1
|
Luo Y, Chang T, Huang S, Xiang J, Tang S, Shen H. Protective Effects and Mechanisms of Esculetin against H 2O 2-Induced Oxidative Stress, Apoptosis, and Pyroptosis in Human Hepatoma HepG2 Cells. Molecules 2024; 29:1415. [PMID: 38611695 PMCID: PMC11013120 DOI: 10.3390/molecules29071415] [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: 02/22/2024] [Revised: 03/15/2024] [Accepted: 03/20/2024] [Indexed: 04/14/2024] Open
Abstract
Oxidative stress plays a crucial role in the pathogenesis of many diseases. Esculetin is a natural coumarin compound with good antioxidant and anti-inflammatory properties. However, whether esculetin can protect HepG2 cells through inhibiting H2O2-induced apoptosis and pyroptosis is still ambiguous. Therefore, this study aimed to investigate the protective effects and mechanisms of esculetin against oxidative stress-induced cell damage in HepG2 cells. The results of this study demonstrate that pretreatment with esculetin could significantly improve the decrease in cell viability induced by H2O2 and reduce intracellular ROS levels. Esculetin not only apparently reduced the apoptotic rates and prevented MMP loss, but also markedly decreased cleaved-Caspase-3, cleaved-PARP, pro-apoptotic protein (Bax), and MMP-related protein (Cyt-c) expression, and increased anti-apoptotic protein (Bcl-2) expression in H2O2-induced HepG2 cells. Meanwhile, esculetin also remarkably reduced the level of LDH and decreased the expression of the pyroptosis-related proteins NLRP3, cleaved-Caspase-1, Il-1β, and GSDMD-N. Furthermore, esculetin pretreatment evidently downregulated the protein expression of p-JNK, p-c-Fos, and p-c-Jun. Additionally, anisomycin, a specific activator of JNK, blocked the protection of esculetin against H2O2-induced HepG2 cells apoptosis and pyroptosis. In conclusion, esculetin can protect HepG2 cells against H2O2-induced oxidative stress, apoptosis, and pyroptosis via inhibiting the JNK signaling pathway. These findings indicate that esculetin has the potential to be used as an antioxidant that improves oxidative stress-related diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Haiyan Shen
- The Institute of Biochemistry and Molecular Biology, Hunan Provincial Key Laboratory for Special Pathogens Prevention and Control, Hengyang Medical College, University of South China, Hengyang 421001, China; (Y.L.); (T.C.); (S.H.); (J.X.); (S.T.)
| |
Collapse
|
2
|
Rezoan Hossain M, Zahra Shova FT, Akter M, Shuvo S, Ahmed N, Akter A, Haque M, Salma U, Roman Mogal M, Saha HR, Sarkar BC, Sohel M. Esculetin unveiled: Decoding its anti-tumor potential through molecular mechanisms-A comprehensive review. Cancer Rep (Hoboken) 2024; 7:e1948. [PMID: 38062981 PMCID: PMC10809201 DOI: 10.1002/cnr2.1948] [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: 08/23/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The growing complexity of cancer has made it a significant concern in the medical community. Although cancer research has advanced, it is still challenging to create new effective medications due to the limitations and side effects of existing treatment strategies. These are enforcing the development of some alternative drugs from natural compounds with fewer drawbacks and side effects. AIM Therefore, this review aims to provide up-to-date, crucial, and all-encompassing data on esculetin's anticancer activity, including all relevant molecular and cellular processes based on in vivo and in vitro investigations. RESULTS According to the literature review, esculetin is available in nature and is effective against 16 different types of cancer. The general mechanism shown by esculetin is modulating signaling cascades and its related pathways, like cell proliferation, cell growth, autophagy, apoptosis, necrosis, inflammation, angiogenesis, metastasis, invasion, and DNA damage. Nanoformulation of esculetin improves this natural product's efficacy by improving water solubility. Esculetin's synergistic effects with both natural substances and conventional treatments have been shown, and this method aids in reversing resistance mechanisms by modulating resistance-related proteins. In addition, it has fewer side effects on humans than other phytochemicals and standard drugs with some good pharmacokinetic features. CONCLUSION Therefore, until standard chemotherapeutics are available in pharmaceutical markets, esculetin should be used as a therapeutic drug against various cancer types.
Collapse
Affiliation(s)
| | - Fatema Tuj Zahra Shova
- Biotechnology and Genetic EngineeringMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Munni Akter
- Department of MedicalDinajpure Nursing College (Affiliated Rajshahi University)DinajpurBangladesh
| | - Shahporan Shuvo
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Nasim Ahmed
- Department of PharmacyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Afroza Akter
- Departmnet of MicrobiologyNoakhali Science and Technology UniversityNoakhaliBangladesh
| | - Munira Haque
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Umme Salma
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Md Roman Mogal
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Hasi Rani Saha
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | | | - Md Sohel
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| |
Collapse
|
3
|
Murata Y, Suzuki K, Shigeta Y, Iso T, Hirose N, Umano T, Horibata K, Sugiyama KI, Hirose A, Masumura K, Matsumoto M. In vivo mutagenicity assessment of orally treated tert-butyl hydroperoxide in the liver and glandular stomach of MutaMouse. Genes Environ 2023; 45:29. [PMID: 37990244 PMCID: PMC10662197 DOI: 10.1186/s41021-023-00285-2] [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: 08/16/2023] [Accepted: 10/19/2023] [Indexed: 11/23/2023] Open
Abstract
BACKGROUND tert-Butyl hydroperoxide (TBHP; CAS 75-91-2), a hydroperoxide, is mainly used as a polymerization initiator to produce polyethylene, polyvinyl chloride, and unsaturated polyester. It is a high-production chemical, widely used in industrial countries, including Japan. TBHP is also used as an additive for the manufacturing of food utensils, containers, and packaging (UCP). Therefore, there could be consumer exposure through oral intake of TBHP eluted from UCPs. TBHP was investigated in various in vitro and in vivo genotoxicity assays. In Ames tests, some positive results were reported with and/or without metabolic activation. As for the mouse lymphoma assay, the positive result was reported, regardless of the presence or absence of metabolic activation enzymes. The results of some chromosomal aberrations test and comet assay in vitro also demonstrated the genotoxic positive results. On the other hand, in in vivo tests, there are negative results in the bone marrow micronucleus test of TBHP-administered mice by single intravenous injection and the bone marrow chromosomal aberration test using rats exposed to TBHP for 5 days by inhalation. Also, about dominant lethal tests, the genotoxic positive results appeared. In contrast, there is little information about in vivo mutagenicity and no information about carcinogenicity by oral exposure. RESULTS We conducted in vivo gene mutation assay using MutaMice according to the OECD Guidelines for the Testing of Chemicals No. 488 to investigate in vivo mutagenicity of TBHP through oral exposure. After repeated dosing for 28 days, there were no significant differences in the mutant frequencies (MFs) of the liver and glandular stomach up to 300 mg/kg/day (close to the maximum tolerable dose (MTD)). The positive and negative controls produced the expected responses. CONCLUSIONS These findings show that orally administrated TBHP is not mutagenic in the mouse liver and glandular stomach under these experimental conditions.
Collapse
Affiliation(s)
- Yasumasa Murata
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan.
| | - Kenichiro Suzuki
- Genotoxicology Laboratory, BioSafety Research Center Inc., Shizuoka, Japan
| | - Yoshiyuki Shigeta
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan
- Division of Chemical Information, National Institute of Occupational Safety and Health, Kanagawa, Japan
| | - Takako Iso
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan
| | - Nozomu Hirose
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan
| | - Takaaki Umano
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan
| | - Katsuyoshi Horibata
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Kanagawa, Japan
| | - Kei-Ichi Sugiyama
- Division of Genetics and Mutagenesis, National Institute of Health Sciences, Kanagawa, Japan
| | - Akihiko Hirose
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan
- Chemicals Evaluation and Research Institute, Tokyo, Japan
| | - Kenichi Masumura
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan
| | - Mariko Matsumoto
- Division of Risk Assessment, National Institute of Health Sciences, Kanagawa, Japan.
| |
Collapse
|
4
|
Jung WK, Park SB, Yu HY, Kim YH, Kim J. Effect of Esculetin on Tert-Butyl Hydroperoxide-Induced Oxidative Injury in Retinal Pigment Epithelial Cells In Vitro. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248970. [PMID: 36558102 PMCID: PMC9781126 DOI: 10.3390/molecules27248970] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Esculetin is a coumarin-derived compound with antioxidant and anti-inflammatory properties. The current study aims to evaluate the therapeutic implications of esculetin on retinal dysfunction and uncover the underlying mechanisms. Tert-butyl hydroperoxide (t-BHP) at a concentration of 300 μM was used to induce oxidative stress in human retinal pigment epithelial cell line (ARPE-19) cells. Esculetin at concentrations below 250 μM did not cause cytotoxicity to ARPE-19 cells. Cell viability analysis confirmed that t-BHP induced oxidative injury of ARPE-19 cells. However, ARPE-19 cells were protected from t-BHP-induced oxidative injury by esculetin in a concentration-dependent manner. As a result of the TUNEL assay to confirm apoptosis, esculetin treatment reduced the number of TUNEL-positive cells. Esculetin down-regulated the expression levels of Bax, Caspase-3, and PARP and up-regulated the expression level of Bcl2. Collectively, this study demonstrates that esculetin exerts potent antioxidant properties in ARPE-19 cells, inhibiting t-BHP-induced apoptosis under the regulation of apoptotic factors.
Collapse
Affiliation(s)
| | | | | | | | - Junghyun Kim
- Correspondence: ; Tel.: +82-63-270-4032; Fax: +82-63-270-4025
| |
Collapse
|
5
|
Jung WK, Park SB, Yu HY, Kim YH, Kim J. Antioxidant Efficacy of Esculetin against Tert-Butyl Hydroperoxide-Induced Oxidative Stress in HEK293 Cells. Curr Issues Mol Biol 2022; 44:5986-5994. [PMID: 36547068 PMCID: PMC9777115 DOI: 10.3390/cimb44120407] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Esculetin is an antioxidant and anti-inflammatory compound derived from coumarin. Oxidative stress can cause overproduction of reactive oxygen species (ROS), which can lead to the development of chronic kidney failure. In this study, human embryonic kidney 293 (HEK293) cells were treated with tert-butyl hydroperoxide (t-BHP) to determine the antioxidant effects of esculetin. HEK293 cells were treated with t-BHP to validate changes in cell viability, ROS production, and apoptosis, and then treated with esculetin to evaluate the changes. Changes in mRNA and protein levels were analyzed using a proteome kit, PCR, and Western blotting. Esculetin improved HEK293 cell viability and reduced apoptosis caused by t-BHP-induced oxidative stress. At the mRNA and protein levels, esculetin decreased pro-apoptotic factor expression as well as increased anti-apoptotic factor expression. The antioxidant efficacy of esculetin was validated when it inhibited the apoptosis caused by t-BHP-induced oxidative stress in HEK293 cells.
Collapse
Affiliation(s)
| | | | | | | | - Junghyun Kim
- Correspondence: ; Tel.: +82-63-270-4032; Fax: +82-63-270-4025
| |
Collapse
|
6
|
Park SB, Jung W, Kim H, Yu HY, Kim Y, Kim J. Esculetin has therapeutic potential via the proapoptotic signaling pathway in A253 human submandibular salivary gland tumor cells. Exp Ther Med 2022; 24:533. [PMID: 35837055 PMCID: PMC9257944 DOI: 10.3892/etm.2022.11460] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2021] [Accepted: 03/16/2022] [Indexed: 11/24/2022] Open
Abstract
Esculetin is a natural lactone that is commonly derived from coumarins. According to previous experiments using human cancer cells, esculetin has potent antitumor activity; it also inhibits proliferation and induces the apoptosis of cancer cells. In the present study, the anti-proliferative effect of esculetin on the submandibular salivary gland tumor cell line, A253, was evaluated via in vitro and in vivo analyses. Furthermore, the anti-cancer effects of esculetin in A253 cells and a xenograft model of salivary gland tumors were determined using 3-(4,5-dimethylthiazol)-2,5-diphenyltetrazolium bromide and TUNEL assay, apoptosis protein array, quantitative polymerase chain reaction and western blot analysis. Esculetin (50-150 µM) was demonstrated to have an anti-proliferative effect in the A253 cell line in vitro; this observed effect was dependent on the dose and duration of treatment. Esculetin also increased the levels of Bax, cleaved caspase-3, cleaved-9 and cleaved poly (ADP-ribose) polymerase apoptosis-related proteins, and decreased the expression levels of the Bcl-2 anti-apoptotic protein. With respect to apoptosis regulation, esculetin significantly decreased the proliferation of tumor cells in a xenograft model (100 mg/kg/day) for 18 days. Overall, esculetin could be a potential oral anticancer drug against salivary gland cancer.
Collapse
Affiliation(s)
- Su-Bin Park
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, Republic of Korea
| | - Woo Jung
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, Republic of Korea
| | - Hyung Kim
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, Republic of Korea
| | - Hwa-Young Yu
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, Republic of Korea
| | - Yong Kim
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, Republic of Korea
| | - Junghyun Kim
- Department of Oral Pathology, School of Dentistry, Jeonbuk National University, Jeonju, Jeollabuk-do 54896, Republic of Korea
| |
Collapse
|
7
|
Luo G, Meng Y, Zhang L, Li X, Tao Z, Zhang Q. Selective recognition of aluminum ions using an esculetin@Q[8] host–guest supramolecular fluorescent probe. NEW J CHEM 2022. [DOI: 10.1039/d1nj04782e] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Esculetin (ESC) and non-toxic Q[8] form a supramolecular compound combination, which can effectively monitor Al3+ in the water environment.
Collapse
Affiliation(s)
- Guangyan Luo
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Ye Meng
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Lin Zhang
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Xiaoyue Li
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Zhu Tao
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| | - Qianjun Zhang
- Key Laboratory of Macrocyclic and Supramolecular Chemistry of Guizhou Province, Guizhou University, Guiyang 550025, China
| |
Collapse
|
8
|
Zhang L, Xie Q, Li X. Esculetin: A review of its pharmacology and pharmacokinetics. Phytother Res 2021; 36:279-298. [PMID: 34808701 DOI: 10.1002/ptr.7311] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Revised: 09/12/2021] [Accepted: 09/30/2021] [Indexed: 12/30/2022]
Abstract
Esculetin is a natural dihydroxy coumarin; it is mainly extracted from twig skin and the trunk bark of the Chinese herbal medicine Fraxinus rhynchophylla Hance. Emerging evidence suggests that esculetin has a wide range of pharmacological activities. Based on its fundamental properties, including antioxidant, antiinflammatory, antiapoptotic, anticancer, antidiabetic, neuroprotective, and cardiovascular protective activities, as well as antibacterial activity, among others, esculetin is expected to be a therapeutic drug for specific disease indications, such as cancer, diabetes, atherosclerosis, Alzheimer's disease (AD), Parkinson's disease (PD), nonalcoholic fatty liver disease (NAFLD), and other diseases. The oral bioavailability of esculetin was shown by studies to be low. The extensive glucuronidation was described to be the main metabolic pathway of esculetin and C-7 phenolic hydroxyl to be its major metabolic site. With the development of scientific research technology, the pharmacological effects of esculetin are identified and its potential for the treatment of diseases is demonstrated. The underlining mechanisms of action and biological activities as well as the pharmacokinetic data of the analyzed compound reported so far are highlighted in this review with the aim of becoming a proven, and applicable insight and reference for further studies on the utilization of esculetin.
Collapse
Affiliation(s)
- Linlin Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qingxuan Xie
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaofang Li
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
9
|
Mohamadi-Zarch SM, Baluchnejadmojarad T, Nourabadi D, Ramazi S, Nazari-Serenjeh M, Roghani M. Esculetin Alleviates Acute Liver Failure following Lipopolysaccharide/D-Galactosamine in Male C57BL/6 Mice. IRANIAN JOURNAL OF MEDICAL SCIENCES 2021; 46:373-382. [PMID: 34539012 PMCID: PMC8438341 DOI: 10.30476/ijms.2020.84909.1474] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Revised: 08/17/2020] [Accepted: 08/31/2020] [Indexed: 12/03/2022]
Abstract
Background: Acute liver failure (ALF) is a fatal clinical situation that rapidly leads to the loss of normal liver function. Esculetin is a natural herbal compound used
for the management of various diseases such as cardiovascular and renal disorders. In this study, we evaluated the protective effects of esculetin in a mouse model of ALF. Methods: This article is a report on an experimental study that was conducted at Iran University of Medical Sciences in 2019. Forty-eight male C57BL/6 mice were randomly
divided into control, LPS/D-Gal, and LPS/D-Gal+Esculetin (40 mg/kg) groups (n=16 per group). ALF was induced with an intraperitoneal injection of lipopolysaccharide
(LPS)/D-galactosamine (D-Gal).The LPS/D-Gal group received a mixture of LPS (50 μg/kg) and D-Gal (400 mg/kg). The LPS/D-Gal+Esculetin group received esculetin by
gavage 24 hours and one hour before receiving LPS/D-Gal. Six hours after LPS/D-Gal injection, the mice were sacrificed. Liver injury markers, including alanine
aminotransferase (ALT), aspartate transaminase (AST), and alkaline phosphatase (ALP), were measured in the serum. Oxidative stress indices and inflammatory
markers such as interleukin-1 beta (IL-1β), IL-6, and tumor necrosis factor-alpha (TNF-α) were measured in hepatic tissue. The histopathology of liver tissue
was also assessed. The data were analyzed using one-way ANOVA, followed by the post hoc Tukey test. Results: Esculetin lowered oxidative stress and myeloperoxidase activity (P<0.001); reduced the serum levels of ALT (P=0.037), AST (P=0.032), and ALP (P=0.004);
and decreased the hepatic levels of IL-1β (P=0.002), IL-6 (P=0.004), toll-like receptor 4 (P<0.001), TNF-α (P=0.003), and nuclear factor-kappa B (P<0.001)
as compared with LPS/D-Gal. Additionally, esculetin ameliorated hepatic tissue injury following LPS/D-Gal challenge. Conclusion: Esculetin can reduce liver injury through the mitigation of oxidative burden, inflammation, and neutrophil infiltration and also exerts hepatoprotective effects against ALF.
Collapse
Affiliation(s)
| | | | - Davood Nourabadi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Ramazi
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Nazari-Serenjeh
- Department of Physiology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Roghani
- Neurophysiology Research Center, Shahed University, Tehran, Iran
| |
Collapse
|
10
|
Boudreau A, Richard AJ, Harvey I, Stephens JM. Artemisia scoparia and Metabolic Health: Untapped Potential of an Ancient Remedy for Modern Use. Front Endocrinol (Lausanne) 2021; 12:727061. [PMID: 35211087 PMCID: PMC8861327 DOI: 10.3389/fendo.2021.727061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 12/16/2021] [Indexed: 11/19/2022] Open
Abstract
Botanicals have a long history of medicinal use for a multitude of ailments, and many modern pharmaceuticals were originally isolated from plants or derived from phytochemicals. Among these, artemisinin, first isolated from Artemisia annua, is the foundation for standard anti-malarial therapies. Plants of the genus Artemisia are among the most common herbal remedies across Asia and Central Europe. The species Artemisia scoparia (SCOPA) is widely used in traditional folk medicine for various liver diseases and inflammatory conditions, as well as for infections, fever, pain, cancer, and diabetes. Modern in vivo and in vitro studies have now investigated SCOPA's effects on these pathologies and its ability to mitigate hepatotoxicity, oxidative stress, obesity, diabetes, and other disease states. This review focuses on the effects of SCOPA that are particularly relevant to metabolic health. Indeed, in recent years, an ethanolic extract of SCOPA has been shown to enhance differentiation of cultured adipocytes and to share some properties of thiazolidinediones (TZDs), a class of insulin-sensitizing agonists of the adipogenic transcription factor PPARγ. In a mouse model of diet-induced obesity, SCOPA diet supplementation lowered fasting insulin and glucose levels, while inducing metabolically favorable changes in adipose tissue and liver. These observations are consistent with many lines of evidence from various tissues and cell types known to contribute to metabolic homeostasis, including immune cells, hepatocytes, and pancreatic beta-cells. Compounds belonging to several classes of phytochemicals have been implicated in these effects, and we provide an overview of these bioactives. The ongoing global epidemics of obesity and metabolic disease clearly require novel therapeutic approaches. While the mechanisms involved in SCOPA's effects on metabolic, anti-inflammatory, and oxidative stress pathways are not fully characterized, current data support further investigation of this plant and its bioactives as potential therapeutic agents in obesity-related metabolic dysfunction and many other conditions.
Collapse
Affiliation(s)
- Anik Boudreau
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Allison J. Richard
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Innocence Harvey
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
| | - Jacqueline M. Stephens
- Adipocyte Biology Laboratory, Pennington Biomedical Research Center, Baton Rouge, LA, United States
- Department of Biological Sciences, Louisiana State University, Baton Rouge, LA, United States
- *Correspondence: Jacqueline M. Stephens,
| |
Collapse
|
11
|
Shaikh S, Pavale G, Ramana MMV. Synthesis and biological evaluation of novel N-substituted (3-(1-aminoethylidene)-2-oxochroman-4-yl)phosphonic acid diethyl ester derivatives as anti-Alzheimer agent. CHEMICAL PAPERS 2020. [DOI: 10.1007/s11696-020-01099-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
12
|
Serralha RS, Rodrigues IF, Bertolini A, Lima DY, Nascimento M, Mouro MG, Punaro GR, Visoná I, Rodrigues AM, Higa EMS. Esculin reduces P2X7 and reverses mitochondrial dysfunction in the renal cortex of diabetic rats. Life Sci 2020; 254:117787. [PMID: 32417372 DOI: 10.1016/j.lfs.2020.117787] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 05/09/2020] [Accepted: 05/11/2020] [Indexed: 02/07/2023]
Abstract
AIMS To evaluate the effects of esculin treatment on P2X7 receptor and mitochondrial dysfunction in the renal cortex of diabetic rats. MAIN METHODS Male Wistar rats, 7 weeks old, were unilaterally nephrectomized. Part of these animals were induced to diabetes using streptozotocin (60 mg/kg). Diabetes was confirmed 48 h after induction, with blood glucose levels ≥200 mg/dL. Part of control and diabetic animals were selected to receive daily doses of esculin (50 mg/kg), during 8 weeks. The animals were placed in metabolic cages at the eighth week of protocol for 24 h urine collection and a small aliquot of blood was collected for biochemical analysis. After this procedure, the animals were euthanized and the remaining kidney was stored for histopathological analysis, Western blotting and mitochondrial high-resolution respirometry. KEY FINDINGS Although esculin did not change metabolic parameters, renal biochemical function, neither TBARS in DM rats, esculin reduced P2X7 levels in these animals and restored mitochondrial function via glycolysis substrates and β-oxidation. Besides, at the histological analysis, we observed that esculin reduced inflammatory infiltrates and collagen IV deposits as compared to diabetic group. SIGNIFICANCE Esculin attenuated the development of renal injuries caused by hyperglycemia, proinflammatory and oxidative mechanisms mediated by P2X7 receptor, as seen by histological findings and improved mitochondrial function in diabetic animals. This suggests that esculin could be used as an adjuvant therapy to prevent the diabetic nephropathy.
Collapse
Affiliation(s)
- R S Serralha
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil.
| | - I F Rodrigues
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - A Bertolini
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - D Y Lima
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - M Nascimento
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology, Universidade Federal de Sao Paulo, Brazil
| | - M G Mouro
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - G R Punaro
- Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - I Visoná
- Pathology Department, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - A M Rodrigues
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| | - E M S Higa
- Translational Medicine, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Laboratory of Nitric Oxide and Oxidative Stress, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Nephrology Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil; Emergency Division, Universidade Federal de Sao Paulo (UNIFESP-EPM), Brazil
| |
Collapse
|
13
|
Sadik NAH, El-Boghdady NA, Omar NN, Al-Hamid HA. Esculetin and idebenone ameliorate galactose-induced cataract in a rat model. J Food Biochem 2020; 44:e13230. [PMID: 32301145 DOI: 10.1111/jfbc.13230] [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] [Received: 10/30/2019] [Revised: 02/23/2020] [Accepted: 03/20/2020] [Indexed: 01/20/2023]
Abstract
Cataract is the principal cause of blindness. The enzyme, aldose reductase (AR) is a key player in polyol pathway. Buildup of polyols and oxidative stress are the main causes of cataractogenesis. This study investigated the cytoprotective properties of esculetin and idebenone in galactose-induced cataract. Rats were partitioned into four groups each of ten rats. Control group was fed with normal diet; group 2 rats were fed with galactose diet (50%); groups 3, 4 rats were fed with galactose diet concurrently with either esculetin (50 mg/kg BW) or idebenone (100 mg/kg BW), for 20 days. The study revealed that esculetin and idebenone significantly reduced the elevated levels of Bax/Bcl-2 ratio, malondialdehyde, and DNA fragmentation and increased total antioxidant capacity level in lenses compared to the cataract-induced group. Only esculetin decreased AR, galactitol, and advanced glycated end products levels in lenses. Histopathological examinations supported the biochemical findings. Esculetin and idebenone may have chemopreventive effects for sugar cataract. PRACTICAL APPLICATIONS: Cataract is an age-related disease that might cause blindness in older adult people. Presently, no absolute pharmacological treatment is accessible for cataract. The use of natural products or their derivatives attract particular attention in modern medicines as they are believed to be safer with few or no side effects. Esculetin is a polyphenolic compound found in many medicinal plants. Idebenone is a synthetic analogue of coenzyme Q10. The current study is an approach to explore the anticataract effects of esculetin and idebenone in galactose-induced cataract in rats. Our study proved that both agents have anticataractogenic potentials due to their antioxidant and antiapoptotic properties.
Collapse
Affiliation(s)
| | | | - Nesreen Nabil Omar
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| | - Hager Abd Al-Hamid
- Biochemistry Department, Faculty of Pharmacy, Modern University for Technology and Information, Cairo, Egypt
| |
Collapse
|
14
|
Ruthenium derivatives attenuate LPS-induced inflammatory responses and liver injury via suppressing NF-κB signaling and free radical production. Bioorg Chem 2020; 96:103639. [DOI: 10.1016/j.bioorg.2020.103639] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Revised: 01/10/2020] [Accepted: 01/28/2020] [Indexed: 12/12/2022]
|
15
|
Karatug Kacar A, Bahadori F, Kepekci Tekkeli SE, Topcu G, Bolkent S. Investigation of cell death mechanism and activity of esculetin-loaded PLGA nanoparticles on insulinoma cells in vitro. ACTA ACUST UNITED AC 2020; 72:592-606. [PMID: 31978266 DOI: 10.1111/jphp.13228] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Revised: 12/13/2019] [Accepted: 12/17/2019] [Indexed: 01/03/2023]
Abstract
AIM The purpose of this study was to prepare targeted cancer therapy formulation against insulinoma INS-1 cells and to study its effect on cell death with related mechanisms in vitro. METHODS Polylactide-co-glycolide (PLGA) nano-micelles were used for preparation of esculetin nano-formulation (nano-esculetin). The cells were treated with nano-esculetin and free esculetin. Apoptotic and necrotic cell death percentages, cell proliferation, ATP and GTP reductions and insulin levels were investigated on insulinoma INS-1 cells for both free and nano-esculetin formulations. RESULTS About 50 mg of PLGA was able to carry 20 mg esculetin in 20 ml of formulation. The obtained optimized formulation was 150 nm, with 92% encapsulation efficiency and a slow-release behaviour was observed during release studies. Nano-esculetin bearing 25, 50 and 100 μg esculetin and free esculetin in equivalent doses successfully decreased cell viability. The prevailing cell death mechanism was necrosis. Along with cell proliferation, intracellular insulin and the ratio of ATP and GTP were decreased even with 12.5, 25 and 50 μg esculetin bearing nano-formulation and its equivalent free esculetin. CONCLUSIONS The results revealed that esculetin is able to show its anti-tumor afficacy after loading to PLGA nano-micelles and nano-encapsulation intensifies its cytotoxic activity in vitro. Current study shows that esculetin and its nano formulations are promising agents in treatment of insulinoma.
Collapse
Affiliation(s)
- Ayse Karatug Kacar
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| | - Fatemeh Bahadori
- Department of Pharmaceutical Biotechnology, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Turkey
| | | | - Gulacti Topcu
- Department of Pharmacognosy, Faculty of Pharmacy, Bezmialem Vakif University, Istanbul, Turkey
| | - Sehnaz Bolkent
- Department of Biology, Faculty of Science, Istanbul University, Istanbul, Turkey
| |
Collapse
|
16
|
Yang SY, Pyo MC, Nam MH, Lee KW. ERK/Nrf2 pathway activation by caffeic acid in HepG2 cells alleviates its hepatocellular damage caused by t-butylhydroperoxide-induced oxidative stress. Altern Ther Health Med 2019; 19:139. [PMID: 31221142 PMCID: PMC6585055 DOI: 10.1186/s12906-019-2551-3] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2018] [Accepted: 06/07/2019] [Indexed: 02/06/2023]
Abstract
Background Several studies have found that caffeic acid (CA), a well-known phytochemical, displays important antioxidant and anti-cancer activities. However, no evidence exists on the protective effect and its mechanisms that CA treatment alone has against oxidative stress induced by tert-butyl hydroperoxide (t-BHP) in HepG2 cells. Methods Hepatoprotective activities such as cell viability, mRNA expression, and report gene assay were measured using HepG2 cell. Three types of genes and proteins related with detoxification in liver were used for measuring the hepatoprotective effects. Statistical analysis was performed using one-way ANOVA test and differences among groups were evaluated by Tukey’s studentized range tests. Results The present study indicate that treatment with CA up-regulates heme oxygenase-1 (HO-1) and glutamate-cysteine ligase (GCL) mRNA and protein expressions in a CA-dose-dependent manner. In addition, translocation of nuclear factor-E2 p45-related factor (Nrf2) from the cytoplasm to the nucleus and phosphorylation of extracellular signal-regulated kinase, ERK and c-Jun N-terminal kinase, JNK which have been shown to be involved in mitogen-activated protein kinases, MAPKs are significantly enhanced by CA treatment. Furthermore, in cell nuclei, CA enhances the 5′-flanking regulatory region of human antioxidant response element (ARE) and activates the ARE binding site. Conclusion Therefore, CA proved to be a stimulant of the expression of detoxification enzymes such as HO-1, GCLC, and GCLM through the ERK/Nrf2 pathway, and it may be an effective chemoprotective agent for protecting liver damage against oxidative damage. Graphical abstract ![]()
Electronic supplementary material The online version of this article (10.1186/s12906-019-2551-3) contains supplementary material, which is available to authorized users.
Collapse
|
17
|
de Jesus Souza M, de Moraes JA, Da Silva VN, Helal-Neto E, Uberti AF, Scopel-Guerra A, Olivera-Severo D, Carlini CR, Barja-Fidalgo C. Helicobacter pylori urease induces pro-inflammatory effects and differentiation of human endothelial cells: Cellular and molecular mechanism. Helicobacter 2019; 24:e12573. [PMID: 30907046 DOI: 10.1111/hel.12573] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2018] [Revised: 01/21/2019] [Accepted: 01/30/2019] [Indexed: 12/26/2022]
Abstract
BACKGROUND Helicobacter pylori urease (HPU) is a key virulence factor that enables bacteria to colonize and survive in the stomach. We early demonstrated that HPU, independent of its catalytic activity, induced inflammatory and angiogenic responses in vivo and directly activated human neutrophils to produce reactive oxygen species (ROS). We have investigated the effects of HPU on endothelial cells, focusing on the signaling mechanism involved. METHODS Monolayers of human microvascular endothelial cells (HMEC-1) were stimulated with HPU (up to 10 nmol/L): Paracellular permeability was accessed through dextran-FITC passage. NO and ROS production was evaluated using intracellular probes. Proteins or mRNA expressions were detected by Western blotting and fluorescence microscopy or qPCR assays, respectively. RESULTS Treatment with HPU enhanced paracellular permeability of HMEC-1, preceded by VE-cadherin phosphorylation and its dissociation from cell-cell junctions. This caused profound alterations in actin cytoskeleton dynamics and focal adhesion kinase (FAK) phosphorylation. HPU triggered ROS and nitric oxide (NO) production by endothelial cells. Increased intracellular ROS resulted in nuclear factor kappa B (NF-κB) activation and upregulated expression of cyclooxygenase-2 (COX-2), hemeoxygenase-1 (HO-1), interleukin-1β (IL-1β), and intercellular adhesion molecule-1 (ICAM-1). Higher ICAM-1 and E-selectin expression was associated with increased neutrophil adhesion on HPU-stimulated HMEC monolayers. The effects of HPU on endothelial cells were dependent on ROS production and lipoxygenase pathway activation, being inhibited by esculetin. Additionally, HPU improved vascular endothelial growth factor receptor 2 (VEGFR-2) expression. CONCLUSION The data suggest that the pro-inflammatory properties of HPU drive endothelial cell to a ROS-dependent program of differentiation that contributes to the progression of H pylori infection.
Collapse
Affiliation(s)
- Mariele de Jesus Souza
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - João Alfredo de Moraes
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil.,Laboratory of Redox Biology, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Vany Nascimento Da Silva
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Edward Helal-Neto
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Augusto Frantz Uberti
- Laboratory of Neurotoxins, Brain Institute (BRAINS-InsCer), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil
| | - Adriele Scopel-Guerra
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil
| | - Deiber Olivera-Severo
- Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil
| | - Célia R Carlini
- Laboratory of Neurotoxins, Brain Institute (BRAINS-InsCer), Pontifícia Universidade Católica do Rio Grande do Sul, Porto Alegre, Brazil.,Center of Biotechnology, Universidade Federal Rio Grande do Sul, Porto Alegre, Brazil
| | - Christina Barja-Fidalgo
- Laboratory of Cellular and Molecular Pharmacology, Universidade do Estado do Rio de Janeiro, Rio de Janeiro, Brazil
| |
Collapse
|
18
|
Wang Y, Zhang B, Zhang J, Tian X, Sun D, Li Q, Wang R. Qualitative and quantitative analysis of Yifei Tongluo granules to identify main bioactive components using LC–DAD/MS and pharmacokinetic studies. J Pharm Biomed Anal 2019; 163:130-136. [DOI: 10.1016/j.jpba.2018.09.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2018] [Revised: 09/06/2018] [Accepted: 09/07/2018] [Indexed: 10/28/2022]
|
19
|
Zhu A, Zhang T, Wang Q. The phytochemistry, pharmacokinetics, pharmacology and toxicity of Euphorbia semen. JOURNAL OF ETHNOPHARMACOLOGY 2018; 227:41-55. [PMID: 30144497 DOI: 10.1016/j.jep.2018.08.024] [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: 05/04/2018] [Revised: 08/07/2018] [Accepted: 08/19/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Euphorbia semen, the dried and ripe seed of Euphorbia lathyris Linnaeus, is widely cultivated for traditional medicine use. This semen is used to expel water, help with phlegm retention, promote blood circulation, remove blood stasis, cure tinea and scabies, and treat amenorrhea, snakebites, terminal schistosomiasis, anuria and constipation. AIM OF THE REVIEW This review provides updated, comprehensive and categorized information on the local and traditional uses, phytochemistry, pharmacokinetics, pharmacological activities and toxicity of Euphorbia semen. Future research to deepen the recognition and utilization of Euphorbia semen is proposed. MATERIALS AND METHODS This article conducted a literature review on information about Euphorbia semen in multiple Internet databases, including PubMed, Web of Science, Wiley, Science Direct, Elsevier, ACS publications, SciFinder, Google Scholar and China National Knowledge Internet, until March of 2018. In this manuscript, a number of books, PhD and MSc dissertations, and Chinese Pharmacopeia were also used as references. RESULTS Approximately 240 chemical constituents have been isolated and identified from Euphorbia semen, namely, diterpenoids, coumarins, flavonoids, fatty acids, amino acids, and steroids. Pharmacokinetic study focused on investigating absorption, distribution, metabolism and excretion (ADME). The chemical constituents have extensive pharmacological effects, such as diuresis and anti-hyperuricaemia, anti-inflammation, antiviral, anticancer, antioxidant, antipigmentation, anti-platelet aggregation and anti-allergic activities, as well as hepatoprotection and neuroprotection. The toxicity of Euphorbia semen, including acute toxicity, target organ irritation and cocareinogenic effects, have been reported, and the detoxification methods are reviewed. CONCLUSION Euphorbia semen has extensive pharmacological activity and excellent clinical value, along with intense intestinal irritation. Although plenty of chemical constituents have been isolated and identified, the exact pharmacological and toxicological mechanisms still need to be explored.
Collapse
Affiliation(s)
- An Zhu
- Department of Toxicology, School of Public Health, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing 100191, China.
| | - Tao Zhang
- Department of Toxicology, School of Public Health, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing 100191, China.
| | - Qi Wang
- Department of Toxicology, School of Public Health, Peking University, No. 38 Xueyuan Road, Haidian District, Beijing 100191, China; Key Laboratory of State Administration of Traditional Chinese Medicine for Compatibility Toxicology, Beijing 100191, China; Beijing Key Laboratory of Toxicological Research and Risk Assessment for Food Safety, Beijing 100191, China.
| |
Collapse
|
20
|
Ando T, Nagumo M, Ninomiya M, Tanaka K, Linhardt RJ, Koketsu M. Synthesis of coumarin derivatives and their cytoprotective effects on t -BHP-induced oxidative damage in HepG2 cells. Bioorg Med Chem Lett 2018; 28:2422-2425. [DOI: 10.1016/j.bmcl.2018.06.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2018] [Revised: 06/02/2018] [Accepted: 06/11/2018] [Indexed: 02/08/2023]
|
21
|
Rubio V, García-Pérez AI, Herráez A, Tejedor MC, Diez JC. Esculetin modulates cytotoxicity induced by oxidants in NB4 human leukemia cells. ACTA ACUST UNITED AC 2017; 69:700-712. [DOI: 10.1016/j.etp.2017.08.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2016] [Revised: 08/25/2017] [Accepted: 08/30/2017] [Indexed: 11/26/2022]
|
22
|
Esculetin Neutralises Cytotoxicity of t-BHP but Not of H 2O 2 on Human Leukaemia NB4 Cells. BIOMED RESEARCH INTERNATIONAL 2017; 2017:9491045. [PMID: 28367450 PMCID: PMC5359438 DOI: 10.1155/2017/9491045] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 02/09/2017] [Indexed: 01/27/2023]
Abstract
The coumarin esculetin shows antioxidant action on some cell types, both by scavenging ROS and by decreasing ROS production. We have previously demonstrated the induction of apoptosis by esculetin on NB4 human leukaemia cells by an ill-defined mechanism related to ROS levels. In this work, we analyze the effect of the simultaneous treatment with esculetin and two oxidants to observe the early events in the mechanism of esculetin-induced apoptosis. Our results show that, from the early time of 15 min, esculetin acts synergistically with H2O2 to decrease cell viability and metabolic activity and to increase apoptosis in NB4 cells. In contrast, the early oxidative effects of t-BHP are neutralised by esculetin, protecting human leukaemia NB4 cells from apoptosis. Esculetin seems to restrict the increase in peroxides caused by H2O2 or t-BHP in the time interval analyzed. These results contribute to a better understanding of the cytotoxic effect caused by esculetin on NB4 cells. At the same time, the early neutralisation of exogenous oxidants could be of interest to prevent diseases related to oxidative stress imbalance.
Collapse
|
23
|
Liang C, Ju W, Pei S, Tang Y, Xiao Y. Pharmacological Activities and Synthesis of Esculetin and Its Derivatives: A Mini-Review. Molecules 2017; 22:E387. [PMID: 28257115 PMCID: PMC6155195 DOI: 10.3390/molecules22030387] [Citation(s) in RCA: 73] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2017] [Revised: 02/20/2017] [Accepted: 02/24/2017] [Indexed: 11/17/2022] Open
Abstract
Esculetin, synonymous with 6,7-dihydroxycoumarin, is the main active ingredient of the traditional Chinese medicine Cortex Fraxini. The twig skin or trunk bark of Cortex Fraxini are used by herb doctors as a mild, bitter liver and gallbladder meridians' nontoxic drug as well as dietary supplement. Recently, with a variety of novel esculetin derivatives being reported, the molecular mechanism research as well as clinical application of Cortex Fraxini and esculetin are becoming more attractive. This mini-review will consolidate what is known about the biological activities, the mechanism of esculetin and its synthetic derivatives over the past decade in addition to providing a brief synopsis of the properties of esculetin.
Collapse
Affiliation(s)
- Chengyuan Liang
- Faculty of Pharmacy, Shaanxi University of Science & Technology, 6 Xuefu Road, Xi'an 710021, China.
| | - Weihui Ju
- Faculty of Pharmacy, Shaanxi University of Science & Technology, 6 Xuefu Road, Xi'an 710021, China.
| | - Shaomeng Pei
- Faculty of Pharmacy, Shaanxi University of Science & Technology, 6 Xuefu Road, Xi'an 710021, China.
| | - Yonghong Tang
- Xi'an Taikomed Pharmaceutical technology Co., LTD, Xi'an 710077, China.
| | - Yadong Xiao
- Faculty of Finance and Economics, Guangzhou Vocational College of Science and Technology, Guangzhou 510550, China.
| |
Collapse
|
24
|
Kadakol A, Sharma N, Kulkarni YA, Gaikwad AB. Esculetin: A phytochemical endeavor fortifying effect against non-communicable diseases. Biomed Pharmacother 2016; 84:1442-1448. [DOI: 10.1016/j.biopha.2016.10.072] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/18/2016] [Accepted: 10/24/2016] [Indexed: 02/06/2023] Open
|
25
|
Choi RY, Ham JR, Lee MK. Esculetin prevents non-alcoholic fatty liver in diabetic mice fed high-fat diet. Chem Biol Interact 2016; 260:13-21. [PMID: 27769711 DOI: 10.1016/j.cbi.2016.10.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2016] [Revised: 09/22/2016] [Accepted: 10/17/2016] [Indexed: 12/19/2022]
Abstract
This study investigated the effects and mechanism of esculetin (6,7-dihydroxycoumarin) on non-alcoholic fatty liver in diabetic mice fed high-fat diet (HFD). The diabetic mice model was induced by injection of streptozotocin, after which they were fed HFD diet with or without esculetin for 11 weeks. Non-diabetic mice were provided a normal diet. Diabetes induced hepatic hypertrophy, lipid accumulation and droplets; however, esculetin reversed these changes. Esculetin treatment in diabetic mice fed HFD significantly down-regulated expression of lipid synthesis genes (Fasn, Dgat2 and Plpp2) and inflammation genes (Tlr4, Myd88, Nfkb, Tnfα and Il6). Moreover, the activities of hepatic lipid synthesis enzymes (fatty acid synthase and phosphatidate phosphohydrolase) and gluconeogenesis enzyme (glucose-6-phosphatase) in the esculetin group were decreased compared with the diabetic group. In addition, esculetin significantly reduced blood HbA1c, serum cytokines (TNF-α and IL-6) and chemokine (MCP-1) levels compared with the diabetic group without changing the insulin content in serum and the pancreas. Hepatic SOD activity was lower and lipid peroxidation level was higher in the diabetic group than in the normal group; however, esculetin attenuates these differences. Overall, these results demonstrated that esculetin supplementation could protect against development of non-alcoholic fatty liver in diabetes via regulation of lipids, glucose and inflammation.
Collapse
Affiliation(s)
- Ra-Yeong Choi
- Department of Food and Nutrition, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Ju Ri Ham
- Department of Food and Nutrition, Sunchon National University, Suncheon, 57922, Republic of Korea
| | - Mi-Kyung Lee
- Department of Food and Nutrition, Sunchon National University, Suncheon, 57922, Republic of Korea.
| |
Collapse
|
26
|
Kim KH, Park H, Park HJ, Choi KH, Sadikot RT, Cha J, Joo M. Glycosylation enables aesculin to activate Nrf2. Sci Rep 2016; 6:29956. [PMID: 27417293 PMCID: PMC4945939 DOI: 10.1038/srep29956] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/28/2016] [Indexed: 02/08/2023] Open
Abstract
Since aesculin, 6,7-dihydroxycoumarin-6-O-β-glucopyranoside, suppresses inflammation, we asked whether its anti-inflammatory activity is associated with the activation of nuclear factor-E2-related factor 2 (Nrf2), a key anti-inflammatory factor. Our results, however, show that aesculin marginally activated Nrf2. Since glycosylation can enhance the function of a compound, we then asked whether adding a glucose makes aesculin activate Nrf2. Our results show that the glycosylated aesculin, 3-O-β-d-glycosyl aesculin, robustly activated Nrf2, inducing the expression of Nrf2-dependent genes, such as heme oxygenase-1, glutamate-cysteine ligase catalytic subunit, and NAD(P)H quinone oxidoreductase 1 in macrophages. Mechanistically, 3-O-β-d-glycosyl aesculin suppressed ubiquitination of Nrf2, retarding degradation of Nrf2. Unlike aesculin, 3-O-β-d-glycosyl aesculin significantly suppressed neutrophilic lung inflammation, a hallmark of acute lung injury (ALI), in mice, which was not recapitulated in Nrf2 knockout mice, suggesting that the anti-inflammatory function of the compound largely acts through Nrf2. In a mouse model of sepsis, a major cause of ALI, 3-O-β-d-glycosyl aesculin significantly enhanced the survival of mice, compared with aesculin. Together, these results show that glycosylation could confer the ability to activate Nrf2 on aesculin, enhancing the anti-inflammatory function of aesculin. These results suggest that glycosylation can be a way to improve or alter the function of aesculin.
Collapse
Affiliation(s)
- Kyun Ha Kim
- School of Korean Medicine, Pusan National University, Yangsan 626-870, Korea
| | - Hyunsu Park
- Department of Microbiology, Pusan National University, Busan 609-735, Korea
| | - Hee Jin Park
- Department of Microbiology, Pusan National University, Busan 609-735, Korea
| | - Kyoung-Hwa Choi
- Department of Microbiology, Pusan National University, Busan 609-735, Korea
| | - Ruxana T Sadikot
- Section of Pulmonary and Critical Care Medicine, Atlanta Veterans Affairs Medical Center, Emory University, Decatur, GA30033, USA
| | - Jaeho Cha
- Department of Microbiology, Pusan National University, Busan 609-735, Korea
| | - Myungsoo Joo
- School of Korean Medicine, Pusan National University, Yangsan 626-870, Korea
| |
Collapse
|
27
|
Karnewar S, Vasamsetti SB, Gopoju R, Kanugula AK, Ganji SK, Prabhakar S, Rangaraj N, Tupperwar N, Kumar JM, Kotamraju S. Mitochondria-targeted esculetin alleviates mitochondrial dysfunction by AMPK-mediated nitric oxide and SIRT3 regulation in endothelial cells: potential implications in atherosclerosis. Sci Rep 2016; 6:24108. [PMID: 27063143 PMCID: PMC4827087 DOI: 10.1038/srep24108] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2015] [Accepted: 03/21/2016] [Indexed: 01/10/2023] Open
Abstract
Mitochondria-targeted compounds are emerging as a new class of drugs that can potentially alter the pathophysiology of those diseases where mitochondrial dysfunction plays a critical role. We have synthesized a novel mitochondria-targeted esculetin (Mito-Esc) with an aim to investigate its effect during oxidative stress-induced endothelial cell death and angiotensin (Ang)-II-induced atherosclerosis in ApoE−/− mice. Mito-Esc but not natural esculetin treatment significantly inhibited H2O2- and Ang-II-induced cell death in human aortic endothelial cells by enhancing NO production via AMPK-mediated eNOS phosphorylation. While L-NAME (NOS inhibitor) significantly abrogated Mito-Esc-mediated protective effects, Compound c (inhibitor of AMPK) significantly decreased Mito-Esc-mediated increase in NO production. Notably, Mito-Esc promoted mitochondrial biogenesis by enhancing SIRT3 expression through AMPK activation; and restored H2O2-induced inhibition of mitochondrial respiration. siSIRT3 treatment not only completely reversed Mito-Esc-mediated mitochondrial biogenetic marker expressions but also caused endothelial cell death. Furthermore, Mito-Esc administration to ApoE−/− mice greatly alleviated Ang-II-induced atheromatous plaque formation, monocyte infiltration and serum pro-inflammatory cytokines levels. We conclude that Mito-Esc is preferentially taken up by the mitochondria and preserves endothelial cell survival during oxidative stress by modulating NO generation via AMPK. Also, Mito-Esc-induced SIRT3 plays a pivotal role in mediating mitochondrial biogenesis and perhaps contributes to its anti-atherogenic effects.
Collapse
Affiliation(s)
- Santosh Karnewar
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| | - Sathish Babu Vasamsetti
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| | - Raja Gopoju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| | | | - Sai Krishna Ganji
- National Centre for Mass Spectrometry, Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Sripadi Prabhakar
- National Centre for Mass Spectrometry, Indian Institute of Chemical Technology, Hyderabad, 500007, India
| | - Nandini Rangaraj
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Nitin Tupperwar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Jerald Mahesh Kumar
- CSIR-Centre for Cellular and Molecular Biology, Uppal Road, Hyderabad, 500007, India
| | - Srigiridhar Kotamraju
- Centre for Chemical Biology, CSIR-Indian Institute of Chemical Technology, Uppal Road, Hyderabad, 500007, India.,Academy of Scientific and Innovative Research, Training and Development Complex, Chennai, India
| |
Collapse
|
28
|
Park SL, Won SY, Song JH, Lee SY, Kim WJ, Moon SK. Esculetin Inhibits VEGF-Induced Angiogenesis Both In Vitro and In Vivo. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2016; 44:61-76. [DOI: 10.1142/s0192415x1650004x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Esculetin is known to inhibit tumor growth, but its effect in angiogenesis has not been studied. Here, we report the efficacy of esculetin on VEGF-induced angiogenesis. Esculetin treatment inhibited VEGF-induced proliferation and DNA synthesis of HUVECs with no cell toxicity. G1-phase cell-cycle arrest was associated with a decreased expression of cyclins and CDKs via the binding of p27KIP1. Esculetin down-regulated the MMP-2 expression in VEGF-stimulated HUVECs, which suppressed colony tube formation and migration. Esculetin reduced the phosphorylation of VEGFR-2 and the downstream signaling of VEGFR-2, including ERK1/2 and eNOS/Akt pathways. Esculetin suppressed microvessel outgrowth from an aortic ring ex vivo model treated with VEGF, and blocked the VEGF-induced formation of new blood vessels and hemoglobin content in an in vivo Matrigel plug model. Collectively, VEGF-stimulated responses in angiogenesis were inhibited in vitro and in vivo, providing a theoretical basis for effective use against anti-angiogenic therapies.
Collapse
Affiliation(s)
- Sung Lyea Park
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Se Yeon Won
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Jun-Hui Song
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Sook-Young Lee
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Republic of Korea
| | - Wun-Jae Kim
- Personalized Tumor Engineering Research Center, Department of Urology, Chungbuk National University, Cheongju 361-763, Republic of Korea
| | - Sung-Kwon Moon
- Department of Food and Nutrition, Chung-Ang University, Anseong 456-756, Republic of Korea
| |
Collapse
|
29
|
Kim CS, Kim J, Lee YM, Sohn E, Kim JS. Esculetin, a Coumarin Derivative, Inhibits Aldose Reductase Activity in vitro and Cataractogenesis in Galactose-Fed Rats. Biomol Ther (Seoul) 2016; 24:178-83. [PMID: 26902086 PMCID: PMC4774499 DOI: 10.4062/biomolther.2015.101] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2015] [Revised: 10/24/2015] [Accepted: 11/20/2015] [Indexed: 11/05/2022] Open
Abstract
Naturally occurring coumarin compounds have received substantial attention due to their pharmaceutical effects. Esculetin is a coumarin derivative and a polyphenol compound that is used in a variety of therapeutic and pharmacological strategies. However, its effect on aldose reductase activity remains poorly understood. In this study, the potential beneficial effects of esculetin on lenticular aldose reductase were investigated in galactose-fed (GAL) rats, an animal model of sugar cataracts. Cataracts were induced in Sprague-Dawley (SD) rats via a 50% galactose diet for 2 weeks, and groups of GAL rats were orally treated with esculetin (10 or 50 mg/kg body weight). In vehicle-treated GAL rats, lens opacification was observed, and swelling and membrane rupture of the lens fiber cells were increased. Additionally, aldose reductase was highly expressed in the lens epithelium and superficial cortical fibers during cataract development in the GAL rats. Esculetin reduced rat lens aldose reductase (RLAR) activity in vitro, and esculetin treatment significantly inhibited lens opacity, as well as morphological alterations, such as swelling, vacuolation and liquefaction of lens fibers, via the inhibition of aldose reductase in the GAL rats. These results indicate that esculetin is a useful treatment for galactose-induced cataracts.
Collapse
Affiliation(s)
- Chan-Sik Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Junghyun Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Yun Mi Lee
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Eunjin Sohn
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| | - Jin Sook Kim
- Korean Medicine Convergence Research Division, Korea Institute of Oriental Medicine, Daejeon 34054, Republic of Korea
| |
Collapse
|
30
|
Pan H, Wang BH, Lv W, Jiang Y, He L. Esculetin induces apoptosis in human gastric cancer cells through a cyclophilin D-mediated mitochondrial permeability transition pore associated with ROS. Chem Biol Interact 2015; 242:51-60. [PMID: 26388407 DOI: 10.1016/j.cbi.2015.09.015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Revised: 08/20/2015] [Accepted: 09/16/2015] [Indexed: 01/20/2023]
Abstract
Esculetin is a coumarin derivative from natural plants that has been commonly used as a folk medicine and has been reported to have beneficial pharmacological and biochemical activities; however, the mechanism by which esculetin prevents human gastric cancer cell growth is still largely unknown. In this study, we investigated the effect of esculetin on human gastric cancer cells and explored the cell death mechanism. Our data indicated that esculetin inhibited the growth of human gastric cancer cells in a dose- and time-dependent manner and apoptosis was the main cause of decreased cell viability in esculetin-treated cells. Additionally, esculetin treatment increased the activity of caspase-9 and caspase-3, and resulted in the appearance of the PARP cleavage product; and esculetin-induced cell death and apoptosis was decreased by pretreatment with CsA and NAC, but not BA; these results demonstrate that esculetin induced apoptosis via the caspase-dependent mitochondrial pathway in human gastric cancer cells in which cyclophilin D mediated the cytotoxic action by triggering the opening of the mitochondrial permeability transition pore; and the generation of ROS not only was a consequence of mitochondrial dysfunction, but also triggered esculetin-induced apoptosis. These results reveal a novel mechanism of esculetin on gastric cancer cells and suggest that esculetin could be a novel agent in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Hui Pan
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, China.
| | - Bao-Hui Wang
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Wang Lv
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, China
| | - Yan Jiang
- Zhejiang Hospital of Traditional Chinese Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Lei He
- The First Affiliated Hospital, College of Medicine, Zhejiang University, Qingchun Road 79, Hangzhou, China
| |
Collapse
|
31
|
Esculetin, a natural coumarin compound, evokes Ca2+ movement and activation of Ca2+-associated mitochondrial apoptotic pathways that involved cell cycle arrest in ZR-75-1 human breast cancer cells. Tumour Biol 2015; 37:4665-78. [DOI: 10.1007/s13277-015-4286-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 10/19/2015] [Indexed: 11/24/2022] Open
|
32
|
Anti-Inflammatory activities of licorice extract and its active compounds, glycyrrhizic acid, liquiritin and liquiritigenin, in BV2 cells and mice liver. Molecules 2015. [PMID: 26205049 PMCID: PMC6332102 DOI: 10.3390/molecules200713041] [Citation(s) in RCA: 150] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
This study provides the scientific basis for the anti-inflammatory effects of licorice extract in a t-BHP (tert-butyl hydrogen peroxide)-induced liver damage model and the effects of its ingredients, glycyrrhizic acid (GA), liquiritin (LQ) and liquiritigenin (LG), in a lipopolysaccharide (LPS)-stimulated microglial cell model. The GA, LQ and LG inhibited the LPS-stimulated elevation of pro-inflammatory mediators, such as inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), tumor necrosis factor (TNF)-alpha, interleukin (IL)-1beta and interleukin (IL)-6 in BV2 (mouse brain microglia) cells. Furthermore, licorice extract inhibited the expression levels of pro-inflammatory cytokines (TNF-α, IL-1β and IL-6) in the livers of t-BHP-treated mice models. This result suggested that mechanistic-based evidence substantiating the traditional claims of licorice extract and its three bioactive components can be applied for the treatment of inflammation-related disorders, such as oxidative liver damage and inflammation diseases.
Collapse
|
33
|
Baek KS, Ahn S, Lee J, Kim JH, Kim HG, Kim E, Kim JH, Sung NY, Yang S, Kim MS, Hong S, Kim JH, Cho JY. Immunotoxicological Effects of Aripiprazole: In vivo and In vitro Studies. THE KOREAN JOURNAL OF PHYSIOLOGY & PHARMACOLOGY : OFFICIAL JOURNAL OF THE KOREAN PHYSIOLOGICAL SOCIETY AND THE KOREAN SOCIETY OF PHARMACOLOGY 2015; 19:365-72. [PMID: 26170741 PMCID: PMC4499649 DOI: 10.4196/kjpp.2015.19.4.365] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2015] [Revised: 05/15/2015] [Accepted: 06/02/2015] [Indexed: 12/20/2022]
Abstract
Aripiprazole (ARI) is a commonly prescribed medication used to treat schizophrenia and bipolar disorder. To date, there have been no studies regarding the molecular pathological and immunotoxicological profiling of aripiprazole. Thus, in the present study, we prepared two different formulas of aripiprazole [Free base crystal of aripiprazole (ARPGCB) and cocrystal of aripiprazole (GCB3004)], and explored their effects on the patterns of survival and apoptosis-regulatory proteins under acute toxicity and cytotoxicity test conditions. Furthermore, we also evaluated the modulatory activity of the different formulations on the immunological responses in macrophages primed by various stimulators such as lipopolysaccharide (LPS), pam3CSK, and poly(I:C) via toll-like receptor 4 (TLR4), TLR2, and TLR3 pathways, respectively. In liver, both ARPGCB and GCB3004 produced similar toxicity profiles. In particular, these two formulas exhibited similar phospho-protein profiling of p65/nuclear factor (NF)-κB, c-Jun/activator protein (AP)-1, ERK, JNK, p38, caspase 3, and bcl-2 in brain. In contrast, the patterns of these phospho-proteins were variable in other tissues. Moreover, these two formulas did not exhibit any cytotoxicity in C6 glioma cells. Finally, the two formulations at available in vivo concentrations did not block nitric oxide (NO) production from activated macrophage-like RAW264.7 cells stimulated with LPS, pam3CSK, or poly(I:C), nor did they alter the morphological changes of the activated macrophages. Taken together, our present work, as a comparative study of two different formulas of aripiprazole, suggests that these two formulas can be used to achieve similar functional activation of brain proteins related to cell survival and apoptosis and immunotoxicological activities of macrophages.
Collapse
Affiliation(s)
- Kwang-Soo Baek
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | | | - Jaehwi Lee
- College of Pharmacy, Chung-Ang University, Seoul 156-756, Korea
| | - Ji Hye Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Han Gyung Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Eunji Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Jun Ho Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Nak Yoon Sung
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Sungjae Yang
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Mi Seon Kim
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Sungyoul Hong
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| | - Jong-Hoon Kim
- Department of Veterinary Physiology, College of Veterinary Medicine, Biosafety Research Institute, Chonbuk National University, Jeonju 561-756, Korea
| | - Jae Youl Cho
- Department of Genetic Engineering, Sungkyunkwan University, Suwon 440-746, Korea
| |
Collapse
|
34
|
Protective effect of carvacrol from Thymus quinquecostatus Celak against tert-butyl hydroperoxide-induced oxidative damage in Chang cells. Food Sci Biotechnol 2015. [DOI: 10.1007/s10068-015-0095-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
|
35
|
Kim Y, Park Y, Namkoong S, Lee J. Esculetin inhibits the inflammatory response by inducing heme oxygenase-1 in cocultured macrophages and adipocytes. Food Funct 2015; 5:2371-7. [PMID: 25088305 DOI: 10.1039/c4fo00351a] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Obesity is associated with chronic low-grade inflammation of adipose tissue. In this study, we investigated the anti-inflammatory effects of esculetin (ECT) through up-regulation of heme oxygenase-1 (HO-1) in cocultured macrophages and adipocytes. RAW264.7 macrophages and differentiated 3T3-L1 adipocytes were cocultured in serum-free Dulbecco's modified Eagle's medium with or without ECT for 24 h. Nitric oxide (NO), tumor necrosis factor-α (TNF-α), and monocyte chemoattractant protein-1 (MCP-1) production was measured in the coculture supernatant. ECT decreased the secretion of NO, TNF-α, and MCP-1. The expression of adipogenic proteins, including peroxisome proliferator-activated receptors γ (PPARγ) and CCAAT/enhancer binding protein α (C/EBPα) in cocultured adipocytes and inducible nitric oxide synthase (iNOS) in cocultured macrophages, was inhibited by ECT. Additionally, HO-1 expression was induced in cocultured macrophages and adipocytes. Silencing of HO-1 expression increased the production of NO, TNF-α, and MCP-1 in cocultured cells, in spite of the presence of ECT. This study demonstrated that ECT exhibited anti-inflammatory properties by inhibiting the production of proinflammatory cytokines in the interaction between adipocytes and macrophages through HO-1 expression. ECT may have the potential to improve chronic inflammation in obesity.
Collapse
Affiliation(s)
- Younghwa Kim
- Department of Food Science and Technology, College of Agriculture, Life and Environment Sciences, Chungbuk National University, 52 Naesudong-ro, Heungdeok-gu, Cheongju, Chungbuk 361-763, Korea.
| | | | | | | |
Collapse
|
36
|
Zhu L, Lu L, Zeng S, Luo F, Dai P, Wu P, Wang Y, Liu L, Hu M, Liu Z. UDP-Glucuronosyltransferases 1A6 and 1A9 are the Major Isozymes Responsible for the 7-O-Glucuronidation of Esculetin and 4-Methylesculetin in Human Liver Microsomes. Drug Metab Dispos 2015; 43:977-83. [PMID: 25854527 DOI: 10.1124/dmd.115.063552] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 04/08/2015] [Indexed: 11/22/2022] Open
Abstract
Esculetin (6,7-dihydroxycoumarin, ET) and 4-methylesculetin (6,7-dihydroxy-4-methylcoumarin, 4-ME) are typical coumarin derivatives that are attracting considerable attention because of their wide spectrum of biologic activities, but their metabolism remains unknown. This study aimed to elucidate the in vitro UDP-glucuronosyltransferase (UGT) metabolism characteristics of ET and 4-ME. 7-O-monoglucuronide esculetin (ET-G) and 7-O-monoglucuronide 4-methylesculetin (4-ME-G) were identified by liquid chromatography-mass spectrometry (LC-MS) and (1)H-nuclear magnetic resonance ((1)HNMR) when ET or 4-ME was incubated with human liver (HLM) in the presence of UDP-glucuronic acid. Screening assays with 12 human expressed UGTs demonstrated that the formations of ET-G and 4-ME-G were almost exclusively catalyzed by UGT1A6 and UGT1A9. Phenylbutazone and carvacrol (UGT1A6 and UGT1A9 chemical inhibitors, respectively) at different concentrations (50, 100, and 200 μM) significantly inhibited the formation of glucuronidates of ET and 4-ME in HLM, UGT1A6, and UGT1A9 when the concentrations of ET and 4-ME ranged from 10 to 300 μM (P < 0.05). Clearance rates of ET in HLM, HIM, UGT1A6, and UGT1A9 were 0.54, 0.16, 0.69, and 0.14 ml/min/mg, respectively. Corresponding clearance rates values of 4-ME were 0.59, 0.03, 0.14, and 0.04 ml/min/mg, respectively. In conclusion, 7-O-monoglucuronidation by UGT1A6 and UGT1A9 was the predominant UGT metabolic pathway for both ET and 4-ME in vitro. The liver is probably the major contributor to the glucuronidation metabolism of ET and 4-ME. ET showed more rapid metabolism than 4-ME in glucuronidation.
Collapse
Affiliation(s)
- Lijun Zhu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Linlin Lu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Shan Zeng
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Feifei Luo
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Peimin Dai
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Peng Wu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Ying Wang
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Liang Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Ming Hu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| | - Zhongqiu Liu
- International Institute for Translational Chinese Medicine, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, PR China (Z.L.J., L.L.L., Z.S., L.F.F., D.P.M., W.P., W.Y., H.M., L.Z.Q.); Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, Texas (H.M.); Department of Pharmaceutics, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, Guangdong, PR China (D.P.M., W.Y.); and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau (SAR), PR China (L.L.)
| |
Collapse
|
37
|
Łuczaj W, Jarocka-Karpowicz I, Bielawska K, Skrzydlewska E. Sweet grass protection against oxidative stress formation in the rat brain. Metab Brain Dis 2015; 30:183-90. [PMID: 25108595 PMCID: PMC4544646 DOI: 10.1007/s11011-014-9599-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 08/04/2014] [Indexed: 12/15/2022]
Abstract
The aims of this study were to investigate the influences of sweet grass on chronic ethanol-induced oxidative stress in the rat brain. Chronic ethanol intoxication decreased activities and antioxidant levels resulting in enhanced lipid peroxidation. Administration of sweet grass solution to ethanol-intoxicated rats partially normalized the activity activities of Cu,Zn-superoxide dismutase, catalase, glutathione peroxidase, and glutathione reductase, as well as levels of reduced glutathione and vitamins C, E, and A. Sweet grass also protected unsaturated fatty acids (arachidonic and docosahexaenoic) from oxidations and decreased levels of lipid peroxidation products: 4-hydroxynonenal, isoprostanes, and neuroprostanes. The present in vivo study confirms previous in vitro data demonstrating the bioactivity of sweet grass and suggests a possible role for sweet grass in human health protection from deleterious consequences associated with oxidative stress formation.
Collapse
|
38
|
Rzodkiewicz P, Gasinska E, Maslinski S, Bujalska-Zadrozny M. Antinociceptive properties of esculetin in non-inflammatory and inflammatory models of pain in rats. Clin Exp Pharmacol Physiol 2015; 42:213-9. [DOI: 10.1111/1440-1681.12346] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2014] [Revised: 11/12/2014] [Accepted: 11/18/2014] [Indexed: 11/30/2022]
Affiliation(s)
- Przemyslaw Rzodkiewicz
- Department of Biochemistry and Molecular Biology; Institute of Rheumatology; Warsaw Poland
- Department of Pharmacodynamics; Centre for Preclinical Research and Technology; Medical University of Warsaw; Warsaw Poland
| | - Emilia Gasinska
- Department of Pharmacodynamics; Centre for Preclinical Research and Technology; Medical University of Warsaw; Warsaw Poland
| | - Slawomir Maslinski
- Department of General and Experimental Pathology; Medical University of Warsaw; Warsaw Poland
| | - Magdalena Bujalska-Zadrozny
- Department of Pharmacodynamics; Centre for Preclinical Research and Technology; Medical University of Warsaw; Warsaw Poland
| |
Collapse
|
39
|
Ferreres F, Bernardo J, Andrade PB, Sousa C, Gil-Izquierdo A, Valentão P. Pennyroyal and gastrointestinal cells: multi-target protection of phenolic compounds against t-BHP-induced toxicity. RSC Adv 2015. [DOI: 10.1039/c5ra02710a] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Pennyroyal infusion maintains gastrointestinal cells antioxidant balance.
Collapse
Affiliation(s)
- F. Ferreres
- Research Group on Quality
- Safety and Bioactivity of Plant Foods
- Department of Food Science and Technology
- CEBAS (CSIC)
- Murcia
| | - J. Bernardo
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| | - P. B. Andrade
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| | - C. Sousa
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| | - A. Gil-Izquierdo
- Research Group on Quality
- Safety and Bioactivity of Plant Foods
- Department of Food Science and Technology
- CEBAS (CSIC)
- Murcia
| | - P. Valentão
- REQUIMTE/LAQV
- Laboratório de Farmacognosia
- Departamento de Química
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
40
|
Kim Y, Lee J. Esculetin, a coumarin derivative, suppresses adipogenesis through modulation of the AMPK pathway in 3T3-L1 adipocytes. J Funct Foods 2015. [DOI: 10.1016/j.jff.2014.12.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022] Open
|
41
|
Molecularly imprinted polymer for pre-concentration of esculetin from tobacco followed by the UPLC analysis. Sci China Chem 2014. [DOI: 10.1007/s11426-014-5180-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
42
|
Nath M, Vats M, Roy P. Design, spectral characterization, anti-tumor and anti-inflammatory activity of triorganotin(IV) hydroxycarboxylates, apoptosis inducers: In vitro assessment of induction of apoptosis by enzyme, DNA-fragmentation, acridine orange and comet assays. Inorganica Chim Acta 2014. [DOI: 10.1016/j.ica.2014.02.034] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
43
|
CHO JINHYOUNG, SHIN JAECHEON, CHO JUNGJAE, CHOI YUNGHYUN, SHIM JUNGHYUN, CHAE JUNGIL. Esculetin (6,7-dihydroxycoumarin): A potential cancer chemopreventive agent through suppression of Sp1 in oral squamous cancer cells. Int J Oncol 2014; 46:265-71. [DOI: 10.3892/ijo.2014.2700] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Accepted: 09/10/2014] [Indexed: 11/05/2022] Open
|
44
|
Fan D, Zhao Y, Zhou X, Gong X, Zhao C. Simultaneous determination of esculetin, quercetin-3-O-β-D-glucuronide, quercetin-3-O-β -D-glucuronopyranside methyl ester and quercetin in effective part of Polygonum Perfoliatum L. using high performace liquid chromatography. Pharmacogn Mag 2014; 10:359-66. [PMID: 25210326 PMCID: PMC4159932 DOI: 10.4103/0973-1296.137379] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2013] [Revised: 11/20/2013] [Accepted: 07/24/2014] [Indexed: 11/29/2022] Open
Abstract
Objective: In the present study, a high performance liquid chromatography (HPLC) coupled with photodiode array detection was developed for simultaneous quantitation of esculetin, quercetin-3-O-β-D-glucuronide, quercetin-3-O-β-D- glucuronopyranoside methyl ester and quercetin in Polygonum perfoliatum L. Materials and Methods: The chromatographic separations were performed on a reversed-phase C18 column using a mobile phase composed of acetonitrile -0.5% aqueous acetic acid with gradient elution. The calibration curves for the analytes demonstrated good linearities within the investigated ranges. The satisfactory intra- and inter-day precision, repeatability and stability of the developed analytical method were shown in the method validation procedure. The recoveries of the established method ranged from 95.76 to 102.10% for all the analytes. Results: This proposed method was successfully applied for simultaneous quantification of the four compounds in effective part of Polygonum perfoliatum L. from different regions. Hierarchical clustering analysis (HCA) and principal components analysis (PCA) were performed to characterize and classify the samples based on the contents of the four compounds in Polygonum perfoliatum L. Conclusion: The established HPLC method combined with chemometric approaches was proven to be useful and efficient for quality control of Polygonum perfoliatum L.
Collapse
Affiliation(s)
- Dongsheng Fan
- The Research Center for Quality Control of Natural Medicine, Guiyang, China
| | - Yang Zhao
- The Research Center for Quality Control of Natural Medicine, Guiyang, China ; Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Enviroment, Guizhou Normal University, Guiyang, China
| | - Xin Zhou
- The Research Center for Quality Control of Natural Medicine, Guiyang, China ; Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Enviroment, Guizhou Normal University, Guiyang, China
| | - Xiaojian Gong
- The Research Center for Quality Control of Natural Medicine, Guiyang, China ; Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Enviroment, Guizhou Normal University, Guiyang, China
| | - Chao Zhao
- The Research Center for Quality Control of Natural Medicine, Guiyang, China ; Key Laboratory for Information System of Mountainous Areas and Protection of Ecological Enviroment, Guizhou Normal University, Guiyang, China
| |
Collapse
|
45
|
V G M N, Atmakur H, Katragadda SB, Devabakthuni B, Kota A, S CKR, Kuncha M, M V P S VV, Kulkarni P, Janaswamy MR, Sistla R. Antioxidant, hepatoprotective and cytotoxic effects of icetexanes isolated from stem-bark of Premna tomentosa. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2014; 21:497-505. [PMID: 24183951 DOI: 10.1016/j.phymed.2013.09.025] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 08/16/2013] [Accepted: 09/29/2013] [Indexed: 05/04/2023]
Abstract
The study investigates the antioxidant, hepatoprotective and antiproliferative effects of novel icetexane diterpenoids (ice 1-4) isolated from hexane extract of stem bark of Premna tomentosa. A549, HT-29, MCF-7, MDA-MB-231, A431 cells were used to assess the antiproliferative activity by MTT assay. Cell death induced by apoptosis was determined by morphological assessment studies using acridine orange/ethidium bromide staining (dual staining), mitochondrial potential measurement by JC-1 staining, and cell cycle analysis by propidium iodide staining method by Muse cell analyser. Anti oxidant activity was investigated by in vitro assays such as DPPH, nitric oxide and superoxide scavenging activities. Hepatoprotective activity was determined in vitro with HepG2 cells and in vivo by tBHP induced hepatic damage mice model. Based on the in vitro cytotoxic assays and morphological assessment studies using fluorescence microscopic study (acridine orange and ethidium bromide double staining) and mitochondrial potential measurements, it was found that ice 2 and 3 possess good antiproliferative effect via mitochondrial mediated apoptosis in human lung and breast cancer cells. Results of in vitro antioxidant studies demonstrated that ice-4 has showed good antioxidant activity. The restoration of serum levels of SGOT, SGPT and ALKP, liver GSH status and reduction or inhibition of lipid peroxidation in liver of tBHP intoxicated mice after administration of ice-4 at dose of 250mg/kg indicated its potential use for hepatoprotective activity.
Collapse
Affiliation(s)
- Naidu V G M
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad 500037, India
| | - Hymavathi Atmakur
- Division of Natural Product Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Suresh Babu Katragadda
- Division of Natural Product Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Bhavana Devabakthuni
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Anudeep Kota
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Chenna Keshava Reddy S
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Madhusudana Kuncha
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Vishnu Vardhan M V P S
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Prasad Kulkarni
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Madhusudana Rao Janaswamy
- Division of Natural Product Chemistry, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India
| | - Ramakrishna Sistla
- Division of Medicinal Chemistry and Pharmacology, CSIR-Indian Institute of Chemical Technology, Hyderabad 500007, India.
| |
Collapse
|
46
|
Kang KS, Lee W, Jung Y, Lee JH, Lee S, Eom DW, Jeon Y, Yoo HH, Jin MJ, Song KI, Kim WJ, Ham J, Kim HJ, Kim SN. Protective effect of esculin on streptozotocin-induced diabetic renal damage in mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2014; 62:2069-2076. [PMID: 24484395 DOI: 10.1021/jf403840c] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
The present study investigated the presence and mechanism of esculin-mediated renoprotection to assess its therapeutic potential. Esculin was orally administered at 20 mg/kg/day for 2 weeks to streptozotocin-induced diabetic mice, and its effects were compared with those of the vehicle in normal and diabetic mice. After oral administration of esculin to mice, the concentrations of esculin and esculetin in blood were 159.5 ± 29.8 and 9.7 ± 4.9 ng/mL at 30 min, respectively. Food and water intake were significantly increased in the diabetic mice compared to normal mice but attenuated in mice receiving esculin. The elevated blood glucose level and hepatic glucose-6-phosphatase expression were significantly reduced in esculin-treated diabetic mice, supporting the antidiabetic effect of esculin. Esculin also increased the uptake of glucose and induced the insulin-evoked phosphorylation of insulin receptor, Akt, and glycogen synthase kinase 3β in C2C12 myotubes, indicating a potential for improvement of insulin sensitivity. In addition, esculin lessened the elevated blood creatinine levels in diabetic mice and ameliorated diabetes-induced renal dysfunction by reducing caspase-3 activation in the kidney. Data support the beneficial effect of esculin against diabetes and oxidative stress-related inflammatory processes in the kidney.
Collapse
Affiliation(s)
- Ki Sung Kang
- Natural Medicine Center, Korea Institute of Science and Technology , Gangneung 210-340, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Rojas F, Cortes N, Abarzua S, Dyrda A, van Zundert B. Astrocytes expressing mutant SOD1 and TDP43 trigger motoneuron death that is mediated via sodium channels and nitroxidative stress. Front Cell Neurosci 2014; 8:24. [PMID: 24570655 PMCID: PMC3916762 DOI: 10.3389/fncel.2014.00024] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2013] [Accepted: 01/17/2014] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal paralytic disorder caused by dysfunction and degeneration of motor neurons. Multiple disease-causing mutations, including in the genes for SOD1 and TDP-43, have been identified in ALS. Astrocytes expressing mutant SOD1 are strongly implicated in the pathogenesis of ALS: we have shown that media conditioned by astrocytes carrying mutant SOD1G93A contains toxic factor(s) that kill motoneurons by activating voltage-sensitive sodium (Nav) channels. In contrast, a recent study suggests that astrocytes expressing mutated TDP43 contribute to ALS pathology, but do so via cell-autonomous processes and lack non-cell-autonomous toxicity. Here we investigate whether astrocytes that express diverse ALS-causing mutations release toxic factor(s) that induce motoneuron death, and if so, whether they do so via a common pathogenic pathway. We exposed primary cultures of wild-type spinal cord cells to conditioned medium derived from astrocytes (ACM) that express SOD1 (ACM-SOD1G93A and ACM-SOD1G86R) or TDP43 (ACM-TDP43A315T) mutants; we show that such exposure rapidly (within 30–60 min) increases dichlorofluorescein (DCF) fluorescence (indicative of nitroxidative stress) and leads to extensive motoneuron-specific death within a few days. Co-application of the diverse ACMs with anti-oxidants Trolox or esculetin (but not with resveratrol) strongly improves motoneuron survival. We also find that co-incubation of the cultures in the ACMs with Nav channel blockers (including mexiletine, spermidine, or riluzole) prevents both intracellular nitroxidative stress and motoneuron death. Together, our data document that two completely unrelated ALS models lead to the death of motoneuron via non-cell-autonomous processes, and show that astrocytes expressing mutations in SOD1 and TDP43 trigger such cell death through a common pathogenic pathway that involves nitroxidative stress, induced at least in part by Nav channel activity.
Collapse
Affiliation(s)
- Fabiola Rojas
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello Santiago, Chile
| | - Nicole Cortes
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello Santiago, Chile
| | - Sebastian Abarzua
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello Santiago, Chile
| | - Agnieszka Dyrda
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello Santiago, Chile
| | - Brigitte van Zundert
- Faculty of Biological Sciences and Faculty of Medicine, Center for Biomedical Research, Universidad Andres Bello Santiago, Chile
| |
Collapse
|
48
|
Medina ME, Galano A, Alvarez-Idaboy JR. Theoretical study on the peroxyl radicals scavenging activity of esculetin and its regeneration in aqueous solution. Phys Chem Chem Phys 2014; 16:1197-207. [DOI: 10.1039/c3cp53889c] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
49
|
Witaicenis A, Luchini A, Hiruma-Lima C, Felisbino S, Justulin L, Garrido-Mesa N, Utrilla P, Gálvez J, Di Stasi L. Mechanism and Effect of Esculetin in an Experimental Animal Model of Inflammatory Bowel Disease. EUR J INFLAMM 2013. [DOI: 10.1177/1721727x1301100213] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Affiliation(s)
- A. Witaicenis
- Department of Pharmacology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - A.C. Luchini
- Department of Pharmacology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - C.A. Hiruma-Lima
- Department of Physiology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - S.L. Felisbino
- Department of Morphology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - L.A. Justulin
- Department of Morphology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| | - N. Garrido-Mesa
- Department of Pharmacology, Centre for Biomedical Research, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EHD), University of Granada, Granada, Spain
| | - P. Utrilla
- Department of Pharmacology, Centre for Biomedical Research, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EHD), University of Granada, Granada, Spain
| | - J. Gálvez
- Department of Pharmacology, Centre for Biomedical Research, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBER-EHD), University of Granada, Granada, Spain
| | - L.C. Di Stasi
- Department of Pharmacology, Institute of Biosciences, Univ Estadual Paulista - UNESP, Botucatu, SP, Brazil
| |
Collapse
|
50
|
Slapšytė G, Dedonytė V, Lazutka JR, Mierauskienė J, Morkūnas V, Kazernavičiūtė R, Pukalskas A, Venskutonis PR. Evaluation of the biological activity of naturally occurring 5,8-dihydroxycoumarin. Molecules 2013; 18:4419-36. [PMID: 23591923 PMCID: PMC6269819 DOI: 10.3390/molecules18044419] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2013] [Revised: 03/21/2013] [Accepted: 04/09/2013] [Indexed: 11/16/2022] Open
Abstract
5,8-Dihydroxycoumarin (5,8-DHC) was isolated from aerial parts of sweet grass (Hierochloë odorata L.) and screened for antioxidant and genotoxic activities. A clear linear dependency of radical scavenging capacity in DPPH• and ABTS•+ assays was determined. 5,8-DHC was very efficient in retarding rapeseed oil oxidation (Oxipress test). TPC (total phenols content) and FRAP (the ability to reduce ferric ion to ferrous ion) assays revealed a somewhat lower antioxidant capacity of 5,8-DHC as compared with gallic acid. Genotoxic activity was tested using different genetic end-points: chromosome aberrations (CAs) and micronuclei (MN) in Wistar rat bone marrow in vivo, CAs and sister chromatid exchanges (SCEs) in human lymphocytes in vitro, and somatic mutations and recombination in Drosophila melanogaster wing cells in vivo. 5,8-DHC did not increase frequency of CAs in rat bone marrow cells, but induced a significant increase of MN. It was slightly mutagenic in the Drosophila melanogaster assay after 120 h of treatment, but not after 48 h of treatment. 5,8-DHC induced both CAs and SCEs in vitro in human lymphocytes in a clear dose-dependent manner. Thus, 5,8-DHC may be classified as weakly genotoxic both in vivo and in vitro.
Collapse
Affiliation(s)
- Gražina Slapšytė
- Department of Botany and Genetics, Vilnius University, M.K.Čiurlionis Str. 21/27, LT-03101 Vilnius, Lithuania; E-Mails: (G.S.); (V.D.); (J.R.L.); (J.M.); (V.M.)
| | - Veronika Dedonytė
- Department of Botany and Genetics, Vilnius University, M.K.Čiurlionis Str. 21/27, LT-03101 Vilnius, Lithuania; E-Mails: (G.S.); (V.D.); (J.R.L.); (J.M.); (V.M.)
| | - Juozas R. Lazutka
- Department of Botany and Genetics, Vilnius University, M.K.Čiurlionis Str. 21/27, LT-03101 Vilnius, Lithuania; E-Mails: (G.S.); (V.D.); (J.R.L.); (J.M.); (V.M.)
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +370-5-239-8257; Fax: +370-5-239-8204
| | - Jūratė Mierauskienė
- Department of Botany and Genetics, Vilnius University, M.K.Čiurlionis Str. 21/27, LT-03101 Vilnius, Lithuania; E-Mails: (G.S.); (V.D.); (J.R.L.); (J.M.); (V.M.)
| | - Vaidotas Morkūnas
- Department of Botany and Genetics, Vilnius University, M.K.Čiurlionis Str. 21/27, LT-03101 Vilnius, Lithuania; E-Mails: (G.S.); (V.D.); (J.R.L.); (J.M.); (V.M.)
| | - Rita Kazernavičiūtė
- Department of Food Technology, Kaunas University of Technology, Radvilėnų pl. 19, LT-50254 Kaunas, Lithuania; E-Mails: (R.K.); (A.P.); (P.R.V.)
| | - Audrius Pukalskas
- Department of Food Technology, Kaunas University of Technology, Radvilėnų pl. 19, LT-50254 Kaunas, Lithuania; E-Mails: (R.K.); (A.P.); (P.R.V.)
| | - Petras Rimantas Venskutonis
- Department of Food Technology, Kaunas University of Technology, Radvilėnų pl. 19, LT-50254 Kaunas, Lithuania; E-Mails: (R.K.); (A.P.); (P.R.V.)
| |
Collapse
|