1
|
Fan W, Zhang Y, Yang X, Liu Z, Wu X, Li L, Zhao T, Li H, Liu X, Cui D, Wan Q, Li W, Yang H, Zhang H, Jia Y. BNIP3 as a potential target of esculetin for treating ovarian cancer and prognostic biomarker in ovarian cancer patients. Eur J Pharmacol 2025; 999:177698. [PMID: 40334727 DOI: 10.1016/j.ejphar.2025.177698] [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: 10/29/2024] [Revised: 03/15/2025] [Accepted: 04/30/2025] [Indexed: 05/09/2025]
Abstract
Among gynecological tumors, ovarian cancer has the highest mortality rate and worst prognosis. Therefore, it is crucial to identify and develop novel methods and targets for treating ovarian cancer. Previous studies have shown that esculetin exerts antitumor effects in a variety of cancers, however, its anti-ovarian cancer effects and mechanisms of action remain unclear. In the present study, we investigated the anti-ovarian cancer effects and mechanisms of esculetin in A2780 and OVCAR3 ovarian cancer cells and established an xenograft ovarian cancer mouse model. Esculetin significantly inhibited ovarian cancer cell proliferation, blocked cell cycle progression, and promoted apoptosis and DNA damage in a concentration-dependent manner. Furthermore, esculetin inhibited tumor growth in an xenograft ovarian cancer mouse model. Moreover, RNA-seq showed that 2114 genes were significantly altered in A2780 cells after esculetin treatment. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis indicated that these differentially expressed genes were mainly enriched in the calcium, HIF-1, and Rap1 signaling pathways. Interestingly, BNIP3 expression was notably upregulated in ovarian cancer cells after esculetin treatment. Finally, we found that low BNIP3 expression was correlated with poor prognosis in patients with ovarian cancer. These results prove that esculetin may be a valuable anti-ovarian cancer drug, and that BNIP3 is a potential treatment target for esculetin and a potential prognostic biomarker for ovarian cancer.
Collapse
Affiliation(s)
- Wen Fan
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China
| | - Yongjun Zhang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China
| | - Xiangqun Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China
| | - Zhenyan Liu
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China
| | - Xingyan Wu
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China
| | - Lvzhou Li
- Department of Obstetrics and Gynecology, Dehong Affiliated Hospital of Kunming Medical University, Dehong People's Hospital of Yunnan Province, Dehong, 678400, China
| | - Taoyu Zhao
- Department of Obstetrics and Gynecology, Dehong Affiliated Hospital of Kunming Medical University, Dehong People's Hospital of Yunnan Province, Dehong, 678400, China
| | - Haoran Li
- Medical School, Kunming University of Science and Technology, Kunming, 650500, China
| | - Xiaolin Liu
- Department of Gastroenterology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, China
| | - Daolei Cui
- Institute of Environmental Remediation and Human Health, School of Ecology and Environment, Southwest Forestry University, Kunming, 650224, China
| | - Qianqian Wan
- Medical Affairs Department, The First Affiliated Hospital of Yunnan University of Traditional Chinese Medicine, Kunming, 650021, China
| | - Wenliang Li
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China
| | - Hongying Yang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China.
| | - Hongping Zhang
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China.
| | - Yue Jia
- Department of Gynecology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Kunming, 650118, China.
| |
Collapse
|
2
|
Kim AT, Park Y. Esculetin Inhibits Fat Accumulation Through Insulin/Insulin-like Growth Factor- and AMP-Activated Protein Kinase-Dependent Pathways in Caenorhabditis elegans. Nutrients 2025; 17:1565. [PMID: 40362874 PMCID: PMC12073564 DOI: 10.3390/nu17091565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2025] [Revised: 04/29/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
Background: Esculetin, 6,7-dihydroxycoumarin, is a bioactive compound found in various herbal plants, and is known to have health-beneficial properties including anti-obesity effects. However, there is a lack of in vivo studies to clearly determine esculetin's role in lipid metabolism. Objectives: In this study, we studied esculetin's effect on lipid accumulation using Caenorhabditis elegans and its underlying mechanisms. Methods:C. elegans were treated with esculetin (100 or 200 μM) for 48 h, and their triglyceride and protein levels were measured. Additionally, behavioral patterns such as pharyngeal pumping rate, body bending rate, body sizes, and locomotive activity were analyzed. Genetic dependencies were examined by utilizing mutant worms and testing relative gene expressions. Results:C. elegans treated with esculetin displayed significantly reduced fat accumulation compared to the controls without effects on the pharyngeal pumping rate, body bending rate, or locomotive activity. Esculetin's fat-lowering effect was dependent on DAF-2 (insulin/insulin-like growth factor-1 [IGF-1] receptor homolog), DAF-16 (Forkhead box protein O homolog), and AAK-2 (5'-adenosine monophosphate-activated protein kinase [AMPK] catalytic subunit α2) in the mutant experiments. Esculetin also significantly increased the relative expression of downstream targets of DAF-16 (hsp-16.2 and sod-3), AMPK-related genes (aak-1 and aak-2), a sirtuin gene, sir-2.1, and a lipolysis-related gene, atgl-1. Conclusions: These findings suggest that esculetin inhibited fat accumulation in C. elegans and this effect was dependent on the insulin/IGF-1 and 5'-adenosine monophosphate-activated protein kinase signaling pathways.
Collapse
Affiliation(s)
| | - Yeonhwa Park
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
| |
Collapse
|
3
|
Niu C, Zhang J, Okolo PI, Daglilar E. Plant polyphenols in gastric cancer: Nature's healing touch. Semin Oncol 2025; 52:152333. [PMID: 40073717 DOI: 10.1053/j.seminoncol.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 01/22/2025] [Accepted: 01/23/2025] [Indexed: 03/14/2025]
Abstract
Globally, gastric cancer ranks as the fifth most common cancer and is the third most common cause of malignancy-associated mortality. Although surgery is the primary treatment option for gastric cancer, adjuvant chemotherapy improves survival in patients following surgery. Proverbially, plant polyphenols have many beneficial health effects, including anticancer properties. Extensive studies have shown that plant polyphenols exhibit potential anticancer effects against gastric cancer in vitro and in vivo, as well as very few human studies. However, this topic has not yet been reviewed. The present review shows that the potential anticancer effect of plant polyphenols on gastric cancer was preliminarily attributed to their antiproliferative, antimetastatic, and antiangiogenic effects and modulations of apoptosis, autophagy, and intracellular reactive oxygen species. Moreover, conventional therapeutics combined with plant polyphenols make gastric cancer cells more sensitive to conventional therapy. We also discuss challenges and opportunities in translating plant polyphenol-based therapy to clinical applications. The content provided in this review is of interest to pharmacologists, ethnobotanists, and oncologists who are involved in phytomedicine.
Collapse
Affiliation(s)
- Chengu Niu
- Internal medicine residency program, Rochester General Hospital, Rochester, NY, USA.
| | - Jing Zhang
- Rainier Springs Behavioral Health Hospital, Vancouver, WA, USA
| | - Patrick I Okolo
- Division of Gastroenterology, Carillion Clinic, Roanoke, VA, USA
| | - Ebubekir Daglilar
- Division of Gastroenterology, Charleston Area Medical Center/CAMC Institute for Academic Medicine Program, Charleston, WV, USA
| |
Collapse
|
4
|
Cai B, Cai T, Feng Z, Zhu H. The possible anti-tumor actions and mechanisms of active metabolites from Cortex Fraxini. Front Pharmacol 2024; 15:1404172. [PMID: 39346560 PMCID: PMC11427270 DOI: 10.3389/fphar.2024.1404172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Accepted: 09/02/2024] [Indexed: 10/01/2024] Open
Abstract
Cortex Fraxini is a traditional Chinese herb that is widely available, inexpensive, and has low toxicity. Modern pharmacological studies have demonstrated that the active metabolites in Cortex Fraxini, including esculin, esculetin, and fraxetin, exert anti-tumor activities by regulating genes and proteins involved in cancer cell proliferation, apoptosis, invasion, and migration. Additionally, these metabolites play a pivotal role in the regulation of several tumor-associated signaling pathways, including the PI3K/Akt, MAPK/ERK, JAK/STAT3, and Wnt/β-catenin pathways. Due to their pro-apoptotic and anti-proliferative properties in vitro and in vivo, Cortex Fraxini and its active metabolites may be considered as potential candidates for the treatment of tumor. The aim of this review is to highlight the anti-tumor biological activities and underlying mechanisms of action of the active metabolites of Cortex Fraxini, with a view to providing a reference for their further development and application in the treatment of tumors.
Collapse
Affiliation(s)
- Bin Cai
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Ting Cai
- Department of Nephrology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Wuxi, China
| | - Zeyu Feng
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| | - Huanhuan Zhu
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
5
|
Liu M, Sheng Y, Guo F, Wu J, Huang Y, Yang X, Wang M, Zhang S, Li P. Therapeutic potential of esculetin in various cancer types (Review). Oncol Lett 2024; 28:305. [PMID: 38774454 PMCID: PMC11106741 DOI: 10.3892/ol.2024.14438] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 04/05/2024] [Indexed: 05/24/2024] Open
Abstract
Esculetin (Esc), a coumarin derivative and herbal medicinal compound used in traditional Chinese medicine, is extracted from Fraxinus chinensis. Esc has shown notable potential in the inhibition of proliferation, metastasis and cell cycle arrest in various cancer cell lines. The present review is based on research articles regarding Esc in the field of carcinoma, published between 2009 and 2023. These studies have unanimously demonstrated that Esc can effectively inhibit cancer cell proliferation through diverse mechanisms and modulate multiple signaling pathways, such as Wnt/β-catenin, PI3K/Akt, MAPK and janus kinase/signal transducer and activator of transcription-3. In addition, the safety profile of Esc has been demonstrated in credible animal experiments, which has indicated Esc as an effective compound. Furthermore, the combination therapy of Esc with commonly used chemotherapeutic drugs holds great promise. The aim of the present review was to encourage further studies and applications of Esc in cancer therapy.
Collapse
Affiliation(s)
- Mengying Liu
- Department of Basic Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Yuyan Sheng
- Teaching Department, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Fangyue Guo
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Jing Wu
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Yufei Huang
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan 450052, P.R. China
| | - Xiaoning Yang
- Department of Basic Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Mengying Wang
- Department of Basic Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Shanfeng Zhang
- Department of Basic Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Pei Li
- Department of Basic Medicine, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
6
|
Luo W, Chang G, Lin D, Xie H, Sun H, Li Z, Mo S, Wang R, Wang Y, Zheng Z. 3,3'-((3,4,5-trifluoropHenyl)methylene)bis(4-hydroxy-2H-chromen-2-one) inhibit lung cancer cell proliferation and migration. PLoS One 2024; 19:e0303186. [PMID: 38776295 PMCID: PMC11111047 DOI: 10.1371/journal.pone.0303186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 04/19/2024] [Indexed: 05/24/2024] Open
Abstract
Lung cancer is a major public health challenge and, despite therapeutic improvements, is the first leading cause of cancer worldwide. The current cure rate from advanced cancer treatment is excessively low. Therefore, it is of great importance to identify novel, potent and less toxic anticancer agents for the treatment of lung cancer. The aim of our research is to synthesize a new biscoumarin 3,3'-((3,4,5-trifluorop -phenyl)methylene)bis(4-hydroxy-2H-chromen-2-one) (C35) as an anticancer agent. C35 was simply prepared by 4-hydroxycoumarin and 3,4,5-trifluorobenzaldehyde under ethanol and its structure was analyzed by spectroscopic analyses. The anti-proliferation effect of C35 was detected using CCK-8 assay. Migration abilities were measured by Transwell assay. The expression of correlated proteins was determined by Western blot. The results showed that C35 displayed strong cytostatic effects on lung cancer cell proliferation. In addition, C35 possessed a significant inhibition of migration by reducing the expression of matrix metalloproteinases-2 (MMP-2) and MMP-9 in lung cancer cells. Furthermore, C35 treatment suppressed the phosphorylation of p38 in lung cancer cells. Moreover, in vivo experiments were carried out, in which we treated Lewis tumor-bearing C57 mice via intraperitoneal injection of C35. Results showed that C35 inhibited tumor growth in vivo. In conclusion, our study demonstrated the anticancer activity of C35 via suppression of lung cancer cell proliferation and migration, which is possibly involved with the inhibition of the p38 pathway.
Collapse
Affiliation(s)
- Wenhui Luo
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
- Guangdong Provincial Key Laboratory of Traditional Chinese Medicine Formula Granule, Guangdong Yifang Pharmaceutical Co., Ltd., Foshan, Guangdong Province, PR China
| | - Guoxin Chang
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Dingmei Lin
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Hongyi Xie
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Huilong Sun
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Zhibin Li
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Shirong Mo
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Ruixue Wang
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| | - Yan Wang
- College of Traditional Chinese Medicine, Guangdong Pharmacuetical University, Guangzhou, Guangdong Province, PR China
| | - Zhaoguang Zheng
- School of Medicine, Foshan University, Foshan, Guangdong Province, PR China
| |
Collapse
|
7
|
da Silva VJ, Baumgarten LG, Dreyer JP, Santana ER, Spinelli A, Winiarski JP, Vieira IC. Heparin-stabilized gold nanoparticles embedded in graphene for the electrochemical determination of esculetin. ANALYTICAL METHODS : ADVANCING METHODS AND APPLICATIONS 2024; 16:2256-2266. [PMID: 38517319 DOI: 10.1039/d4ay00229f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/23/2024]
Abstract
A conductive nanocomposite consisting of heparin-stabilized gold nanoparticles embedded in graphene was prepared and characterized to develop an electrochemical sensor for the determination of esculetin in tea and jam samples. The gold nanoparticles were characterized by spectroscopic and microscopic techniques. The different proportions of graphene in the nanocomposite were evaluated and characterized by electrochemical practices. The heterostructure material on the glassy carbon electrode with esculetin showed π-π stacking interactions with an adsorption-controlled process. The voltammetric profile of esculetin using the proposed nanomaterial presented oxidation and reduction peaks at +0.61 and +0.58 V vs. Ag/AgCl, respectively, facilitating the electron transfer with esculetin through the transfer of two moles of protons and two moles of electrons per mole of esculetin. Using optimized conditions and square wave voltammetry, the calibration curve was obtained with two linear ranges, from 0.1 to 20.5 μmol L-1, with a detection limit of 43.0 nmol L-1. The electrochemical sensor showed satisfactory results for repeatability and stability, although interferences were observed in the presence of high concentrations of ascorbic acid or quercetin. The sensor was successfully applied in the determination of esculetin in samples of mulberry jam, white mulberry leaf tea, and white mulberry powder tea, presenting adequate recovery ranges. This directive provides valuable insights for the development of novel electrochemical sensors using heparin-based conductive nanomaterials with improved sensitivity and sensibility.
Collapse
Affiliation(s)
- Vinicius José da Silva
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Luan Gabriel Baumgarten
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Juliana Priscila Dreyer
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Edson Roberto Santana
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Almir Spinelli
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - João Paulo Winiarski
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| | - Iolanda Cruz Vieira
- Laboratory of Biosensors - Department of Chemistry, Federal University of Santa Catarina, Campus Trindade, Florianópolis, SC, 88040-900, Brazil.
| |
Collapse
|
8
|
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: 2] [Impact Index Per Article: 2.0] [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
|
9
|
Abstract
Esculin and esculetin are 2 widely studied coumarin components of Cortex Fraxini, which is a well-known herbal medicine with a 2000-year history. In vivo and in vitro studies have demonstrated that both have a variety of pharmacological activities, including antioxidant, anti-tumor, anti-inflammatory, antibacterial, antidiabetic, immunomodulatory, anti-atherosclerotic, and so on. Their underlying mechanisms of action and biological activities include scavenging free radicals, modulating the nuclear factor erythroid 2-related factor 2 pathway, regulating the cell cycle, inhibiting tumor cell proliferation and migration, promoting mitochondrial pathway apoptosis, inhibiting the NF-κB and MAPK signaling pathways, regulating CD4+ T cells differentiation and associated cytokine release, inhibiting vascular smooth muscle cells, etc. This review aims to provide comprehensive information on pharmacological studies of esculin and esculetin, which is of noteworthy importance in exploring the therapeutic potential of both coumarin compounds.
Collapse
Affiliation(s)
- Ting Cai
- Department of Nephrology, The Affiliated Wuxi People’s Hospital of Nanjing Medical University, Wuxi People’s Hospital, Wuxi Medical Center, Wuxi, China
| | - Bin Cai
- Department of Anorectal Surgery, Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
10
|
Wróblewska-Łuczka P, Góralczyk A, Łuszczki JJ. Synergy, Additivity and Antagonism between Esculetin and Six Commonly Used Chemotherapeutics in Various Malignant Melanoma Cell Lines-An Isobolographic Analysis. Molecules 2023; 28:molecules28093889. [PMID: 37175299 PMCID: PMC10180450 DOI: 10.3390/molecules28093889] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/30/2023] [Accepted: 05/03/2023] [Indexed: 05/15/2023] Open
Abstract
(1) Malignant melanomas are dangerous skin cancers, and the treatment of melanomas with various cytostatic drugs often causes side effects and after their prolonged use resistance to these drugs appears. The aim of this study was to evaluate the anticancer effects of esculetin (a simple coumarin) and to assess pharmacodynamic interactions between esculetin and six commonly used cytostatic drugs (cisplatin, epirubicin, docetaxel, paclitaxel, mitoxantrone and vemurafenib) using an isobolographic analysis. (2) The experiments were carried out on four human malignant melanoma cell lines (FM55P, A375, FM55M2 and SK-MEL28). The effects of esculetin on viability, cell proliferation and cytotoxicity were verified in the range of concentrations of 2-200 μM. (3) Esculetin inhibited, in a dose-dependent manner, malignant melanoma cell viability and proliferation. The IC50 for esculetin ranged from 18.20 ± 2.93 to 120.64 ± 30.39 μM depending on the melanoma cell lines used. The combinations of esculetin with epirubicin and vemurafenib showed antagonistic interactions, the combinations of esculetin with cisplatin, docetaxel and paclitaxel showed additive interactions. For the combinations of esculetin with mitoxantrone, the isobolographic analysis displayed synergy. (4) In the treatment of malignant melanoma, esculetin should not be combined with epirubicin or vemurafenib, due to the reduction of their anticancer effects, while the synergistic interactions (esculetin + mitoxantrone) deserve a preclinical recommendation as a beneficial combination during anticancer therapy.
Collapse
Affiliation(s)
- Paula Wróblewska-Łuczka
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Agnieszka Góralczyk
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| | - Jarogniew J Łuszczki
- Department of Occupational Medicine, Medical University of Lublin, ul. Jaczewskiego 8b, 20-090 Lublin, Poland
| |
Collapse
|
11
|
Zhao M, Yang Y, Nian Q, Shen C, Xiao X, Liao W, Zheng Q, Zhang G, Chen N, Gong D, Tang J, Wen Y, Zeng J. Phytochemicals and mitochondria: Therapeutic allies against gastric cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 110:154608. [PMID: 36586205 DOI: 10.1016/j.phymed.2022.154608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 12/09/2022] [Accepted: 12/16/2022] [Indexed: 06/17/2023]
Abstract
BACKGROUND Mitochondria are the energy factories of cells with the ability to modulate the cell cycle, cellular differentiation, signal transduction, growth, and apoptosis. Existing drugs targeting mitochondria in cancer treatment have disadvantages of drug resistance and side effects. Phytochemicals, which are widely found in plants, are bioactive compounds that could facilitate the development of new drugs for gastric cancer. Studies have shown that some phytochemicals can suppress the development of gastric cancer. METHODS We searched for data from PubMed, China National Knowledge Infrastructure, Web of Science, and Embase databases from initial establishment to December 2021 to review the mechanism by which phytochemicals suppress gastric cancer cell growth by modulating mitochondrial function. Phytochemicals were classified and summarized by their mechanisms of action. RESULTS Phytochemicals can interfere with mitochondria through several mechanisms to reach the goal of promoting apoptosis in gastric cancer cells. Some phytochemicals, e.g., daidzein and tetrandrine promoted cytochrome c spillover into the cytoplasm by modulating the members of the B-cell lymphoma-2 protein family and induced apoptotic body activity by activating the caspase protein family. Phytochemicals (e.g., celastrol and shikonin) could promote the accumulation of reactive oxygen species and reduce the mitochondrial membrane potential. Several phytochemicals (e.g., berberine and oleanolic acid) activated mitochondrial apoptotic submission via the phosphatidylinositol-3-kinase/Akt signaling pathway, thereby triggering apoptosis in gastric cancer cells. Several well-known phytochemicals that target mitochondria, including berberine, ginsenoside, and baicalein, showed the advantages of multiple targets, high efficacy, and fewer side effects. CONCLUSIONS Phytochemicals could target the mitochondria in the treatment of gastric cancer, providing potential directions and evidence for clinical translation. Drug discovery focused on phytochemicals has great potential to break barriers in cancer treatment.
Collapse
Affiliation(s)
- Maoyuan Zhao
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Yi Yang
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Qing Nian
- Department of Blood Transfusion, Sichuan Academy of Medical Science & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, PR China
| | - Caifei Shen
- Department of Endoscopy center, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Xiaolin Xiao
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Wenhao Liao
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Qiao Zheng
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Gang Zhang
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Nianzhi Chen
- Department of Oncology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China
| | - Daoyin Gong
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China
| | - Jianyuan Tang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China.
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, PR China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China; Department of Geriatrics, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, PR China.
| |
Collapse
|
12
|
Choi J, Yoo MJ, Park SY, Seol JW. Antitumor Effects of Esculetin, a Natural Coumarin Derivative, against Canine Mammary Gland Tumor Cells by Inducing Cell Cycle Arrest and Apoptosis. Vet Sci 2023; 10:vetsci10020084. [PMID: 36851388 PMCID: PMC9961495 DOI: 10.3390/vetsci10020084] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/13/2023] [Accepted: 01/19/2023] [Indexed: 01/24/2023] Open
Abstract
Mammary gland tumors are the most common neoplasms in female dogs, of which 50% are malignant. Esculetin, a coumarin derivative, reportedly induces death in different types of cancer cells. In this study, we explore the anticancer effects of esculetin against CMT-U27 and CF41.mg canine mammary gland tumor cells. Esculetin significantly inhibited the viability and migration of both CMT-U27 and CF41.mg cells in a dose- and time-dependent manner. Flow cytometric analysis and terminal deoxynucleotidyl transferase dUTP nick-end labeling assay revealed increased numbers of annexin-V-positive cells and DNA fragmentation. Furthermore, a cell cycle analysis demonstrated that esculetin blocked the cell progression at the G0/G1 phase and the S phase in CMT-U27 and CF41.mg cells. These results were supported by a Western blot analysis, which revealed upregulated protein expression of cleaved caspase-3, a marker of apoptosis, and downregulated cyclin-dependent kinase 4 and cyclin D1 protein, the cell cycle regulators. In conclusion, this novel study proves that esculetin exerts in vitro antitumor effects by inducing apoptosis and cell cycle arrest in canine mammary gland tumors.
Collapse
Affiliation(s)
- Jawun Choi
- Correspondence: (J.C.); (J.-W.S.); Tel.: +82-63-850-0964 (J.-W.S.)
| | | | | | - Jae-Won Seol
- Correspondence: (J.C.); (J.-W.S.); Tel.: +82-63-850-0964 (J.-W.S.)
| |
Collapse
|
13
|
Xiu Z, Li Y, Fang J, Han J, Li S, Li Y, Yang X, Song G, Li Y, Jin N, Zhu Y, Zhu G, Sun L, Li X. Inhibitory Effects of Esculetin on Liver Cancer Through Triggering NCOA4 Pathway-Mediation Ferritinophagy in vivo and in vitro. J Hepatocell Carcinoma 2023; 10:611-629. [PMID: 37069958 PMCID: PMC10105581 DOI: 10.2147/jhc.s395617] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 01/18/2023] [Indexed: 04/19/2023] Open
Abstract
Objective To explore the effects of Esculetin on liver cancer and explore potential mechanisms of Esculetin-inducing cells death. Methods Esculetin's effects on the proliferation, migration and apoptosis of HUH7 and HCCLM3 cells were detected by using CCK8, crystal violet staining, wound healing, TranswellTM and Annexin V-FITC/PI. Flow cytometry, fluorescence staining, Western blot, T-AOC, DPPH radical scavenging assay, hydroxyl radical's inhibitory capability and GSH test were used to examine the esculetin's effects on the ROS level, the oxidation-related substances and proteins' expression in hepatoma cells. In vivo experiment was performed by xenograft model. Ferrostatin-1 was used to determine the death way of hepatoma cells induced by esculetin. Live cell probe, Western blot, Fe2+ content, MDA, HE staining, Prussian blue staining and immunohistochemistry were used to examine the ferritinophagy-related phenomenon induced by esculetin in hepatoma cells. The relationship between esculetin and NCOA4-mediated ferritinophagy was confirmed through gene silence and overexpression, immunofluorescence staining and Western blot. Results Esculetin suppressed the proliferation, migration and apoptosis of HUH7 and HCCLM3 cells significantly, influenced the oxidative stress level, altered the autophagy and iron metabolism levels in cells, and produced a ferritinophagy-related phenomena. Esculetin increased the levels of cellular lipid peroxidation and reactive oxygen species. In vivo, esculetin could decrease tumour volume, promote LC3 and NCOA4 expressions, suppresse hydroxyl radical's inhibiting capacity and GSH, increase Fe2+ and MDA levels, decrease antioxidant proteins expression in tumour tissue. In addition, Esculetin could also increase the iron deposition of tumour tissues, promote ferritinophagy, and induce tumours' ferroptosis. Conclusion Esculetin has an inhibitory effect on liver cancer in vivo and in vitro through triggering NCOA4 pathway-mediation ferritinophagy.
Collapse
Affiliation(s)
- Zhiru Xiu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yiquan Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Jinbo Fang
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Jicheng Han
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Shanzhi Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Yaru Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
- Medical College, Yanbian University, Yanji, People’s Republic of China
| | - Xia Yang
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Gaojie Song
- Medical College, Jiujiang· University, Jiujiang, People’s Republic of China
| | - Yue Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Ningyi Jin
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- Jiangsu Co-Innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, People’s Republic of China
| | - Yilong Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Guangze Zhu
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
| | - Lili Sun
- Department of Head and Neck Surgery, Tumor Hospital of Jilin Province, Changchun, People’s Republic of China
- Correspondence: Lili Sun; Xiao Li, Boshuo Road, 1035, Jingyue Economic & Technological Development Zone, Changchun, Jilin, 130122, People’s Republic of China, Tel +86-431-86985923, Fax +86-431-87985861, Email ;
| | - Xiao Li
- Academician Workstation of Jilin Province, Changchun University of Chinese Medicine, Changchun, People’s Republic of China
- Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, People’s Republic of China
- College of Life Sciences, Shandong Normal University, Jinan, People’s Republic of China
| |
Collapse
|
14
|
The immunoregulatory effects of natural products on psoriasis via its action on Th17 cells versus regulatory T cells balance. Int Immunopharmacol 2022; 110:109032. [PMID: 35810491 DOI: 10.1016/j.intimp.2022.109032] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 06/27/2022] [Accepted: 07/03/2022] [Indexed: 01/08/2023]
Abstract
Psoriasis is an incurable, chronic inflammatory disease, which brings a substantial burden on individuals and society. Currently, the treatment of psoriasis has entered the era of biologics, but its highly targeting of inflammatory mediators may enable the immune response to circumvent the blockade, leading to disease recurrence, or other clinical and immunological characteristics. Therefore, the discovery of new therapies that have the ability of multidirectional regulation on immunity and maintain the dynamic balance of immunity in psoriasis, may be the key to the treatment of the disease. Natural products extracted from herbal medicines have synergistic effects to alleviate psoriasis and its comorbidities because of their structural diversity and multiple active mechanisms. To date, the characteristics of natural products regulating T helper 17 (Th17) cells/regulatory T (Treg) cells balance in the treatment of psoriasis have attracted more and more attention from basic and clinical studies. In this review, we systematically introduced the natural products regulating the balance of Th17/Treg and their specific mechanism of action, finding Datura metel L, Grape seed proanthocyanidin extract (GSPE), Thymol, Kaempferol, Aloperine, Abietic acid (AA), Isogarcinol, Luteolin reduced the frequency and function of Th17 cells and simultaneously increased that of Treg cells. It is expected that our work can provide a reference for clinicians in drug use.
Collapse
|
15
|
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: 1.0] [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
|
16
|
Sair AT, Liu RH. Molecular regulation of phenolic compounds on IGF-1 signaling cascade in breast cancer. Food Funct 2022; 13:3170-3184. [PMID: 35253808 DOI: 10.1039/d1fo03283f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Breast cancer is a highly aggressive and heterogeneous disease with complex features that remains a major health problem and undermines the span and quality of life of women worldwide. Primary literature has shown the role of phenolic compounds in controlling the onset of breast cancer. The mechanism of action of phenolic compounds can be explained by their interaction with signal transduction pathways that regulate cell proliferation and induction of apoptosis. One of the targets of phenolic compounds is the insulin like growth factor 1 (IGF-1) signaling cascade, which plays a significant role in the growth and development of mammary tissues by leading proliferative and anti-apoptotic events. Increasing research evidence points to the function of the IGF-1 cascade system in the commencement, progression, and metastasis of breast tissue malignancy. In this review, we mainly discuss the function of the IGF-1 system, and the role of phenolic compounds in regulating the IGF-1 signaling cascade and curbing breast malignancies.
Collapse
Affiliation(s)
- Ali Tahir Sair
- Department of Food Science, Cornell University, 245 Stocking Hall, Ithaca, New York 14853, USA.
| | - Rui Hai Liu
- Department of Food Science, Cornell University, 245 Stocking Hall, Ithaca, New York 14853, USA.
| |
Collapse
|
17
|
Chen C, Yu LT, Cheng BR, Xu JL, Cai Y, Jin JL, Feng RL, Xie L, Qu XY, Li D, Liu J, Li Y, Cui XY, Lu JJ, Zhou K, Lin Q, Wan J. Promising Therapeutic Candidate for Myocardial Ischemia/Reperfusion Injury: What Are the Possible Mechanisms and Roles of Phytochemicals? Front Cardiovasc Med 2022; 8:792592. [PMID: 35252368 PMCID: PMC8893235 DOI: 10.3389/fcvm.2021.792592] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Percutaneous coronary intervention (PCI) is one of the most effective reperfusion strategies for acute myocardial infarction (AMI) despite myocardial ischemia/reperfusion (I/R) injury, causing one of the causes of most cardiomyocyte injuries and deaths. The pathological processes of myocardial I/R injury include apoptosis, autophagy, and irreversible cell death caused by calcium overload, oxidative stress, and inflammation. Eventually, myocardial I/R injury causes a spike of further cardiomyocyte injury that contributes to final infarct size (IS) and bound with hospitalization of heart failure as well as all-cause mortality within the following 12 months. Therefore, the addition of adjuvant intervention to improve myocardial salvage and cardiac function calls for further investigation. Phytochemicals are non-nutritive bioactive secondary compounds abundantly found in Chinese herbal medicine. Great effort has been put into phytochemicals because they are often in line with the expectations to improve myocardial I/R injury without compromising the clinical efficacy or to even produce synergy. We summarized the previous efforts, briefly outlined the mechanism of myocardial I/R injury, and focused on exploring the cardioprotective effects and potential mechanisms of all phytochemical types that have been investigated under myocardial I/R injury. Phytochemicals deserve to be utilized as promising therapeutic candidates for further development and research on combating myocardial I/R injury. Nevertheless, more studies are needed to provide a better understanding of the mechanism of myocardial I/R injury treatment using phytochemicals and possible side effects associated with this approach.
Collapse
Affiliation(s)
- Cong Chen
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Lin-Tong Yu
- Department of Cardiology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Bai-Ru Cheng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jiang-Lin Xu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Yun Cai
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Jia-Lin Jin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Ru-Li Feng
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Long Xie
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Xin-Yan Qu
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
| | - Dong Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jing Liu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yan Li
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Xiao-Yun Cui
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Jin-Jin Lu
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Kun Zhou
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Qian Lin
- Department of Cardiology, Dongzhimen Hospital Affiliated to Beijing University of Chinese Medicine, Beijing, China
- *Correspondence: Qian Lin
| | - Jie Wan
- Department of Cardiology, Dongfang Hospital Beijing University of Chinese Medicine, Beijing, China
- Jie Wan
| |
Collapse
|
18
|
Zhang H, Zhang L, Lu M. Inhibition of integrin subunit alpha 11 restrains gastric cancer progression through phosphatidylinositol 3-kinase/Akt pathway. Bioengineered 2021; 12:11909-11921. [PMID: 34802381 PMCID: PMC8810121 DOI: 10.1080/21655979.2021.2006551] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 11/08/2021] [Accepted: 11/09/2021] [Indexed: 01/02/2023] Open
Abstract
Gastric cancer (GC) is among the most frequent malignancies originating from the digestive system worldwide, while the role and specific mechanism of integrin-subunit alpha 11 (ITGA11) in GC remain unclear. This study probes the expression characteristics and function of ITGA11 in GC. Firstly, the ITGA11 profile in GC tissues and paracancerous non-tumor tissues was assessed by quantitative reverse transcription-polymerase chain reaction (qRT-PCR) and Western blot (WB), and the association between ITGA11 and GC patients' clinicopathological indicators was evaluated. ITGA11 knockdown models were set up in GC cell lines MKN45 and AGS. Cell proliferation was determined by the cell counting kit-8 (CCK-8) assay and colony formation assay. WB was utilized to gauge the expression of apoptosis-related proteins (Bax, Bcl2, Bad, and C-Caspase3) and the PI3K/AKT pathway. We discovered that the ITGA11 expression was boosted in GC tissues and was related to the unfavorable prognosis of GC patients. Additionally, ITGA11 knockdown abated GC cell proliferation, invasion and migration, and enhanced cell apoptosis. In animal experiments, the tumorigenesis of GC cells knocking down ITGA11 was reduced. Mechanically, knocking down ITGA11 notably inactivated the PI3K/AKT axis. The tumor-suppressive effect mediated by ITGA11 knockdown was attenuated after activating the PI3K/AKT pathway with insulin-like growth factor 1 (IGF-1). Overall, this study substantiated that the ITGA11 expression was heightened in GC tissues, which affected GC progression by modulating the PI3K/AKT pathway.
Collapse
Affiliation(s)
- Haijun Zhang
- Second Department of General Surgery, The First Hospital of Qiqihar, Qiqihar, P. R. China
| | - Lin Zhang
- Pharmacy Department of the Second Affiliated Hospital of Qiqihar Medical College, Qiqihar, P. R. China
| | - Ming Lu
- First Department of Surgery, Gannan People’s Hospital, Qiqihar, P. R. China
| |
Collapse
|
19
|
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: 48] [Impact Index Per Article: 12.0] [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
|
20
|
Banikazemi Z, Mirazimi SM, Dashti F, Mazandaranian MR, Akbari M, Morshedi K, Aslanbeigi F, Rashidian A, Chamanara M, Hamblin MR, Taghizadeh M, Mirzaei H. Coumarins and Gastrointestinal Cancer: A New Therapeutic Option? Front Oncol 2021; 11:752784. [PMID: 34707995 PMCID: PMC8542999 DOI: 10.3389/fonc.2021.752784] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 09/20/2021] [Indexed: 12/24/2022] Open
Abstract
Cancers of the gastrointestinal (GI) tract are often life-threatening malignancies, which can be a severe burden to the health care system. Globally, the mortality rate from gastrointestinal tumors has been increasing due to the lack of adequate diagnostic, prognostic, and therapeutic measures to combat these tumors. Coumarin is a natural product with remarkable antitumor activity, and it is widely found in various natural plant sources. Researchers have explored coumarin and its related derivatives to investigate their antitumor activity, and the potential molecular mechanisms involved. These mechanisms include hormone antagonists, alkylating agents, inhibitors of angiogenesis, inhibitors of topoisomerase, inducers of apoptosis, agents with antimitotic activity, telomerase inhibitors, inhibitors of human carbonic anhydrase, as well as other potential mechanisms. Consequently, drug design and discovery scientists and medicinal chemists have collaborated to identify new coumarin-related agents in order to produce more effective antitumor drugs against GI cancers. Herein, we summarize the therapeutic effects of coumarin and its derivatives against GI cancer.
Collapse
Affiliation(s)
- Zarrin Banikazemi
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Seyed Mohammad Mirazimi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.,School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Dashti
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.,School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Reza Mazandaranian
- Department of Community Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences, Tehran, Iran
| | - Maryam Akbari
- Department of Surgery, Kashan University of Medical Sciences, Kashan, Iran
| | - Korosh Morshedi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.,School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Fatemeh Aslanbeigi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran.,School of Medicine, Kashan University of Medical Sciences, Kashan, Iran
| | - Amir Rashidian
- Department of Pharmacology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran
| | - Mohsen Chamanara
- Department of Pharmacology, School of Medicine, Aja University of Medical Sciences, Tehran, Iran.,Toxicology Research Center, Aja University of Medical Sciences, Tehran, Iran
| | - Michael R Hamblin
- Laser Research Centre, Faculty of Health Science, University of Johannesburg, Doornfontein, South Africa
| | - Mohsen Taghizadeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
21
|
Jiang R, Su G, Chen X, Chen S, Li Q, Xie B, Zhao Y. Esculetin inhibits endometrial cancer proliferation and promotes apoptosis via hnRNPA1 to downregulate BCLXL and XIAP. Cancer Lett 2021; 521:308-321. [PMID: 34480971 DOI: 10.1016/j.canlet.2021.08.039] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 07/10/2021] [Accepted: 08/30/2021] [Indexed: 11/30/2022]
Abstract
Endometrial cancer represents one of the most common gynecological tumors in the world. Advanced and relapsed patients rely on drug therapy. Therefore, it is extremely important to seek more effective targeted drugs. This study found that esculetin has an anti-tumor effect on endometrial cancer through cellular proliferation and apoptosis. At the same time, its anti-tumor effect has also been verified in human endometrial cancer xenograft models in nude mice. Western blot results showed that BCLXL, XIAP, and pAKT protein expression level were down-regulated. A pulldown experiment and LC-MS/MS analysis technology revealed that esculetin targets the hnRNPA1 protein. Cellular proliferation experiments following si-hnRNPA1 transfection verified the tumor-promoting effect of hnRNPA1 in endometrial cancer cells. Nuclear and cytoplasmic separation experiment demonstrated esculetin affecting the export of the hnRNPA1/mRNA complex from the nucleus into the cytoplasm. Thus, esculetin targets hnRNPA1, thereby downregulates BCLXL and XIAP mRNA transcription and translation, resulting in apoptosis and an arrest in proliferation.
Collapse
Affiliation(s)
- Ruqi Jiang
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Guifeng Su
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Xi Chen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Shuo Chen
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Qianhui Li
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Bumin Xie
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| | - Yang Zhao
- Department of Obstetrics and Gynecology, Department of Gynecologic Oncology Research Office, Key Laboratory for Major Obstetric Diseases of Guangdong Province, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510150, China.
| |
Collapse
|
22
|
Niu Z, Zhang W, Shi J, Li X, Wu H. Effect of silencing C-erbB-2 on esophageal carcinoma cell biological behaviors by inhibiting IGF-1 pathway activation. J Cardiothorac Surg 2021; 16:194. [PMID: 34233689 PMCID: PMC8265138 DOI: 10.1186/s13019-021-01540-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 05/13/2021] [Indexed: 01/10/2023] Open
Abstract
OBJECTIVE C-erbB-2 has been confirmed to be an oncogene that participates in cell growth, differentiation and division of tumors. We are wondered if its silenced expression can exert an anti-tumor effect. Therefore, this study is conducted to investigate the mechanism of C-erbB-2 silencing and IGF-1 pathway on esophageal carcinoma (EC) cell biological behaviors. METHODS The objects of study were 84 EC patients from Heping Hospital Affiliated to Changzhi Medical College, with the collection of EC tissue and adjacent normal tissue (> 5 cm away from cancer tissue). C-erbB-2 protein expression in EC tissues was detected by immunohistochemistry. Human EC cell line Eca-109 was purchased from Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences. Based on different transfection protocols, EC cells with logarithmic growth phase of 3-5 passages were divided into blank control group, oe-C-erbB-2 NC group, siRNA C-erbB-2 NC group, oe-C-erbB-2 group, siRNA C-erbB-2 group, OSI-906 group, Rg5 group, Rg5 + siRNA C-erbB-2 NC group and Rg5 + siRNA C-erbB-2 group. Cell proliferation was detected by MTT assay; cell cycle distribution and apoptosis by flow cytometry; C-erbB-2, IGF-1, IGF-1R and Akt mRNA and protein expressions by qRT-PCR and western blot; and cell invasion and migration by Transwell assay and scratch test. Tumor growth was observed in male BALB/c nude mice (Shanghai Experimental Animal Center) based on Eca109 cell implantation, raising, and measurement. RESULTS C-erbB-2, IGF-1, IGF-1R and Akt expression were higher in EC tissues than those in adjacent tissues (all P < 0.05). Compared with blank control group, both si-C-erbB-2 and OSI-906 groups had decreased IGF-1, IGF-1R and Akt mRNA and protein expressions, decreased cell proliferation, migration and invasion, prolonged G0/G1 phase, shortened S phase, increased cell apoptosis, and inhibited tumor growth (all P < 0.05); while opposite trends were detected in C-erbB-2 vector and Rg5 groups (all P < 0.05), without statistical differences in siRNA C-erbB-2 + Rg5 group (all P > 0.05). CONCLUSION Silencing C-erbB-2 expression may inhibit EC cell proliferation, promote cell apoptosis and block cell cycle progression by inhibiting IGF-1 pathway activation. The beneficial effect of silencing C-erbB-2 expression can be reversed by promoting the activation of IGF-1 pathway. Findings in our study may provide potential reference for understanding the molecular mechanism of EC and supply possible axis for preventing the development of EC from the perspective of molecular biology.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Apoptosis/genetics
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Carcinoma, Squamous Cell/pathology
- Cell Line, Tumor
- Cell Movement
- Cell Proliferation
- Esophageal Neoplasms/genetics
- Esophageal Neoplasms/metabolism
- Esophageal Neoplasms/pathology
- Female
- Flow Cytometry
- Gene Expression Regulation, Neoplastic/physiology
- Gene Silencing/physiology
- Humans
- Immunohistochemistry
- Insulin-Like Growth Factor I/metabolism
- Male
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Neoplasm Transplantation
- RNA, Messenger/genetics
- RNA, Small Interfering/genetics
- Real-Time Polymerase Chain Reaction
- Receptor, ErbB-2/genetics
- Receptor, IGF Type 1
- Transfection
- Mice
Collapse
Affiliation(s)
- Zhigao Niu
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China.
| | - Wenping Zhang
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| | - Jialun Shi
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| | - Xiangdong Li
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| | - Hanlei Wu
- Cardiothoracic surgery department, Heping Hospital Affiliated to Changzhi Medical College, No. 110 Yan'an South Road, Luzhou District, Changzhi, 046000, Shanxi, China
| |
Collapse
|
23
|
Svitina H, Hamman JH, Gouws C. Molecular mechanisms and associated cell signalling pathways underlying the anticancer properties of phytochemical compounds from Aloe species (Review). Exp Ther Med 2021; 22:852. [PMID: 34178125 PMCID: PMC8220653 DOI: 10.3892/etm.2021.10284] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/07/2021] [Indexed: 12/14/2022] Open
Abstract
Naturally occurring components from various species of Aloe have been used as traditional folk medicine since the ancient times. Over the last few decades, the therapeutic effects of extracts and phytochemical compounds obtained from Aloe vera have been proven in preclinical and clinical studies. Recently, compounds from other Aloe species apart from Aloe vera have been investigated for the treatment of different diseases, with a particular focus on cancer. In the present review, the effects of phytochemical compounds obtained from different Aloe species are discussed, with a specific focus on the effects on cell signalling in cancer and normal cells, and their selectivity and efficacy. This information will be useful for the application of Aloe-derived compounds as therapeutic agents, either alone or in combination with other standard drugs for cancer treatment.
Collapse
Affiliation(s)
- Hanna Svitina
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom, North West 2520, South Africa.,Department of Functional Genomics, Institute of Molecular Biology and Genetics of NASU, Kyiv 03143, Ukraine
| | - Josias H Hamman
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom, North West 2520, South Africa
| | - Chrisna Gouws
- Centre of Excellence for Pharmaceutical Sciences (Pharmacen™), North-West University, Potchefstroom, North West 2520, South Africa
| |
Collapse
|
24
|
de Araújo RSA, da Silva-Junior EF, de Aquino TM, Scotti MT, Ishiki HM, Scotti L, Mendonça-Junior FJB. Computer-Aided Drug Design Applied to Secondary Metabolites as Anticancer Agents. Curr Top Med Chem 2021; 20:1677-1703. [PMID: 32515312 DOI: 10.2174/1568026620666200607191838] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 11/06/2019] [Accepted: 01/05/2020] [Indexed: 12/11/2022]
Abstract
Computer-Aided Drug Design (CADD) techniques have garnered a great deal of attention in academia and industry because of their great versatility, low costs, possibilities of cost reduction in in vitro screening and in the development of synthetic steps; these techniques are compared with highthroughput screening, in particular for candidate drugs. The secondary metabolism of plants and other organisms provide substantial amounts of new chemical structures, many of which have numerous biological and pharmacological properties for virtually every existing disease, including cancer. In oncology, compounds such as vimblastine, vincristine, taxol, podophyllotoxin, captothecin and cytarabine are examples of how important natural products enhance the cancer-fighting therapeutic arsenal. In this context, this review presents an update of Ligand-Based Drug Design and Structure-Based Drug Design techniques applied to flavonoids, alkaloids and coumarins in the search of new compounds or fragments that can be used in oncology. A systematical search using various databases was performed. The search was limited to articles published in the last 10 years. The great diversity of chemical structures (coumarin, flavonoids and alkaloids) with cancer properties, associated with infinite synthetic possibilities for obtaining analogous compounds, creates a huge chemical environment with potential to be explored, and creates a major difficulty, for screening studies to select compounds with more promising activity for a selected target. CADD techniques appear to be the least expensive and most efficient alternatives to perform virtual screening studies, aiming to selected compounds with better activity profiles and better "drugability".
Collapse
Affiliation(s)
| | | | - Thiago Mendonça de Aquino
- Laboratory of Medicinal Chemistry, Nursing and Pharmacy School, Federal University of Alagoas, Maceio-AL, Brazil
| | - Marcus Tullius Scotti
- Laboratory of Medicinal Chemistry, Nursing and Pharmacy School, Federal University of Alagoas, Maceio-AL, Brazil
| | - Hamilton M Ishiki
- University of Western Sao Paulo (Unoeste), Presidente Prudente- SP, Brazil
| | - Luciana Scotti
- Postgraduate Program in Natural and Synthetic Bioactive Products, Federal University of Paraiba, Joao Pessoa-PB, Brazil
| | | |
Collapse
|
25
|
Ding F, Yang S. Epigallocatechin-3-gallate inhibits proliferation and triggers apoptosis in colon cancer via the hedgehog/phosphoinositide 3-kinase pathways. Can J Physiol Pharmacol 2021; 99:910-920. [PMID: 33617370 DOI: 10.1139/cjpp-2020-0588] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The present study evaluated whether epigallocatechin-3-gallate (EGCG) effectively attenuates tumor growth in colon cancer cells and in the xenografts of nude mice and investigated the underlying mechanisms by focusing on the sonic hedgehog (Shh) and phosphoinositide 3-kinase (PI3K) pathways. Three kinds of colon cancer cells and BALB/c nude mice were used to evaluate the antiproliferative effect of EGCG. The apoptosis, migration, and invasion of colon cancer cells were analyzed to explore the toxicity effect of EGCG on colon cancer cells. Western blotting was used to demonstrate the expression levels of related proteins. The results showed that EGCG exhibited an antiproliferative effect against colon cancer cells in a dose-dependent manner with low toxicity against normal colon epithelial cells. Administration of EGCG caused significant apoptosis and inhibited the migration and invasion of colon cancer cells. The toxic effect of EGCG on colon cancer cells was accompanied by downregulation of the Shh and PI3K/Akt pathways. In addition, EGCG reduced tumor volume and weight without affecting the body weight of nude mice and inhibited the activation of the Shh and PI3K/AKT pathways in tumor tissue. Further study showed that purmorphamine (smoothened (Smo) agonist) or insulin like growth factor-1 (IGF-1, PI3K agonist) partly abolished the effect of EGCG on cell proliferation, migration, and apoptosis. Cyclopamine (Smo inhibitor) and LY294002 (PI3K inhibitor) showed the similar toxic effects as EGCG on colon cancer cells. In conclusion, EGCG inhibited colon tumor growth via downregulation of the Shh and PI3K pathways and may be a potential chemotherapeutic agent against colon cancer.
Collapse
Affiliation(s)
- Feng Ding
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| | - Su Yang
- Department of Urology, the First Affiliated Hospital of Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
26
|
Chen Y, He X, Feng D, Li S. Compare the Effects of Magnolol on Gastric Cancer Cells Through c-Jun N-Terminal Kinase Signaling Pathway and Gold Magnetic. JOURNAL OF NANOSCIENCE AND NANOTECHNOLOGY 2021; 21:943-948. [PMID: 33183428 DOI: 10.1166/jnn.2021.18688] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
This article explores the effects and mechanisms of magnolol on the proliferation of gastric cancer cells as well as the apoptosis. First, 0 (control group), 20, 40, and 80 /x mol/L magnolol were observed on SGC-7901 cells for 24, 48, and 72 h. We use MTT method to measure the cell viability, and apoptosis and cells were detected by flow cytometry. Cell proliferation inhibition rate, apoptosis and cell cycle experiments showed that P-value < 0.05 means the difference is statistically significant. And the results which compare the control group, the 20, 40, and 80 /x mol/L show that honokiol had lower cell viability (P < 0.01), increased apoptotic rate (P < 0.01), and cell cycle stay in the G1 phase (P < 0.01), so we found that honokiol may suppress the proliferation of SGC-7901 cells and stimulate apoptosis by regulating cyclin and apoptosis-related proteins. With the development of nanomaterials synthesis technology and application in biomedicine, gold magnetic composite nanomaterials have unique properties, so they have been widely concerned in many applications. We combine gold and magnetic nanomaterials through other nanostructures to achieve the integration of diagnosis and treatment of tumors. We have synthesized two kinds of gold magnetic nanocomposites, GNR-PPy-FA nanocomposites. With the role of chemotherapy and heat and light therapy, GNR-PPy-FA nanocomposites have high light-to-heat conversion efficiency. Cell experiments verify the effect of chemotherapy and photothermal treatment of composite nanomaterials. After incubation with gold magnetic composite nanomaterials, the cell survival rate of tumor cells decreased to about 15%. In addition, both types of gold magnetic nanocomposites have the ability to dually target cancer cells, and the modification of folic acid and cancer cell membranes makes the material more biocompatible.
Collapse
Affiliation(s)
- Yu Chen
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Xiuyun He
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Dagang Feng
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| | - Shijie Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, Sichuan Province, China
| |
Collapse
|
27
|
Wang F, Wang L, Sui G, Yang C, Guo M, Xiong X, Chen Z, Lei P. IGF-1 Alleviates Mitochondrial Apoptosis through the GSK3β/NF-κB/NLRP3 Signaling Pathway in LPS-Treated PC-12 Cells. J Mol Neurosci 2021; 71:1320-1328. [PMID: 33403593 DOI: 10.1007/s12031-020-01759-6] [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: 08/25/2020] [Accepted: 11/16/2020] [Indexed: 02/06/2023]
Abstract
Inflammation contributes to mitochondrial dysfunction and neuronal apoptosis. The aim of this study was to determine whether insulin-like growth factor-1 (IGF-1) alleviates mitochondrial apoptosis in lipopolysaccharide (LPS)-treated PC-12 cells, and to further explore the mechanism involved. Prepared PC-12 cells were treated with IGF-1, Mdivi-1 (DRP1 blocker), LY294002 (PI3K blocker), betulinic acid (NF-κB activator) or their combinations. Mitochondrial membrane potential and ATP generation were then measured to assess mitochondrial function. The rate of apoptosis was determined using flow cytometry. The expression of several apoptosis proteins (i.e. Bax, cleaved caspase-9 and cleaved caspase-3) and signaling proteins (i.e. p-GSK3β, NF-κB and NLRP3) was measured using western blotting. Compared with the control cells, the LPS-treated cells showed evidence of mitochondrial dysfunction, increased apoptosis and upregulation of apoptosis proteins, which were significantly alleviated by Mdivi-1. These findings indicate that neuronal apoptosis was activated partly through the mitochondrial pathway. IGF-1 treatment inhibited mitochondrial apoptosis in a dose-dependent manner in the LPS-treated cells. The reagent also increased the expression of p-GSK3β and decreased the expression of NF-κB and NLRP3. Both LY294002 and betulinic acid reversed the protective effect of IGF-1. In addition, LY294002 affected the expression of the three signaling proteins, while betulinic acid only affected the expression of NF-κB and NLRP3. These findings indicated a GSK3β/NF-κB/NLRP3 signaling pathway was existed and was involved in the protective mechanism of IGF-1. In conclusion, IGF-1 alleviated mitochondrial apoptosis through GSK3β/NF-κB/NLRP3 signaling pathway in LPS-treated PC-12 cells.
Collapse
Affiliation(s)
- Feng Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China.
| | - Lu Wang
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China
| | - Guanghong Sui
- Department of Child and Adolescent Psychology, Tianjin Anding Hospital, Tianjin, 300074, China
| | - Caixia Yang
- Department of Rehabilitation, Tianjin Anding Hospital, Tianjin, 300074, China
| | - Mengtian Guo
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China
| | - Xiangyang Xiong
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China
| | - Zheng Chen
- Department of Psychology, Tianjin Anding Hospital, Tianjin, 300074, China
| | - Ping Lei
- Department of Geriatrics, Tianjin Medical University General Hospital, Tianjin Geriatrics Institute, Tianjin, 300052, China
| |
Collapse
|
28
|
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
|
29
|
Association between Polyphenol Intake and Gastric Cancer Risk by Anatomic and Histologic Subtypes: MCC-Spain. Nutrients 2020; 12:nu12113281. [PMID: 33114671 PMCID: PMC7692577 DOI: 10.3390/nu12113281] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Revised: 10/20/2020] [Accepted: 10/21/2020] [Indexed: 02/08/2023] Open
Abstract
Several anticancer properties have been largely attributed to phenolics in in vivo and in vitro studies, but epidemiologic evidence is still scarce. Furthermore, some classes have not been studied in relation to gastric cancer (GC). The aim of this study was to assess the relationship between the intake of phenolic acids, stilbenes, and other phenolics and the risk of developing GC and its anatomical and histological subtypes. We used data from a multi-case-control study (MCC-Spain) obtained from different regions of Spain. We included 2700 controls and 329 GC cases. Odds ratios (ORs) were calculated using mixed effects logistic regression considering quartiles of phenolic intake. Our results showed an inverse association between stilbene and lignan intake and GC risk (ORQ4 vs. Q1 = 0.47; 95% CI: 0.32–0.69 and ORQ4 vs. Q1 = 0.53; 95% CI: 0.36–0.77, respectively). We found no overall association between total phenolic acid and other polyphenol class intake and GC risk. However, hydroxybenzaldehydes (ORQ4 vs. Q1 = 0.41; 95% CI: 0.28–0.61), hydroxycoumarins (ORQ4 vs. Q1 = 0.49; 95% CI: 0.34–0.71), and tyrosols (ORQ4 vs. Q1 = 0.56; 95% CI: 0.39–0.80) were inversely associated with GC risk. No differences were found in the analysis by anatomical or histological subtypes. In conclusion, a diet high in stilbenes, lignans, hydroxybenzaldehydes, hydroxycoumarins, and tyrosols was associated with a lower GC risk. Further prospective studies are needed to confirm our results.
Collapse
|
30
|
Wang SY, Zhao JM, Zhou CL, Zheng HD, Huang Y, Zhao M, Zhang ZY, Wu LY, Wu HG, Liu HR. Herbal cake-partitioned moxibustion inhibits colonic autophagy in Crohn’s disease via signaling involving distinct classes of phosphatidylinositol 3-kinases. World J Gastroenterol 2020; 26:5997-6014. [PMID: 33132650 PMCID: PMC7584057 DOI: 10.3748/wjg.v26.i39.5997] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 07/14/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Autophagy is an evolutionarily conserved biological process in eukaryotic cells that involves lysosomal-mediated degradation and recycling of related cellular components. Recent studies have shown that autophagy plays an important role in the pathogenesis of Crohn’s disease (CD). Herbal cake-partitioned moxibustion (HM) has been historically practiced to treat CD. However, the mechanism by which HM regulates colonic autophagy in CD remains unclear.
AIM To observe whether HM can alleviate CD by regulating colonic autophagy and to elucidate the underlying mechanism.
METHODS Rats were randomly divided into a normal control (NC) group, a CD group, an HM group, an insulin + CD (I + CD) group, an insulin + HM (I + HM) group, a rapamycin + CD (RA + CD) group, and a rapamycin + HM (RA + HM) group. 2,4,6-trinitrobenzenesulfonic acid was administered to establish a CD model. The morphology of the colonic mucosa was observed by hematoxylin-eosin staining, and the formation of autophagosomes was observed by electron microscopy. The expression of autophagy marker microtubule-associated protein 1 light chain 3 beta (LC3B) was observed by immunofluorescence staining. Insulin and rapamycin were used to inhibit and activate colonic autophagy, respectively. The mRNA expression levels of phosphatidylinositol 3-kinase class I (PI3KC1), Akt1, LC3B, sequestosome 1 (p62), and mammalian target of rapamycin (mTOR) were evaluated by RT-qPCR. The protein expression levels of interleukin 18 (IL-18), tumor necrosis factor-α (TNF-α), nuclear factor κB/p65 (NF-κB p65), LC3B, p62, coiled-coil myosin-like BCL2-interacting protein (Beclin-1), p-mTOR, PI3KC1, class III phosphatidylinositol 3-kinase (PI3KC3/Vps34), and p-Akt were evaluated by Western blot analysis.
RESULTS Compared with the NC group, the CD group showed severe damage to colon tissues and higher expression levels of IL-18 and NF-κB p65 in colon tissues (P < 0.01 for both). Compared with the CD group, the HM group showed significantly lower levels of these proteins (PIL-18 < 0.01 and Pp65 < 0.05). There were no significant differences in the expression of TNF-α protein in colon tissue among the rat groups. Typical autophagic vesicles were found in both the CD and HM groups. The expression of the autophagy proteins LC3B and Beclin-1 was upregulated (P < 0.01 for both) in the colon tissues of rats in the CD group compared with the NC group, while the protein expression of p62 and p-mTOR was downregulated (P < 0.01 for both). However, these expression trends were significantly reversed in the HM group compared with the CD group (PLC3B < 0.01, PBeclin-1 < 0.05, Pp62 < 0.05, and Pm-TOR < 0.05). Compared with those in the RA + CD group, the mRNA expression levels of PI3KC1, Akt1, mTOR, and p62 in the RA + HM group were significantly higher (PPI3KC1 < 0.01 and PAkt1, mTOR, and p62 < 0.05), while those of LC3B were significantly lower (P < 0.05). Compared with the RA + CD group, the RA + HM group exhibited significantly higher PI3KC1, p-Akt1, and p-mTOR protein levels (PPI3KC1 < 0.01, Pp-Akt1 < 0.05, and Pp-mTOR < 0.01), a higher p62 protein level (P = 0.057), and significantly lower LC3B and Vps34 protein levels (P < 0.01 for both) in colon tissue.
CONCLUSION HM can activate PI3KC1/Akt1/mTOR signaling while inhibiting the PI3KC3 (Vps34)-Beclin-1 protein complex in the colon tissues of CD rats, thereby inhibiting overactivated autophagy and thus exerting a therapeutic effect.
Collapse
Affiliation(s)
- Shi-Yuan Wang
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Ji-Meng Zhao
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Ci-Li Zhou
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Han-Dan Zheng
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Yan Huang
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Min Zhao
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Zhi-Ying Zhang
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Lu-Yi Wu
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
| | - Huan-Gan Wu
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| | - Hui-Rong Liu
- Key Laboratory of Acupuncture-Moxibustion and Immunology, Shanghai University of Traditional Chinese Medicine, Shanghai 201203, China
- Shanghai Research Institute of Acupuncture and Meridian, Shanghai 200030, China
| |
Collapse
|
31
|
Al-Warhi T, Sabt A, Elkaeed EB, Eldehna WM. Recent advancements of coumarin-based anticancer agents: An up-to-date review. Bioorg Chem 2020; 103:104163. [DOI: 10.1016/j.bioorg.2020.104163] [Citation(s) in RCA: 50] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Revised: 08/09/2020] [Accepted: 08/10/2020] [Indexed: 12/13/2022]
|
32
|
Modified Jian-pi-yang-zheng decoction inhibits gastric cancer progression via the macrophage immune checkpoint PI3Kγ. Biomed Pharmacother 2020; 129:110440. [PMID: 32768942 DOI: 10.1016/j.biopha.2020.110440] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Revised: 06/05/2020] [Accepted: 06/17/2020] [Indexed: 12/21/2022] Open
Abstract
Jian-pi-yang-zheng Decoction (JPYZ) is a traditional Chinese medicine that is used for the treatment of advanced gastric cancer, and it shows good efficacy in patients. A previous study indicated that JPYZ inhibited the progression of gastric cancer via the regulation of tumor-associated macrophages (TAMs), but the underlying molecular target of JPYZ regulation of TAMs has not been determined. The present study used modified-JPYZ (mJPYZ) to extend our investigation of gastric cancer. Our results showed that mJPYZ inhibited gastric cancer progression in vivo and in vitro. We found that mJPYZ decreased the activity of PI3-kinase γ (PI3Kγ) in TAMs, reduced the anti-inflammatory factor IL-10 and increased the expression of pro-inflammatory cytokines, such as TNF-α and IL-1β, which ultimately promoted the conversion of TAMs from M2 to M1. Our findings also indicated that mJPYZ inhibited the growth and metastasis of gastric cancer by alleviating the unfavorable differentiation of TAMs via the PI3Kγ signaling cascades. In conclusion, the present findings indicated that mJPYZ inhibited gastric cancer cell EMT via PI3Kγ-dependent TAM reprogramming, which eventually suppressed gastric cancer growth and metastasis. Our study provides an underlying mechanism of a Chinese medicine in the treatment of gastric cancer via PI3Kγ in macrophages.
Collapse
|
33
|
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: 9] [Impact Index Per Article: 1.8] [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
|
34
|
Chen ZL, Li XN, Ye CX, Chen HY, Wang ZJ. Elevated Levels of circRUNX1 in Colorectal Cancer Promote Cell Growth and Metastasis via miR-145-5p/IGF1 Signalling. Onco Targets Ther 2020; 13:4035-4048. [PMID: 32494158 PMCID: PMC7231768 DOI: 10.2147/ott.s254133] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 04/24/2020] [Indexed: 12/25/2022] Open
Abstract
Background Emerging evidence suggests that circular RNAs (circRNAs) are vital regulators in a range of cancers. “miRNA sponge” is the most reported role played by circRNAs in many tumors. The insulin-like growth factor (IGF) 1 pathway plays a key role in the development and progression of many cancers, including colorectal cancer (CRC). The aim of the study is to establish the potential clinical value and driving molecular mechanisms of circRNAs in CRC. Materials and Methods Real-time quantitative RT-PCR (qRT-PCR) was performed to measure the circRUNX1 expression in 52 tissue samples from CRC patients. We verified the tumor promotor role of circRUNX1 in cell-based in vitro and in vivo assays. Human growth factor array was used to identify circRUNX1-regulated signaling pathways. We then used a double luciferase reporter assay and RNA fluorescence in situ hybridization to identify the downstream miR-145-5p of circRUNX1. Furthermore, we performed Western blotting and biological function assays to demonstrate if the circRUNX1/miR-145-5p/IGF1 axis is responsible for the proliferation of CRC cells and promotes CRC development. Results By performing qRT-PCR from CRC tissues and paired adjacent normal mucosa tissues, we identified that circRUNX1 expression was significantly upregulated in CRC tissues and positively related with lymph node metastasis, distant metastasis and advanced tumor-node-metastasis tumor stage in patients. Functionally, circRUNX1 knockdown inhibited cell proliferation and migration and promoted apoptosis, whereas its overexpression exerted opposite effects. In vivo, circRUNX1 promoted tumor growth and metastasis. Mechanically, circRUNX1 shared miRNA response elements with IGF1. circRUNX1 competitively bound to miR-145-5p and prevented miR-145-5p from decreasing the expression of IGF1, which facilitated tumor growth. Conclusion Our studies verified that circRUNX1 functions as a tumor promotor in CRC cells by targeting the miR-145-5p/IGF1 signaling pathway and may have potential use as a prognostic indicator and therapeutic target in CRC patients.
Collapse
Affiliation(s)
- Zhi-Lei Chen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, People's Republic of China
| | - Xiang-Nan Li
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, People's Republic of China
| | - Chun-Xiang Ye
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, People's Republic of China
| | - Hong-Yu Chen
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, People's Republic of China
| | - Zhen-Jun Wang
- Department of General Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing 10020, People's Republic of China
| |
Collapse
|
35
|
Esculetin inhibits proliferation, migration, and invasion of clear cell renal cell carcinoma cells. Biomed Pharmacother 2020; 125:110031. [PMID: 32164951 DOI: 10.1016/j.biopha.2020.110031] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 02/16/2020] [Accepted: 02/17/2020] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Esculetin, the main active ingredient in the Chinese herbal medicineCortex Fraxini, has been shown to possess antitumor activity. However, the effect of esculetin on clear cell renal cell carcinoma (ccRCC) has not been investigated. METHODS MTS assays and colony formation assays were used to study the cytotoxicity of esculetin. The effects of esculetin on cell cycle and apoptosis were analyzed by flow cytometry. Western blot was used to detect cell cycle-, apoptosis-, and EMT-related proteins. Wound-healing assays and transwell assays were performed to study the effect of esculetin on cell migration and invasion in the ccRCC cell lines 786-O and SN12-PM6. RESULTS Esculetin exerted cytotoxic activities in 786-O and SN12-PM6 cells in a dose- and time-dependent manner. The compound arrested the cell cycle in G0/G1 and G2/M phase with down-regulation of Cyclin D1, CDK4, CDK6, and c-Myc expression. Esculetin also induced apoptosis and the expression of cleaved caspase 3 increased. Additionally, esculetin significantly inhibited 786-O and SN12-PM6 cell migration and invasion, the expression of E-Cadherin increased, and the expression of N-cadherin and vimentin decreased. CONCLUSION Taken together, these results suggest that esculetin inhibits proliferation, migration, and invasion of ccRCC and is a potential novel therapeutic agent for the treatment of ccRCC.
Collapse
|
36
|
Zhang G, Xu Y, Zhou HF. Esculetin Inhibits Proliferation, Invasion, and Migration of Laryngeal Cancer In Vitro and In Vivo by Inhibiting Janus Kinas (JAK)-Signal Transducer and Activator of Transcription-3 (STAT3) Activation. Med Sci Monit 2019; 25:7853-7863. [PMID: 31630150 PMCID: PMC6820344 DOI: 10.12659/msm.916246] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND Laryngeal cancer is one of the most common malignant tumors of the head and neck. Natural compounds in traditional Chinese medicine provide many valuable potential compounds for tumor chemotherapy. Esculetin, a coumarin derivative from several herbs, inhibits proliferation of many types of cancer cells, but its anticancer effect in laryngeal cancer is still not clear. MATERIAL AND METHODS We performed in vitro proliferation assay, invasion assay, and migration assay to assess the effect of esculetin against LC, and in vivo nude mouse xenograft animal model was used as well. Flow cytometry was conducted to analyze the effect of esculetin on cell cycle of LC cells, and Western blot analysis was used to assess the effect esculetin on the JAK-STAT signaling pathway. RESULTS Esculetin remarkably inhibits proliferation, migration, and invasion of LC cells, and reduces in vivo xenograft tumor growth and tumor weight in a dose-dependent manner. Our molecular mechanism study demonstrated that esculetin significantly inhibits STAT3 phosphorylation and blocks translocation of STAT3 into the nucleus, and esculetin also blocks the cell cycle in G1/S phase. CONCLUSIONS In a summary, by inhibiting the STAT3 activation, esculetin shows potential anticancer effects against the laryngeal cancer.
Collapse
Affiliation(s)
- Geng Zhang
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Yi Xu
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| | - Hui-Fang Zhou
- Department of Otorhinolaryngology, Tianjin Medical University General Hospital, Tianjin, China (mainland)
| |
Collapse
|
37
|
Choi YJ, Lee CM, Park SH, Nam MJ. Esculetin induces cell cycle arrest and apoptosis in human colon cancer LoVo cells. ENVIRONMENTAL TOXICOLOGY 2019; 34:1129-1136. [PMID: 31313495 DOI: 10.1002/tox.22815] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2019] [Revised: 06/25/2019] [Accepted: 06/26/2019] [Indexed: 06/10/2023]
Abstract
We investigated the anti-cancer effects of ESC in human colon cancer LoVo cells. Cell counting assay results showed that ESC inhibited the proliferation of LoVo cells. Cell cycle arrest results showed that cell cycle was arrested during the G0/G1 phase in the ESC-treated LoVo cells. Western blot results showed that the cell cycle inhibitory proteins p53, p27, and p21 were increased, and cyclin D1, the cell cycle progressive protein, was decreased. Sp1 is a transcription factor regulating cell proliferation, was decreased in the ESC-treated LoVo cells. Annexin V/propidium iodide staining results showed that ESC induces apoptosis in LoVo cells. Western blot results showed that Bax, cleaved caspase -3, -7, -9, and poly(ADP-ribose) polymerase, which are proapoptotic proteins, were increased and the antiapoptotic protein Bcl-2 was decreased. Taken together, ESC induced apoptosis and has an anti-cancer effect in LoVo cells.
Collapse
Affiliation(s)
- Yong J Choi
- Department of Biological Sciences, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - Chang M Lee
- Department of Biological Sciences, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, Republic of Korea
| | - Myeong J Nam
- Department of Biological Sciences, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
38
|
Xie C, Li Y, Gao J, Wang Y. Esculetin regulates the phenotype switching of airway smooth muscle cells. Phytother Res 2019; 33:3008-3015. [PMID: 31435973 DOI: 10.1002/ptr.6483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 06/30/2019] [Accepted: 08/04/2019] [Indexed: 12/18/2022]
Abstract
Airway remodeling is one important feature of childhood asthma, which is one of the most common chronic childhood diseases. Phenotype switching of airway smooth muscle cells (ASMCs), defined as a reversible switching between contractile and proliferative phenotypes, plays an important role in the process of airway remodeling. Esculetin has shown antiinflammatory action in animal models of asthma; however, the effects of esculetin on ASMC phenotype switching have not been investigated. In the present study, platelet-derived growth factor (PDGF) was used to induce the phenotype modulation of ASMCs. The results demonstrated that esculetin pretreatment mitigated the PDGF-caused inhibitory effects on expressions of contractile phenotype protein markers, including calponin and SM22α. Esculetin also inhibited PDGF-induced migration and proliferation of ASMCs. Besides, the PDGF-induced expressions of extracellular matrix components, collagen I and fibronectin, were attenuated by esculetin pretreatment. Furthermore, PDGF-caused activation of PI3K/Akt pathway in ASMCs was inhibited by esculetin. These findings suggest that esculetin might exert its inhibitory effect on PDGF-induced ASMC phenotype switching through inhibition of PI3K/Akt pathway.
Collapse
Affiliation(s)
- Chundan Xie
- Department of Pediatrics, Huaihe Hospital of Henan University, Kaifeng, PR China
| | - Yanyang Li
- Department of Pediatrics, Huaihe Hospital of Henan University, Kaifeng, PR China
| | - Jie Gao
- Department of Pediatrics, Huaihe Hospital of Henan University, Kaifeng, PR China
| | - Yingying Wang
- Department of Pediatrics, Huaihe Hospital of Henan University, Kaifeng, PR China
| |
Collapse
|
39
|
Dong M, Ye T, Bi Y, Wang Q, Kuerban K, Li J, Feng M, Wang K, Chen Y, Ye L. A novel hybrid of 3-benzyl coumarin seco-B-ring derivative and phenylsulfonylfuroxan induces apoptosis and autophagy in non-small-cell lung cancer. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2019; 52:79-88. [PMID: 30599915 DOI: 10.1016/j.phymed.2018.09.216] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2018] [Revised: 09/20/2018] [Accepted: 09/25/2018] [Indexed: 06/09/2023]
Abstract
BACKGROUND Compound 6, as a novel hybrid of 3-benzyl coumarin seco-B-ring derivative and nitric oxide (NO) donor phenylsulfonylfuroxan, has the potential to develop into an anticancer drug because it displays significant antiproliferation activitity for various solid cancer cell lines including non-small-cell lung cancer (NSCLC) cells. PURPOSE We attempt to uncover the capacities of compound 6 to induce apoptosis and autophagy in NSCLC cells, as well as the underlying mechanism involved in this process. METHODS The effect of compound 6 on cell viability was evaluated in A549 cells by MTT assay. Apoptosis was mainly detected by flow cytometry. The induction of autophagy was observed by transmission electron microscopy (TEM), confocal microscopy as well as western-blotting technique. The expression of all related-proteins including PI3K/Akt/mTOR signaling pathway were also examined by western-blotting technique. RESULTS Above all, distinct growth inhibition and caspase-dependent apoptosis were detected in A549 cells administered with compound 6. Then, we confirmed the induction of autophagy triggered by compound 6 in A549 cells. Noticeably, blocking autophagy using a series of inhibitors and ATG5 siRNA had little effect on the cytotoxicity of compound 6, elucidating nonprotective autophagy triggered in NSCLC cells. Further research illustrated that PI3K/Akt/mTOR signaling pathway was involved in compound 6-induced apoptosis, and 3-MA as well as LY294002 had synergistic inhibiting effect on proliferation of A549 cells through the pathway mentioned above. CONCLUSION These findings raise a rationale that this 3-benzyl coumarin seco-B-ring derivative and phenylsulfonylfuroxan hybrid could be a promising candidate for developing as a therapeutic agent toward NSCLC, and the combination therapy through PI3K/Akt/mTOR signaling pathway may result in optimized treatment outcomes.
Collapse
Affiliation(s)
- Mengxue Dong
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Tao Ye
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Yongyan Bi
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Qian Wang
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Kudelaidi Kuerban
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Jiyang Li
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Meiqing Feng
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China
| | - Ke Wang
- Department of Medicinal Chemistry at School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ying Chen
- Department of Medicinal Chemistry at School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Li Ye
- Minghang Hospital & Department of Microbiological and Biochemical Pharmacy at School of Pharmacy, Fudan University, Shanghai 201199, China.
| |
Collapse
|
40
|
Caban M, Owczarek K, Chojnacka K, Lewandowska U. Overview of polyphenols and polyphenol-rich extracts as modulators of IGF-1, IGF-1R, and IGFBP expression in cancer diseases. J Funct Foods 2019. [DOI: 10.1016/j.jff.2018.11.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
41
|
Chen Y, Zhang Q, Liu H, Lu C, Liang CL, Qiu F, Han L, Dai Z. Esculetin Ameliorates Psoriasis-Like Skin Disease in Mice by Inducing CD4 +Foxp3 + Regulatory T Cells. Front Immunol 2018; 9:2092. [PMID: 30258447 PMCID: PMC6143660 DOI: 10.3389/fimmu.2018.02092] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 08/24/2018] [Indexed: 12/11/2022] Open
Abstract
Psoriasis is an autoimmune and inflammatory skin disease affecting around 2-3% of the world's population. Patients with psoriasis need extensive treatments with global immunosuppressive agents that may cause severe side effects. Esculetin, a type of coumarins, is an active ingredient extracted mainly from the bark of Fraxinus rhynchophylla, which has been used to treat inflammatory and autoimmune diseases in China. However, the antipsoriatic effects of esculetin have not been reported. In this study, we aimed to investigate the effects of esculetin on psoriatic skin inflammation in a mouse model and explored the potential molecular mechanisms underlying its action. We found that esculetin ameliorated the skin lesion and reduced PASI scores as well as weight loss in imiquimod-induced psoriasis-like mice, accompanied with weakened proliferation and differentiation of keratinocytes and T cell infiltration in esculetin-treated psoriatic mice. In addition, esculetin reduced the frequency of CD8+CD44highCD62Llow effector T cells in psoriatic mice. In contrast, it increased the frequency of CD4+Foxp3+ Tregs in both lymph nodes and spleens of the psoriatic mice while promoting the differentiation of CD4+CD25- T cells into CD4+Foxp3+ Tregs in vitro. Interestingly, depleting CD4+Foxp3+ Tregs largely reversed esculetin-mediated reduction in PASI scores, indicating that esculetin attenuates murine psoriasis mainly by inducing CD4+Foxp3+ Tregs. Furthermore, the mRNA levels of proinflammatory cytokines in the psoriatic mouse skin, including IL-6, IL-17A, IL-22, IL-23, TNF-α, and IFN-γ, were dramatically decreased by the treatment with esculetin. Finally, we found that esculetin inhibited the phosphorylation of IKKα and P65 in the psoriatic skin, suggesting that it inhibits the activation of NF-κB signaling. Thus, we have demonstrated that esculetin attenuates psoriasis-like skin lesion in mice and may be a potential therapeutic candidate for the treatment of psoriasis in clinic.
Collapse
Affiliation(s)
| | | | | | - Chuanjian Lu
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | | | | | | | - Zhenhua Dai
- Section of Immunology and Joint Immunology Program, Guangdong Provincial Academy of Chinese Medical Sciences and Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| |
Collapse
|
42
|
He Y, Li C, Ma Q, Chen S. Esculetin inhibits oxidative stress and apoptosis in H9c2 cardiomyocytes following hypoxia/reoxygenation injury. Biochem Biophys Res Commun 2018; 501:139-144. [DOI: 10.1016/j.bbrc.2018.04.195] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 04/25/2018] [Indexed: 12/20/2022]
|