosis caused by ZEA. Meanwhile, BA alleviated ZEA-triggered testicular damage by restoring hormone levels and sperm motility, and reconstructing the blood-testis-barrier. Moreover, BA alleviated ZEA-exposed testicular oxidative stress by activating Nrf2 pathway. Furthermore, BA moderated ZEA-evoked testicular inflammation by inhibiting p38/ERK MAPK pathway. Overall, our results revealed that BA has a therapeutic protective effect on ZEA-induced testicular injury and oxidative stress via p38/ERK MAPK inhibition and Nrf2-mediated antioxidant defense activation, which provides a viable alternative to alleviate ZEA-induced male reproductive toxicology.
Collapse
Affiliation(s)
- Xing Lin
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Lijuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Xinyu Gao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - You Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Haoqiang Zhao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yazhi Chen
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Lixin Wen
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Rongfang Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
10
|
He E, Ma Y, Kong L, Huang Y, Huang C, Yang W, Yi J, Zhu L. Suppression of endoplasmic reticulum stress-associated pathways and hepatocyte apoptosis participates in the attenuation of betulinic acid on alcohol-provoked liver injury in mice. Food Funct 2022; 13:11489-11502. [DOI: 10.1039/d2fo01042a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
BA protects against alcohol-induced liver damage through the alleviation of oxidative stress and suppression of ERS-induced apoptosis.
Collapse
Affiliation(s)
- Enqi He
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- College of Food Science and Technology, Hunan Agricultural University, Changsha, 410128, China
| | - Yurong Ma
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
- Changsha University of Science & Technology, Changsha 410114, China
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - You Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Chunlin Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Wenjiang Yang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| | - Lijuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha, 410128, China
| |
Collapse
|
11
|
Tuli HS, Sak K, Gupta DS, Kaur G, Aggarwal D, Chaturvedi Parashar N, Choudhary R, Yerer MB, Kaur J, Kumar M, Garg VK, Sethi G. Anti-Inflammatory and Anticancer Properties of Birch Bark-Derived Betulin: Recent Developments. PLANTS (BASEL, SWITZERLAND) 2021; 10:2663. [PMID: 34961132 PMCID: PMC8705846 DOI: 10.3390/plants10122663] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Revised: 11/26/2021] [Accepted: 11/29/2021] [Indexed: 05/03/2023]
Abstract
Birch tree bark-derived betulin has attracted scientific interest already for several centuries, being one of the first natural products identified from plants. However, the cellular events regulated by betulin and precise molecular mechanisms under these processes have been begun to be understood only recently. Today, we know that betulin can exert important anticancer activities through modulation of diverse cellular pathways. In this review article, betulin-regulated molecular signaling is unraveled and presented with a special focus on its participation in anti-inflammatory processes, especially by modulating nuclear factor-κB (NF-κB), prostaglandin/COX, and nuclear factor erythroid2-related factor 2 (Nrf2)-mediated cascades. By regulating these diverse pathways, betulin can not only affect the development and progression of different cancers, but also enhance the antitumor action of traditional therapeutic modalities. It is expected that by overcoming the low bioavailability of betulin by encapsulating it into nanocarriers, this promising natural compound may provide novel possibilities for targeting inflammation-related cancers.
Collapse
Affiliation(s)
- Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India; (D.A.); (N.C.P.); (R.C.)
| | | | - Dhruv Sanjay Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s NMIMS, Mumbai 40056, Maharashtra, India; (D.S.G.); (G.K.)
| | - Ginpreet Kaur
- Shobhaben Pratapbhai Patel School of Pharmacy and Technology Management, SVKM’s NMIMS, Mumbai 40056, Maharashtra, India; (D.S.G.); (G.K.)
| | - Diwakar Aggarwal
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India; (D.A.); (N.C.P.); (R.C.)
| | - Nidarshana Chaturvedi Parashar
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India; (D.A.); (N.C.P.); (R.C.)
| | - Renuka Choudhary
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala 133207, Haryana, India; (D.A.); (N.C.P.); (R.C.)
| | - Mukerrem Betul Yerer
- Department of Pharmacology, Faculty of Pharmacy, Erciyes University, Kayseri 38039, Turkey;
| | - Jagjit Kaur
- ARC Centre of Excellence in Nanoscale Biophotonics (CNBP), Graduate School of Biomedical Engineering, Faculty of Engineering, The University of New South Wales, Sydney 2052, Australia;
| | - Manoj Kumar
- Department of Chemistry, Maharishi Markandeshwar University, Sadopur 134007, Haryana, India;
| | - Vivek Kumar Garg
- Department of Medical Laboratory Technology, University Institute of Applied Health Sciences, Chandigarh University, Gharuan, Mohali 140413, Punjab, India;
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
| |
Collapse
|
12
|
Ou Z, Zhu L, Huang C, Ma C, Kong L, Lin X, Gao X, Huang L, Wen L, Liang Z, Yuan Z, Wu J, Yi J. Betulinic acid attenuates cyclophosphamide-induced intestinal mucosa injury by inhibiting the NF-κB/MAPK signalling pathways and activating the Nrf2 signalling pathway. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 225:112746. [PMID: 34482064 DOI: 10.1016/j.ecoenv.2021.112746] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Revised: 08/29/2021] [Accepted: 08/31/2021] [Indexed: 06/13/2023]
Abstract
Betulinic acid (BA), a pentacyclic triterpenoid, has been associated with several biological effects, such as antioxidant, anti-inflammatory and antiviral activities. Previous studies have demonstrated that BA has the ability to alleviate intestinal mucosal damage, however, the potential mechanism associated with the effect has not been reported. This study aimed to investigate the possible protective mechanism of BA against cyclophosphamide (CYP)-induced intestinal mucosal damage. Here, we found that BA pretreatment prevented intestinal mucosal barrier dysfuction from CYP-challenged mice by repairing the intestinal physical, chemical, and immune barriers. Moreover, BA treatment suppressed the CYP-induced oxidative stress by activating the nuclear factor erythroid 2 [NF-E2]-related factor (Nrf2) pathway blocked reactive oxygen species (ROS) accumulation. In addition, BA inhibited CYP-triggered intestinal inflammation through down-regulating the nuclear transcription factor kappa B (NF-κB)/mitogen-activating protein kinase (MAPK) pathways. Furthermore, BA pretreatment reduced intestinal apoptosis by blocking ROS-activated mitochondrial apoptotic pathway. Overall, the current study demonstrated the protective effect of BA against CYP-caused intestinal mucosal damage by regulating the Nrf2 and NF-κB/MAPK signalling pathways, which may provide new therapeutic targets to attenuate intestinal impairment and maintain intestinal health.
Collapse
Affiliation(s)
- Zhaoping Ou
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Lijuan Zhu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Chenglong Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Chaoyang Ma
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Li Kong
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Xing Lin
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Xinyu Gao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Lin Huang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Lixin Wen
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zengenni Liang
- Department of Hunan Agricultural Product Processing Institute, Changsha 410128, China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, Colleges of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China.
| |
Collapse
|
13
|
Cui N, Liang Y, Wang J, Liu B, Wei B, Zhao Y. Minocycline attenuates oxidative and inflammatory injury in a intestinal perforation induced septic lung injury model via down-regulating lncRNA MALAT1 expression. Int Immunopharmacol 2021; 100:108115. [PMID: 34562841 DOI: 10.1016/j.intimp.2021.108115] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Revised: 08/19/2021] [Accepted: 08/28/2021] [Indexed: 12/21/2022]
Abstract
BACKGROUND Oxidative stress and inflammatory responses play an important role in acute lung injury (ALI). Although minocycline (MINO) has anti-inflammatory effects and is a promising candidate in treating inflammatory diseases, the effect of MINO on ALI during sepsis is still unclear. METHODS In the present study, a mouse model with intestinal perforation was established. C57BL/6 mice received cecal ligation and puncture (CLP) to induce sepsis-associated ALI. MINO was used to treat the mice via intraperitoneal injection at different doses (negative control, 20 mg/kg, 50 mg/kg and 100 mg/kg, respectively) 24 h after CLP. The severity of lung injury was evaluated by pathological examination, and lung wet / dry weight ratio was calculated to evaluate the severity of pulmonary edema. The changes of TNF-α, IL-1β, IL-6, PGE2, MDA, NF-κB, Nrf2, Keap1 and lncRNA MALAT1 levels in lung tissues of the mice were detected with ELISA, chemical colorimetry, Western blot or qRT-PCR. RESULTS MINO ameliorated the lung edema and lung injury of the mice induced by CLP in a dose-dependent manner. MINO administration could significantly down-regulate expressions of TNF-α, IL-6, IL-1β, PGE2 and MDA in lung tissues of the mice. Mechanistically, MINO exerted the effects of anti-inflammation and anti-oxidative stress through down-regulating the expression of MALAT1 and regulating Nrf2/Keap1 and NF-κB signaling pathways. CONCLUSION MINO represses oxidative stress and inflammatory response during sepsis-induced ALI via down-regulating MALAT1 expression, and it has the potential to treat septic ALI.
Collapse
Affiliation(s)
- Ning Cui
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430070, China
| | - Yong Liang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing 100043, China
| | - Junyu Wang
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing 100043, China
| | - Bo Liu
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing 100043, China
| | - Bing Wei
- Emergency Medicine Clinical Research Center, Beijing Chao-Yang Hospital, Capital Medical University & Beijing Key Laboratory of Cardiopulmonary Cerebral Resuscitation, Beijing 100043, China
| | - Yu Zhao
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province 430070, China.
| |
Collapse
|
14
|
Musayeva A, Unkrig JC, Zhutdieva MB, Manicam C, Ruan Y, Laspas P, Chronopoulos P, Göbel ML, Pfeiffer N, Brochhausen C, Daiber A, Oelze M, Li H, Xia N, Gericke A. Betulinic Acid Protects from Ischemia-Reperfusion Injury in the Mouse Retina. Cells 2021; 10:cells10092440. [PMID: 34572088 PMCID: PMC8469383 DOI: 10.3390/cells10092440] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 09/10/2021] [Accepted: 09/14/2021] [Indexed: 12/16/2022] Open
Abstract
Ischemia/reperfusion (I/R) events are involved in the pathophysiology of numerous ocular diseases. The purpose of this study was to test the hypothesis that betulinic acid protects from I/R injury in the mouse retina. Ocular ischemia was induced in mice by increasing intraocular pressure (IOP) to 110 mm Hg for 45 min, while the fellow eye served as a control. One group of mice received betulinic acid (50 mg/kg/day p.o. once daily) and the other group received the vehicle solution only. Eight days after the I/R event, the animals were killed and the retinal wholemounts and optic nerve cross-sections were prepared and stained with cresyl blue or toluidine blue, respectively, to count cells in the ganglion cell layer (GCL) of the retina and axons in the optic nerve. Retinal arteriole responses were measured in isolated retinas by video microscopy. The levels of reactive oxygen species (ROS) were assessed in retinal cryosections and redox gene expression was determined in isolated retinas by quantitative PCR. I/R markedly reduced cell number in the GCL and axon number in the optic nerve of the vehicle-treated mice. In contrast, only a negligible reduction in cell and axon number was observed following I/R in the betulinic acid-treated mice. Endothelial function was markedly reduced and ROS levels were increased in retinal arterioles of vehicle-exposed eyes following I/R, whereas betulinic acid partially prevented vascular endothelial dysfunction and ROS formation. Moreover, betulinic acid boosted mRNA expression for the antioxidant enzymes SOD3 and HO-1 following I/R. Our data provide evidence that betulinic acid protects from I/R injury in the mouse retina. Improvement of vascular endothelial function and the reduction in ROS levels appear to contribute to the neuroprotective effect.
Collapse
Affiliation(s)
- Aytan Musayeva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
- Laboratory of Corneal Immunology, Transplantation and Regeneration, Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA 02114, USA
| | - Johanna C. Unkrig
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Mayagozel B. Zhutdieva
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Caroline Manicam
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Yue Ruan
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Panagiotis Laspas
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Panagiotis Chronopoulos
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Marie L. Göbel
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Norbert Pfeiffer
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
| | - Christoph Brochhausen
- Institute of Pathology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany;
- Institute of Pathology, University of Regensburg, Franz-Josef-Strauß-Allee 11, 93053 Regensburg, Germany
| | - Andreas Daiber
- Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Building 605, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.D.); (M.O.)
| | - Matthias Oelze
- Department of Cardiology 1, Laboratory of Molecular Cardiology, University Medical Center, Johannes Gutenberg University Mainz, Building 605, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.D.); (M.O.)
| | - Huige Li
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (H.L.); (N.X.)
| | - Ning Xia
- Department of Pharmacology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (H.L.); (N.X.)
| | - Adrian Gericke
- Department of Ophthalmology, University Medical Center, Johannes Gutenberg University Mainz, Langenbeckstrasse 1, 55131 Mainz, Germany; (A.M.); (J.C.U.); (M.B.Z.); (C.M.); (Y.R.); (P.L.); (P.C.); (M.L.G.); (N.P.)
- Correspondence: ; Tel.: +49-613-117-8276
| |
Collapse
|
15
|
Song X, Li L, Zhao Y, Song Y. Down-regulation of long non-coding RNA XIST aggravates sepsis-induced lung injury by regulating miR-16-5p. Hum Cell 2021; 34:1335-1345. [PMID: 33978928 PMCID: PMC8114023 DOI: 10.1007/s13577-021-00542-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 04/27/2021] [Indexed: 11/30/2022]
Abstract
This study aims to explain the role and related mechanisms of long non-coding RNA (lncRNA) X inactive specific transcript (XIST) in sepsis-induced acute lung injury (ALI). The in vivo septic models and in vitro septic model were established. In animal models, the lung injury of the rats was evaluated after XIST was overexpressed. In cell models, the effects of XIST and microRNA (miR)-16-5p on ALI was detected by MTT assay, Western blot and ELISA. The interaction between XIST and miR-16-5p was investigated by bioinformatics analysis, dual-luciferase reporter assay, RIP assay and RNA pull-down assay. We found that XIST expression was down-regulated in lung tissues of septic rats and lipopolysaccharide-stimulated cells, while the expression of miR-16-5p was up-regulated. Down-regulation of XIST significantly promoted pulmonary edema, increased the levels of TNF-α, IL-1β and malondialdehyde, inhibited the cell viability and decreased the level of superoxide dismutase. Mechanistically, it was confirmed that XIST could sponge miR-16-5p, and thus repress its expression, and the transfection of miR-16-5p mimics could reverse the effects of XIST over-expression in the cell model. Collectively, it is concluded that XIST reduces sepsis-induced ALI via regulating miR-16-5p.
Collapse
Affiliation(s)
- Xiaofei Song
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Medical College of Henan University, Weiwu Road No. 7, Zhengzhou, 450003 Henan Province China
| | - Linyu Li
- Department of Scientific Research, Sanquan College of Xinxiang Medical University, Xinxiang, 453003 China
| | - Yaying Zhao
- Department of Disinfection Supply Center, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Medical College of Henan University, Zhengzhou, 450003 China
| | - Yucheng Song
- Department of Gastrointestinal Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Medical College of Henan University, Weiwu Road No. 7, Zhengzhou, 450003 Henan Province China
| |
Collapse
|
16
|
Ghiulai R, Roşca OJ, Antal DS, Mioc M, Mioc A, Racoviceanu R, Macaşoi I, Olariu T, Dehelean C, Creţu OM, Voicu M, Şoica C. Tetracyclic and Pentacyclic Triterpenes with High Therapeutic Efficiency in Wound Healing Approaches. Molecules 2020; 25:E5557. [PMID: 33256207 PMCID: PMC7730621 DOI: 10.3390/molecules25235557] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/23/2020] [Accepted: 11/24/2020] [Indexed: 02/06/2023] Open
Abstract
Wounds are among the most common skin conditions, displaying a large etiological diversity and being characterized by different degrees of severity. Wound healing is a complex process that involves multiple steps such as inflammation, proliferation and maturation and ends with scar formation. Since ancient times, a widely used option for treating skin wounds are plant- based treatments which currently have become the subject of modern pharmaceutical formulations. Triterpenes with tetracyclic and pentacyclic structure are extensively studied for their implication in wound healing as well as to determine their molecular mechanisms of action. The current review aims to summarize the main results of in vitro, in vivo and clinical studies conducted on lupane, ursane, oleanane, dammarane, lanostane and cycloartane type triterpenes as potential wound healing treatments.
Collapse
Affiliation(s)
- Roxana Ghiulai
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (R.G.); (O.J.R.); (M.M.); (R.R.); (C.Ş.)
| | - Oana Janina Roşca
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (R.G.); (O.J.R.); (M.M.); (R.R.); (C.Ş.)
| | - Diana Simona Antal
- Department of Pharmaceutical Botany, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania
| | - Marius Mioc
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (R.G.); (O.J.R.); (M.M.); (R.R.); (C.Ş.)
| | - Alexandra Mioc
- Department of Anatomy, Physiology, Pathophysiology, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania;
| | - Roxana Racoviceanu
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (R.G.); (O.J.R.); (M.M.); (R.R.); (C.Ş.)
| | - Ioana Macaşoi
- Department of Toxicology, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd EftimieMurgu Sq., 300041 Timişoara, Romania; (I.M.); (C.D.)
| | - Tudor Olariu
- Department of Organic Chemistry, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd EftimieMurgu Sq., 300041 Timişoara, Romania;
| | - Cristina Dehelean
- Department of Toxicology, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd EftimieMurgu Sq., 300041 Timişoara, Romania; (I.M.); (C.D.)
| | - Octavian Marius Creţu
- Department of Surgery, Faculty of Medicine, Victor Babeş University of Medicine and Pharmacy, 2nd EftimieMurgu Sq., 300041 Timişoara, Romania;
| | - Mirela Voicu
- Department of Pharmacology, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd EftimieMurgu Sq., 300041 Timişoara, Romania
| | - Codruţa Şoica
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Victor Babeş University of Medicine and Pharmacy, 2nd Eftimie Murgu Sq., 300041 Timişoara, Romania; (R.G.); (O.J.R.); (M.M.); (R.R.); (C.Ş.)
| |
Collapse
|
17
|
Zhirong Z, Qiaojian Z, Chunjing X, Shengchen W, Jiahe L, Zhaoyi L, Shu L. Methionine selenium antagonizes LPS-induced necroptosis in the chicken liver via the miR-155/TRAF3/MAPK axis. J Cell Physiol 2020; 236:4024-4035. [PMID: 33151563 DOI: 10.1002/jcp.30145] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Revised: 10/05/2020] [Accepted: 10/26/2020] [Indexed: 12/15/2022]
Abstract
Organic selenium has antioxidation and disease treatment effects. To explore the mechanisms of how methionine selenium alleviates necroptosis in the liver and whether this process is related to microRNA (miRNA) and the mitogen-activated protein kinase (MAPK) pathway, an animal model of methionine selenium and the lipopolysaccharide (LPS) interaction was established. The morphology, inflammatory factor (tumor necrosis factor-α [TNF-α]), necroptosis-related genes (RIP1, RIP3, MLKL, and caspase 8), MAPK pathway-related genes (JNK, ERK, and p38, p-JNK, p-ERK, and p-p38), gga-miR-155, TRAF3 (predicted target of gga-miR-155), and oxidative stress-related indicators (SOD, MDA, CAT, GSH, and GSH-Px) were analyzed from the perspective of the miR-155/TRAF3/MAPK axis to elucidate the mechanism of methionine selenium on the LPS-induced necroptosis mechanism in the chicken liver. The current results suggested that methionine selenium antagonizes oxidative stress, inflammation, and the MAPK pathway, thereby antagonizing the occurrence of necroptosis through multiple mechanisms. At the same time, methionine selenium affects miR-155/TRAF3/MAPK signaling, reduces miR-155 expression, and upregulates TRAF3 expression to inhibit necroptosis. This information provided new ideas and a theoretical basis for the practical application of methionine selenium, and it also enriched the study of miRNAs in birds and provided a reference for comparative medicine.
Collapse
Affiliation(s)
- Zhao Zhirong
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Zhang Qiaojian
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Xu Chunjing
- National Selenium-Rich Product Quality Supervision and Inspection Center, Enshi, People's Republic of China
| | - Wang Shengchen
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Li Jiahe
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Liu Zhaoyi
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| | - Li Shu
- Department of Basic Veterinary Medicine, College of Veterinary Medicine, Northeast Agricultural University, Harbin, People's Republic of China
| |
Collapse
|
18
|
Liu J, Wang S, Zhang Q, Li X, Xu S. Selenomethionine alleviates LPS-induced chicken myocardial inflammation by regulating the miR-128-3p-p38 MAPK axis and oxidative stress. Metallomics 2020; 12:54-64. [PMID: 31720660 DOI: 10.1039/c9mt00216b] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
Selenium is closely related to the occurrence of heart disease, and an appropriate amount of selenium can alleviate inflammatory changes caused by various factors. Lipopolysaccharide (LPS), as a specific component of the cell wall of Gram-negative bacteria, is often used to construct various inflammatory models. In order to explore the effect of selenium on LPS-induced myocardial inflammation in chickens, we chose 4-month-old laying hens to be fed with a selenium-rich diet containing 0.5 g kg-1 Se, and injected LPS into the abdominal cavity at the age of 8 months to establish an inflammation model. We observed the myocardial tissue lesions by light microscopy, and detected miR-128-3p, p38MAPK, and NF-κB pathway-associated inflammatory factors and Th1/Th2 related factors by qRT-PCR and Western blot. The results showed that LPS stimulation inhibited miR-128-3p, which increased the expression of p38MAPK and NF-κB, while the expression of TNF-α, IL-1, PTGE, COX-2 and iNOS increased. Additionally, the expression of IL-4 and IL-6 increased and IFN-γ decreased, suggesting an imbalance of Th1/Th2. We also found that LPS treatment not only increased the content of H2O2 and MDA in the myocardium, but also increased the expression of HSP60, HSP70 and HSP90, while the activity of SOD, GPX and CAT and the content of GSH decreased. Interestingly, the addition of selenium can alleviate the changes in the above indicators. Finally, we concluded that selenium inhibits the occurrence of oxidative stress and ultimately alleviates myocardial inflammation induced by LPS through the miR-128-3p-p38MAPK-NF-κB pathway.
Collapse
Affiliation(s)
- Jing Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin 150030, P. R. China.
| | | | | | | | | |
Collapse
|
19
|
Li W, Sun K, Hu F, Chen L, Zhang X, Wang F, Yan B. Protective effects of natural compounds against oxidative stress in ischemic diseases and cancers via activating the Nrf2 signaling pathway: A mini review. J Biochem Mol Toxicol 2020; 35:e22658. [PMID: 33118292 DOI: 10.1002/jbt.22658] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 08/28/2020] [Accepted: 10/09/2020] [Indexed: 12/11/2022]
Abstract
Oxidative stress, an imbalance between reactive oxygen species and antioxidants, has been seen in the pathological states of many disorders such as ischemic diseases and cancers. Many natural compounds (NCs) have long been recognized to ameliorate oxidative stress due to their inherent antioxidant activities. The modulation of oxidative stress by NCs via activating the Nrf2 signaling pathway is summarized in the review. Three NCs, ursolic acid, betulinic acid, and curcumin, and the mechanisms of their cytoprotective effects are investigated in myocardial ischemia, cerebral ischemia, skin cancer, and prostate cancer. To promote the therapeutic performance of NCs with poor water solubility, the formulation approach, such as the nano drug delivery system, is elaborated as well in this review.
Collapse
Affiliation(s)
- Wenji Li
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| | - Kai Sun
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Fang Hu
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Longfei Chen
- China National Intellectual Property Administration Patent Re-examination and Invalidation Department Pharmaceutical Division, Beijing, China
| | - Xing Zhang
- Departments of Urology, Yangzhou Traditional Chinese Medicine Hospital Affiliated to Nanjing University of Chinese Medicine, Yangzhou, Jiangsu, China
| | - Fuxing Wang
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China
| | - Bingchun Yan
- Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Yangzhou University, Yangzhou, Jiangsu, China.,Jiangsu Key Laboratory of Zoonosis, Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, Jiangsu, China
| |
Collapse
|
20
|
Li G, Yang L, Feng L, Yang J, Li Y, An J, Li D, Xu Y, Gao Y, Li J, Liu J, Yang L, Qi Z. Syringaresinol Protects against Type 1 Diabetic Cardiomyopathy by Alleviating Inflammation Responses, Cardiac Fibrosis, and Oxidative Stress. Mol Nutr Food Res 2020; 64:e2000231. [PMID: 32729956 DOI: 10.1002/mnfr.202000231] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 07/03/2020] [Indexed: 12/18/2022]
Abstract
SCOPE Syringaresinol (SYR) is a phenolic compound, which could be found in various cereals and medicinal plants. It exerts both anti-inflammatory and antioxidant pharmacological properties. However, little is known about the effect of SYR on modulating diabetic cardiomyopathy. The present study aimed to investigate the pharmacodynamic effect of SYR on diabetic cardiomyopathy and the underlying molecular mechanism. METHODS AND RESULTS In STZ-induced type 1 diabetic mice, orally administration with SYR in every other day for 8 weeks significantly improves cardiac dysfunction and preventes cardiac hypertrophy and fibrosis. The macrophage infiltration and oxidative stress biomarkers are also suppressed by SYR without affecting hyperglycemia and body weight. In neonatal cardiomyocytes, high glucose-induced cell apoptosis and fibrosis are potently decreased by SYR, and the inflammatory response and oxidant stress are also alleviated by SYR incubation. Mechanistically, SYR may exert protective effects by restoring suppression of antioxidant kelch-like ECH-associated protein 1 (Keap1)/nuclear factor-E2-related factor 2 (Nrf2) system and abnormal activation of transforming growth factor-β (TGF-β)/mothers against decapentaplegic homolog (Smad) signaling pathway in vitro and in vivo. CONCLUSION The results indicated that SYR could be a potential therapeutic agent for the treatment of diabetic cardiomyopathy by inhibiting inflammation, fibrosis, and oxidative stress. The signaling pathway of Keap1/Nrf2 and TGF-β/Smad could be used as therapeutic targets for diabetic complications.
Collapse
Affiliation(s)
- Guangru Li
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Lei Yang
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Lifeng Feng
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jiu Yang
- Clinical laboratory, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, 300197, China
| | - Yafei Li
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jiale An
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Dihua Li
- Tianjin Key Laboratory of Acute Abdomen Disease Associated Organ Injury and ITCWM Repair, Institute of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin, 300100, China
| | - Yang Xu
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Yang Gao
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jing Li
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Jie Liu
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Liang Yang
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zhi Qi
- Department of Pharmacology, School of Medicine, Nankai University, Tianjin, 300071, China
| |
Collapse
|