1
|
Liu W, Nie F, Jiang H, Zhao Y, Zhang Y, Zhang Z, Zhang J, Xu J, Guo Y. Preparation of pH-Sensitive Polysaccharide-Small Molecule Nanoparticles and Their Applications for Tumor Chemo- and Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2024; 16:68437-68452. [PMID: 39586061 DOI: 10.1021/acsami.4c16504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Hydrophobic chemotherapy drugs face significant challenges in cancer treatment, including low bioavailability, unavoidable toxic side effects, and the development of drug resistance. To address these issues, a multifunctional nanoplatform was developed for cancer therapy, aimed at achieving effective drug delivery and enhancing antitumor efficacy. Poria cocos polysaccharide (PCP), a natural polymer known for its immunomodulatory properties, was utilized as an immunoreactive vector for drug delivery after being cross-linked with 1,4-phenylenebisboronic acid (BDBA). Subsequently, a small-molecule chemotherapy drug, esculetin (EL), was confirmed through density functional theory (DFT) simulations to be encapsulated within the PCP-BDBA nanoparticles via weak interactions. The results demonstrated that the synthesized nanoparticles were spherical, with an average particle size of 162.0 nm. In addition to exhibiting excellent stability, the nanoparticles also displayed pH-responsive drug release properties. In vivo experiments indicated that EL@PCP-BDBA NPs exhibited antitumor effects. Furthermore, EL@PCP-BDBA NPs showed superior in vitro antitumor activity compared to EL at the cellular level. Additionally, EL@PCP-BDBA NPs were found to increase intracellular reactive oxygen species (ROS) levels, induce cell apoptosis, and suppress cell migration to combat cancer. Meanwhile, EL@PCP-BDBA NPs enhanced immune function in vivo. In summary, this study developed a nano-pharmaceutical that combined chemotherapy and immunotherapy functions, which was considered a promising tool for cancer therapy.
Collapse
Affiliation(s)
- Wenhui Liu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Fan Nie
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Haojing Jiang
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yinan Zhao
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yan Zhang
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, People's Republic of China
| | - Zheng Zhang
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300071, People's Republic of China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, People's Republic of China
| | - Jie Zhang
- School of Chemistry and Chemical Engineering/State Key Laboratory Incubation Base for Green Processing of Chemical Engineering, Shihezi University, Shihezi, Xinjiang 832003, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| | - Yuanqiang Guo
- State Key Laboratory of Medicinal Chemical Biology, College of Pharmacy, and Tianjin Key Laboratory of Molecular Drug Research, Nankai University, Tianjin 300350, People's Republic of China
| |
Collapse
|
2
|
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
|
3
|
Park SH. Role of Phytochemicals in Treatment of Aging and Cancer: Focus on Mechanism of FOXO3 Activation. Antioxidants (Basel) 2024; 13:1099. [PMID: 39334758 PMCID: PMC11428386 DOI: 10.3390/antiox13091099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/30/2024] Open
Abstract
There have been many studies reporting that the regular consumption of fruits and vegetables is associated with reduced risks of cancer and age-related chronic diseases. Recent studies have demonstrated that reducing reactive oxygen species and inflammation by phytochemicals derived from natural sources can extend lifespans in a range of model organisms. Phytochemicals derived from fruits and vegetables have been known to display both preventative and suppressive activities against various types of cancer via in vitro and in vivo research by interfering with cellular processes critical for tumor development. The current challenge lies in creating tailored supplements containing specific phytochemicals for individual needs. Achieving this goal requires a deeper understanding of the molecular mechanisms through which phytochemicals affect human health. In this review, we examine recently (from 2010 to 2024) reported plant extracts and phytochemicals with established anti-aging and anti-cancer effects via the activation of FOXO3 transcriptional factor. Additionally, we provide an overview of the cellular and molecular mechanisms by which these molecules exert their anti-aging and anti-cancer effects in specific model systems. Lastly, we discuss the limitations of the current research approach and outline for potential future directions in this field.
Collapse
Affiliation(s)
- See-Hyoung Park
- Department of Biological and Chemical Engineering, Hongik University, Sejong 30016, Republic of Korea
| |
Collapse
|
4
|
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: 0] [Impact Index Per Article: 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
|
5
|
Law YY, Lee HL, Lin CL, Chen PN, Wang PH, Hsieh YH, Chen CM. Asiatic acid inhibits osteosarcoma cell migration and invasion via the AKT/Sp1/MMP1 axis. ENVIRONMENTAL TOXICOLOGY 2024; 39:3920-3929. [PMID: 38567545 DOI: 10.1002/tox.24246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 02/27/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024]
Abstract
Osteosarcoma is a malignant bone tumor affecting adolescents and children. No effective treatment is currently available. Asiatic acid (AA), a triterpenoid compound found in Centella asiatica, possesses anti-tumor, anti-inflammatory, and anti-oxidant properties in various types of tumor cells. This study aims to determine whether AA exerts antitumor effects in human osteosarcoma cells. Our results indicate that AA does not influence the viability, proliferative rate, or cell cycle phase of human osteosarcoma cells under non-toxic conditions. AA suppressed osteosarcoma cell migration and invasion by down-regulating matrix metalloproteinase 1 (MMP1) expression. Data in the TNMplot database suggested MMP1 expression was higher in osteosarcoma than in normal tissues, with associated clinical significance observed in osteosarcoma patients. Overexpression of MMP1 in osteosarcoma cells reversed the AA-induced suppression of cell migration and invasion. AA treatment decreased the expression of specificity protein 1 (Sp1), while Sp1 overexpression abolished the effect of AA on MMP1 expression and cell migration and invasion. AA inhibited AKT phosphorylation, and treatment with a PI3K inhibitor (wortmannin) increased the anti-invasive effect of AA on osteosarcoma cells via the p-AKT/Sp1/MMP1 axis. Thus, AA exhibits the potential for use as an anticancer drug against human osteosarcoma.
Collapse
Affiliation(s)
- Yat-Yin Law
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Orthopedics, Chung Shan Medical University Hospital, Taichung, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Hsiang-Lin Lee
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Surgery, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chu-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Ni Chen
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Pei-Han Wang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chien-Min Chen
- Division of Neurosurgery, Department of Surgery, Changhua Christian Hospital, Changhua, Taiwan
- Department of Leisure Industry Management, National Chin-Yi University of Technology, Taichung, Taiwan
- Department of Biomedical Sciences, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
6
|
Salem MG, Alqahtani AM, Mali SN, Alshwyeh HA, Jawarkar RD, Altamimi AS, Alshawwa SZ, Al-Olayan E, Saied EM, Youssef MF. Synthesis and antiproliferative evaluation of novel 3,5,8-trisubstituted coumarins against breast cancer. Future Med Chem 2024; 16:1053-1073. [PMID: 38708686 PMCID: PMC11216633 DOI: 10.4155/fmc-2023-0375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Accepted: 02/13/2024] [Indexed: 05/07/2024] Open
Abstract
Aim: This study focused on designing and synthesizing novel derivatives of 3,5,8-trisubstituted coumarin. Results: The synthesized compounds, particularly compound 5, exhibited significant cytotoxic effects on MCF-7 cells, surpassing staurosporine, and reduced toxicity toward MCF-10A cells, highlighting potential pharmacological advantages. Further, compound 5 altered the cell cycle and significantly increased apoptosis in MCF-7 cells, involving both early (41.7-fold) and late stages (33-fold), while moderately affecting necrotic signaling. The antitumor activity was linked to a notable reduction (4.78-fold) in topoisomerase IIβ expression. Molecular modeling indicated compound 5's strong affinity for EGFR, human EGF2 and topoisomerase II proteins. Conclusion: These findings highlight compound 5 as a multifaceted antitumor agent for breast cancer.
Collapse
Affiliation(s)
- Manar G Salem
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Suez Canal University, Ismailia, 41522, Egypt
| | - Alaa M Alqahtani
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Umm Al-Qura University, Makkah, 21955, Saudi Arabia
| | - Suraj N Mali
- School of Pharmacy, DY Patil Deemed to be University Sector 7, Nerul, Navi Mumbai, 400706, India
| | - Hussah Abdullah Alshwyeh
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, 31441, Saudi Arabia
- Basic & Applied Scientific Research Centre, Imam Abdulrahman Bin Faisal University, PO Box 1982, Dammam, 31441, Saudi Arabia
| | - Rahul D Jawarkar
- Department of Medicinal Chemistry & Drug Discovery, Dr. Rajendra Gode Institute of Pharmacy, University Mardi Road, Amravati, 444603, India
| | - Abdulmalik S Altamimi
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, PO Box 173, Alkharj, 11942, Saudi Arabia
| | - Samar Z Alshawwa
- Department of Pharmaceutical Sciences, College of Pharmacy, Princess Nourah bint Abdulrahman University, PO Box 84428, Riyadh, 11671, Saudi Arabia
| | - Ebtesam Al-Olayan
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Essa M Saied
- Chemistry Department (Biochemistry Division), Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
- Institute for Chemistry, Humboldt Universität zu Berlin, Brook-Taylor-Str. 2, Berlin, 12489, Germany
| | - Mohamed F Youssef
- Chemistry Department (Organic Chemistry Division), Faculty of Science, Suez Canal University, Ismailia, 41522, Egypt
| |
Collapse
|
7
|
Rezoan Hossain M, Zahra Shova FT, Akter M, Shuvo S, Ahmed N, Akter A, Haque M, Salma U, Roman Mogal M, Saha HR, Sarkar BC, Sohel M. Esculetin unveiled: Decoding its anti-tumor potential through molecular mechanisms-A comprehensive review. Cancer Rep (Hoboken) 2024; 7:e1948. [PMID: 38062981 PMCID: PMC10809201 DOI: 10.1002/cnr2.1948] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/10/2023] [Accepted: 11/16/2023] [Indexed: 01/27/2024] Open
Abstract
BACKGROUND The growing complexity of cancer has made it a significant concern in the medical community. Although cancer research has advanced, it is still challenging to create new effective medications due to the limitations and side effects of existing treatment strategies. These are enforcing the development of some alternative drugs from natural compounds with fewer drawbacks and side effects. AIM Therefore, this review aims to provide up-to-date, crucial, and all-encompassing data on esculetin's anticancer activity, including all relevant molecular and cellular processes based on in vivo and in vitro investigations. RESULTS According to the literature review, esculetin is available in nature and is effective against 16 different types of cancer. The general mechanism shown by esculetin is modulating signaling cascades and its related pathways, like cell proliferation, cell growth, autophagy, apoptosis, necrosis, inflammation, angiogenesis, metastasis, invasion, and DNA damage. Nanoformulation of esculetin improves this natural product's efficacy by improving water solubility. Esculetin's synergistic effects with both natural substances and conventional treatments have been shown, and this method aids in reversing resistance mechanisms by modulating resistance-related proteins. In addition, it has fewer side effects on humans than other phytochemicals and standard drugs with some good pharmacokinetic features. CONCLUSION Therefore, until standard chemotherapeutics are available in pharmaceutical markets, esculetin should be used as a therapeutic drug against various cancer types.
Collapse
Affiliation(s)
| | - Fatema Tuj Zahra Shova
- Biotechnology and Genetic EngineeringMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Munni Akter
- Department of MedicalDinajpure Nursing College (Affiliated Rajshahi University)DinajpurBangladesh
| | - Shahporan Shuvo
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Nasim Ahmed
- Department of PharmacyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Afroza Akter
- Departmnet of MicrobiologyNoakhali Science and Technology UniversityNoakhaliBangladesh
| | - Munira Haque
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Umme Salma
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | - Md Roman Mogal
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
| | - Hasi Rani Saha
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| | | | - Md Sohel
- Biochemistry and Molecular BiologyMawlana Bhashani Science and Technology UniversityTangailBangladesh
- Biochemistry and Molecular BiologyPrimeasia UniversityDhakaBangladesh
| |
Collapse
|
8
|
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
|
9
|
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: 2] [Impact Index Per Article: 1.0] [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
|
10
|
Lin CH, Lin KH, Ku HJ, Lee KC, Lin SS, Hsu FT. Amentoflavone induces caspase-dependent/-independent apoptosis and dysregulates cyclin-dependent kinase-mediated cell cycle in colorectal cancer in vitro and in vivo. ENVIRONMENTAL TOXICOLOGY 2023; 38:1078-1089. [PMID: 36727907 DOI: 10.1002/tox.23749] [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: 10/08/2022] [Revised: 12/27/2022] [Accepted: 01/16/2023] [Indexed: 06/18/2023]
Abstract
Colorectal cancer (CRC) is recognized as the third most common malignancy and the second most deadly in highly developed countries. Although the treatment of CRC has improved in the past decade, the mortality rate of CRC is still increasing. Amentoflavone, one of the flavonoids detected in medical plants, is reported to possess potential anticancer properties in various cancers. However, its role in CRC has not been studied. This study aimed to investigate the role and underlying mechanism of amentoflavone on CRC in vitro and in vivo. We identified the cytotoxicity, apoptosis effect, cell cycle alteration, DNA damage induction and tumor progression inhibition of amentoflavone in HT-29 model by using MTT assay, flow cytometry, immunofluorescence (IF) staining, Western blotting and animal experiments. Amentoflavone induced cytotoxicity is caused by triggering G1 arrest, DNA damage and apoptosis in HT-29 cells. The expression of cyclin D1, CDK4 and CDK6 was decreased by amentoflavone; in contrast, the phosphorylation of ATM and CHK2 and the expression of p21 and p27 were increased. The apoptosis induction of amentoflavone in CRC is not only caspase-dependent but also increases EndoG and AIF nuclear translocation in a caspase-independent manner. Importantly, the apoptosis induction of amentoflavone is not affected by the activity of p53 in CRC. Amentoflavone suppressed the progression of CRC by initiating G1 arrest and ATM/CHK2-mediated DNA damage-responsive, caspase-dependent/independent apoptotic effects. We uncovered a novel tumor-inhibitory role of amentoflavone in CRC that is not associated with p53 activity, which may serve as a potential treatment for CRC.
Collapse
Affiliation(s)
- Cheng-Hsun Lin
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Kuang-Hsuan Lin
- Department of Radiation Oncology, Chang Bing Show Chwan Memorial Hospital, Changhua, Taiwan
| | - Hsiang-Ju Ku
- Institute of Biotechnology, National Tsing Hua University, Hsinchu, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Kun-Ching Lee
- Department of Radiation Oncology, National Yang Ming Chiao Tung University Hospital, Yilan, Taiwan
| | - Song-Shei Lin
- Department of Medical Imaging and Radiological Sciences, Central Taiwan University of Science and Technology, Taichung, Taiwan
| | - Fei-Ting Hsu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| |
Collapse
|
11
|
Zhang D, Fu Y, Liu Y, Wu Y, Chen J, Zhang L, Wang R, Chen Z, Liu T. 8-Methoxyflindersine-Induced Apoptosis and Cell Cycle Disorder Involving MAPK Signaling Activation in Human Colorectal Cancer Cells. Int J Mol Sci 2023; 24:ijms24098039. [PMID: 37175741 PMCID: PMC10179151 DOI: 10.3390/ijms24098039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/23/2023] [Accepted: 04/27/2023] [Indexed: 05/15/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most common malignant tumors with a high lethal rate globally, and novel strategies for its prevention and therapy are urgently needed. In our previous work, 8-methoxyflindersine (8-MF), a quinoline alkaloid, was isolated from the Dictamni cortex, and its bioactivities were largely unknown. In this study, we found that 8-MF significantly inhibited cell viability in the CRC cell lines LoVo and RKO. The 8-MF-induced CRC cell apoptosis, as well as cell cycle disorder, were further verified by cyclins dysregulation in mRNA and protein levels. Further, the activation of MAPK family members p38 and ERK1/2 was observed after 8-MF treatment. Moreover, the protein-protein interaction of 8-MF with cyclins and MAPKs was demonstrated using the STRING database. The 8-MF could bind to p38 and ERK1/2 proteins in molecular docking. Taken together, we found that 8-MF induced apoptosis and cell cycle disorder involving MAPK signaling activation in CRC cells, indicating 8-MF as a novel lead compound candidate for the development of anti-tumor drugs for CRC.
Collapse
Affiliation(s)
- Dianbao Zhang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Yunmei Fu
- Department of Natural Products Chemistry, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Ying Liu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Yifan Wu
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Jiayu Chen
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Luting Zhang
- Department of Natural Products Chemistry, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Rui Wang
- Department of Stem Cells and Regenerative Medicine, Key Laboratory of Cell Biology, National Health Commission of China, and Key Laboratory of Medical Cell Biology, Ministry of Education of China, China Medical University, Shenyang 110122, China
| | - Zaixing Chen
- Central Laboratory, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Tao Liu
- Department of Natural Products Chemistry, School of Pharmacy, China Medical University, Shenyang 110122, China
| |
Collapse
|
12
|
Chen R, Liu J, Hu J, Li C, Liu Y, Pan W. DLGAP5 knockdown inactivates the Wnt/β-catenin signal to repress endometrial cancer cell malignant activities. ENVIRONMENTAL TOXICOLOGY 2023; 38:685-693. [PMID: 36454672 DOI: 10.1002/tox.23720] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Revised: 11/03/2022] [Accepted: 11/20/2022] [Indexed: 06/17/2023]
Abstract
Human discs large-associated protein 5 (DLGAP5), a microtubule-associated protein, has been reported to be upregulated in several tumors. However, the role of DLGAP5 in endometrial cancer (EC) progression and the related underlying mechanism were still unknown. A bioinformatics analysis was performed to analyze the expression and prognostic significance of DLGAP5 in EC tissues using TCGA, CPTAC, Human Protein Atlas, and GSE63678 databases, UALCAN web tool, and the Kaplan-Meier plotter. Effects of DLGAP on EC cell malignant properties were evaluated by CCK-8, flow cytometry analysis, TUNEL assay, caspase-3 activity assay, and Transwell invasion assay. The expression of DLGAP5, Wnt3, c-Myc, Ki67, and cleaved caspase-3 was detected by western blot analysis. DLGAP5 was highly expressed and correlated with poor prognosis in EC patients. DLGAP5 knockdown inhibited proliferation and invasion, triggered apoptosis, and increased caspase-3 activity in EC cells. Additionally, DLGAP5 knockdown inactivated the Wnt/β-catenin signaling pathway in EC cells. Moreover, β-catenin overexpression abolished the effects of DLGAP5 knockdown on the malignant phenotypes of EC cells. DLGAP5 silencing suppressed the malignant properties in EC cells by inactivating the Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Ruipu Chen
- Department of Obstetrics, Fokind Hospital Affiliated to Tibet University, Lhasa, Tibet, China
| | - Jing Liu
- Department of Obstetrics, Fokind Hospital Affiliated to Tibet University, Lhasa, Tibet, China
| | - Jun Hu
- Department of Obstetrics, Fokind Hospital Affiliated to Tibet University, Lhasa, Tibet, China
| | - Chunxia Li
- Department of Obstetrics, Fokind Hospital Affiliated to Tibet University, Lhasa, Tibet, China
| | - Yanhua Liu
- Department of Obstetrics, Fokind Hospital Affiliated to Tibet University, Lhasa, Tibet, China
| | - Weiwei Pan
- Department of Intensive Care Unit, Huai'an Second People's Hospital and The Affiliated Huai'an Hospital of Xuzhou Medical University, Huai'an, Jiangsu, China
| |
Collapse
|
13
|
Yin W, Fu X, Chang W, Han L, Meng J, Cao A, Ren X, Fan Z, Zhou S. Antiovarian cancer mechanism of esculetin: inducing G0/G1 arrest and apoptosis via JAK2/STAT3 signalling pathway. J Pharm Pharmacol 2023; 75:87-97. [PMID: 36332079 DOI: 10.1093/jpp/rgac083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 10/11/2022] [Indexed: 11/06/2022]
Abstract
OBJECTIVES Esculetin is a coumarin derivative, which is extracted from the dried barks of fraxinus chinensis Roxb. Although it is reported esculetin possesses multiple pharmacological activities, its associated regulatory mechanism on ovarian cancer isn't well investigated. METHODS Cytotoxicity is evaluated by MTT, clonogenic and living/dead cells staining assays. Migration and invasion effects are investigated by wound healing, and transwell assays. The effect of cell cycle and apoptosis are analyzed by flow cytometry and western blotting. Mitochondrial membrane potential and intracellular reactive oxygen species (ROS) is assessed by fluorescence microscope. Analysis of animal experiments are carried out by various pathological section assays. KEY FINDINGS Esculetin exerts an anti- ovarian cancer effect. It is found that apoptosis induction is promoted by the accumulation of excessive ROS and inhibition of JAK2/STAT3 signalling pathway. In addition, exposure to esculetin leads to the cell viability reduction, migration and invasion capability decrease and G0/G1 phase cell cycle arrest induced by down-regulating downstream targets of STAT3. In vivo experimental results also indicate esculetin can inhibit tumour growth of mice. CONCLUSIONS Our study provides some strong evidences to support esculetin as a potential anti-cancer agent in ovarian cancer.
Collapse
Affiliation(s)
- Wen Yin
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, China
| | - Xu Fu
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Wenwen Chang
- Lanzhou University Second Clinical Medical College/Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Li Han
- Key Laboratory of Emergency Medicine, Lanzhou University Second Hospital, Lanzhou, China
| | - Jiahao Meng
- Department of biomaterials, College of Materials, Xiamen University, Xiamen, China
| | - Aijia Cao
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, China
| | - Xiaomin Ren
- Lanzhou University Second Clinical Medical College/Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Zhongxiong Fan
- Department of biomaterials, College of Materials, Xiamen University, Xiamen, China.,Institute of Materia Medica, Xinjiang University, Urumqi, China
| | - Suqin Zhou
- Department of Pharmacy, Lanzhou University Second Hospital, Lanzhou, China
| |
Collapse
|
14
|
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
|
15
|
He Q, Liu C, Wang X, Rong K, Zhu M, Duan L, Zheng P, Mi Y. Exploring the mechanism of curcumin in the treatment of colon cancer based on network pharmacology and molecular docking. Front Pharmacol 2023; 14:1102581. [PMID: 36874006 PMCID: PMC9975159 DOI: 10.3389/fphar.2023.1102581] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 02/07/2023] [Indexed: 02/17/2023] Open
Abstract
Objective: Curcumin is a plant polyphenol extracted from the Chinese herb turmeric. It was found that curcumin has good anti-cancer properties in a variety of cancers, but the exact mechanism is not clear. Based on the network pharmacology and molecular docking to deeply investigate the molecular mechanism of curcumin for the treatment of colon cancer, it provides a new research direction for the treatment of colon cancer. Methods: Curcumin-related targets were collected using PharmMapper, SwissTargetPrediction, Targetnet and SuperPred. Colon cancer related targets were obtained using OMIM, DisGeNET, GeneCards and GEO databases. Drug-disease intersection targets were obtained via Venny 2.1.0. GO and KEGG enrichment analysis of drug-disease common targets were performed using DAVID. Construct PPI network graphs of intersecting targets using STRING database as well as Cytoscape 3.9.0 and filter core targets. Molecular docking via AutoDockTools 1.5.7. The core targets were further analyzed by GEPIA, HPA, cBioPortal and TIMER databases. Results: A total of 73 potential targets of curcumin for the treatment of colon cancer were obtained. GO function enrichment analysis yielded 256 entries, including BP(Biological Progress):166, CC(celluar component):36 and MF(Molecular Function):54. The KEGG pathway enrichment analysis yielded 34 signaling pathways, mainly involved in Metabolic pathways, Nucleotide metabolism, Nitrogen metabolism, Drug metabolism - other enzymes, Pathways in cancer,PI3K-Akt signaling pathway, etc. CDK2, HSP90AA1, AURKB, CCNA2, TYMS, CHEK1, AURKA, DNMT1, TOP2A, and TK1 were identified as core targets by Cytoscape 3.9.0. Molecular docking results showed that the binding energies of curcumin to the core targets were all less than 0 kJ-mol-1, suggesting that curcumin binds spontaneously to the core targets. These results were further validated in terms of mRNA expression levels, protein expression levels and immune infiltration. Conclusion: Based on network pharmacology and molecular docking initially revealed that curcumin exerts its therapeutic effects on colon cancer with multi-target, multi-pathway. Curcumin may exert anticancer effects by binding to core targets. Curcumin may interfere with colon cancer cell proliferation and apoptosis by regulating signal transduction pathways such as PI3K-Akt signaling pathway,IL-17 signaling pathway, Cell cycle. This will deepen and enrich our understanding of the potential mechanism of curcumin against colon cancer and provide a theoretical basis for subsequent studies.
Collapse
Affiliation(s)
- Qingmin He
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China
| | - Chuan Liu
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaohan Wang
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Kang Rong
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Mingyang Zhu
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Liying Duan
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Pengyuan Zheng
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China.,Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Mi
- Henan Key Laboratory of Helicobacter Pylori and Microbiota and Gastrointestinal Cancer, Marshall B. J. Medical Research Center of Zhengzhou University, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.,Academy of Medical Science, Zhengzhou University, Zhengzhou, Henan, China.,Department of Gastroenterology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
16
|
Mesas C, Garcés V, Martínez R, Ortiz R, Doello K, Dominguez-Vera JM, Bermúdez F, Porres JM, López-Jurado M, Melguizo C, Delgado-López JM, Prados J. Colon cancer therapy with calcium phosphate nanoparticles loading bioactive compounds from Euphorbia lathyris: In vitro and in vivo assay. Biomed Pharmacother 2022; 155:113723. [PMID: 36156367 DOI: 10.1016/j.biopha.2022.113723] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/15/2022] [Accepted: 09/19/2022] [Indexed: 02/07/2023] Open
Abstract
Amorphous calcium phosphate nanoparticles (ACP NPs) exhibit excellent biocompatibility and biodegradability properties. ACP NPs were functionalized with two coumarin compounds (esculetin and euphorbetin) extracted from Euphorbia lathyris seeds (BC-ACP NPs) showing high loading capacity (0.03% and 0.34% (w/w) for esculetin and euphorbetin, respectively) and adsorption efficiency (2.6% and 33.5%, respectively). BC-ACP NPs, no toxic to human blood cells, showed a more selective cytotoxicity against colorectal cancer (CRC) cells (T-84 cells) (IC50, 71.42 µg/ml) compared to non-tumor (CCD18) cells (IC50, 420.77 µg/ml). Both, the inhibition of carbonic anhydrase and autophagic cell death appeared to be involved in their action mechanism. Interestingly, in vivo treatment with BC-ACPs NPs using two different models of CRC induction showed a significant reduction in tumor volume (62%) and a significant decrease in the number and size of polyps. A poor development of tumor vasculature and invasion of normal tissue were also observed. Moreover, treatment increased the bacterial population of Akkermansia by restoring antioxidant systems in the colonic mucosa of mice. These results show a promising pathway to design innovative and more efficient therapies against CRC based on biomimetic calcium phosphate NPs loaded with natural products.
Collapse
Affiliation(s)
- Cristina Mesas
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Víctor Garcés
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, 18071 Granada, Spain
| | - Rosario Martínez
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Raúl Ortiz
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18014 Granada, Spain
| | - Kevin Doello
- Medical Oncology Service, Virgen de las Nieves Hospital, 18014 Granada, Spain
| | - Jose M Dominguez-Vera
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, 18071 Granada, Spain
| | - Francisco Bermúdez
- Cellbitec S.L., N.I.F. B04847216, Scientific Headquarters of the Almería Technology Park, Universidad de Almería, La Cañada, 04128 Almería, Spain
| | - Jesús M Porres
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - María López-Jurado
- Department of Physiology, Institute of Nutrition and Food Technology (INyTA), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain
| | - Consolación Melguizo
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18014 Granada, Spain.
| | - José M Delgado-López
- Department of Inorganic Chemistry, Faculty of Science, University of Granada, 18071 Granada, Spain
| | - Jose Prados
- Institute of Biopathology and Regenerative Medicine (IBIMER), Center of Biomedical Research (CIBM), University of Granada, 18100 Granada, Spain; Department of Anatomy and Embryology, Faculty of Medicine, University of Granada, 18071 Granada, Spain; Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA), 18014 Granada, Spain
| |
Collapse
|
17
|
Qin X, Wang X, Xu K, Yang X, Wang Q, Liu C, Wang X, Guo X, Sun J, Li L, Li S. Synergistic antitumor effects of polysaccharides and anthocyanins from Lycium ruthenicum Murr. on human colorectal carcinoma LoVo cells and the molecular mechanism. Food Sci Nutr 2022; 10:2956-2968. [PMID: 36171788 PMCID: PMC9469862 DOI: 10.1002/fsn3.2892] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 03/15/2022] [Accepted: 04/05/2022] [Indexed: 11/11/2022] Open
Abstract
The antitumor effects of Lycium ruthenicum Murr. polysaccharides (LRPS) and Lycium ruthenicum Murr. anthocyanins (LRAC) were comprehensively investigated in this study. LPRS was obtained by water extraction and alcohol precipitation and further purified using diethylaminoethyl cellulose (DEAE-Cellulose) and Sephadex G-75 columns. High-performance liquid chromatography (HPLC) and Fourier transform-infrared (FT-IR) spectroscopy were used to characterize the purified LRPS. The results showed that the purified LRPS contained heteropolysaccharides, mainly composed of arabinose, galactose, and glucose with weight percentage of 41.2%, 33.6%, and 10.8%, respectively. More importantly, LRPS (500 μg/ml) and LRAC (80 μg/ml) failed to impede the proliferation of tumor cells when applied solely (48 h incubation), yet remarkable antineoplastic effects were found once they were applied altogether, since the LoVo cells, a typical human colorectal carcinoma cell line, were significantly inhibited by the mixture of LRPS (150 μg/ml) and LRAC (20 μg/ml) (LRPS&AC) in 24 h. The antineoplastic activity resulted from the combination of both LRPS and LRAC (LRPS&AC), by means of blocking the cell cycle at the G0-G1 phase and inducing LoVo cell apoptosis via reactive oxygen species (ROS)-dependent pathway. The inhibitory effects of LRPS&AC were specific to the tumor cells, without imposing on the proliferation of normal cells. Western blotting revealed that the antitumor effect was related to the mitochondria-mediated apoptosis launched by the cross-action of PI3K/Akt (phosphatidylinositol 3-kinase/protein kinase B) and JAK2/STAT3 (janus kinase 2/signal transduction and activator of transcription 3) signaling pathways. These findings for the first time reveal the synergistic antitumor effects of LRPS&AC and the related mechanisms, which enable Lycium ruthenicum Murr. to serve as a natural source to develop therapeutic reagents and functional foods with antineoplastic properties.
Collapse
Affiliation(s)
- Xinshu Qin
- Key Laboratory of Novel Food Resources ProcessingMinistry of Agriculture and Rural AffairsKey Laboratory of Agro‐Products Processing Technology of Shandong ProvinceInstitute of Agro‐Food Science and TechnologyShandong Academy of Agricultural SciencesJi'nanChina
- Shaanxi Engineering Laboratory for Food Green Processing and Safety ControlShaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural ProductsCollege of Food Engineering and Nutritional ScienceShaanxi Normal UniversityXi'anChina
| | - Xingyu Wang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety ControlShaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural ProductsCollege of Food Engineering and Nutritional ScienceShaanxi Normal UniversityXi'anChina
| | - Ke Xu
- Department of Joint Surgery, Hong Hui HospitalXi'an Jiaotong UniversityXi'anChina
| | - Xingbin Yang
- Shaanxi Engineering Laboratory for Food Green Processing and Safety ControlShaanxi Key Laboratory for Hazard Factors Assessment in Processing and Storage of Agricultural ProductsCollege of Food Engineering and Nutritional ScienceShaanxi Normal UniversityXi'anChina
| | - Qing Wang
- Key Laboratory of Novel Food Resources ProcessingMinistry of Agriculture and Rural AffairsKey Laboratory of Agro‐Products Processing Technology of Shandong ProvinceInstitute of Agro‐Food Science and TechnologyShandong Academy of Agricultural SciencesJi'nanChina
| | - Chao Liu
- Key Laboratory of Novel Food Resources ProcessingMinistry of Agriculture and Rural AffairsKey Laboratory of Agro‐Products Processing Technology of Shandong ProvinceInstitute of Agro‐Food Science and TechnologyShandong Academy of Agricultural SciencesJi'nanChina
| | - Xinkun Wang
- Key Laboratory of Novel Food Resources ProcessingMinistry of Agriculture and Rural AffairsKey Laboratory of Agro‐Products Processing Technology of Shandong ProvinceInstitute of Agro‐Food Science and TechnologyShandong Academy of Agricultural SciencesJi'nanChina
| | - Xu Guo
- Key Laboratory of Novel Food Resources ProcessingMinistry of Agriculture and Rural AffairsKey Laboratory of Agro‐Products Processing Technology of Shandong ProvinceInstitute of Agro‐Food Science and TechnologyShandong Academy of Agricultural SciencesJi'nanChina
| | - Jinyue Sun
- Key Laboratory of Novel Food Resources ProcessingMinistry of Agriculture and Rural AffairsKey Laboratory of Agro‐Products Processing Technology of Shandong ProvinceInstitute of Agro‐Food Science and TechnologyShandong Academy of Agricultural SciencesJi'nanChina
| | - Lin Li
- Santa Barbara City CollegeUniversity of California Santa BarbaraSanta BarbaraCaliforniaUSA
| | - Shiqi Li
- Department of Material Science and EngineeringQueen Mary University of London Engineering SchoolNorthwestern Polytechnical UniversityXi'anChina
| |
Collapse
|
18
|
Alam M, Hasan GM, Ansari MM, Sharma R, Yadav DK, Hassan MI. Therapeutic implications and clinical manifestations of thymoquinone. PHYTOCHEMISTRY 2022; 200:113213. [PMID: 35472482 DOI: 10.1016/j.phytochem.2022.113213] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 04/16/2022] [Accepted: 04/17/2022] [Indexed: 06/14/2023]
Abstract
Thymoquinone (TQ), a natural phytochemical predominantly found in Nigella sativa, has been investigated for its numerous health benefits. TQ showed anti-cancer, anti-oxidant, and anti-inflammatory properties, validated in various disease models. The anti-cancer potential of TQ is goverened by anti-proliferation, cell cycle arrest, apoptosis induction, ROS production, anti-metastasis and anti-angiogenesis, inhibition of cell migration and invasion action. Additionally, TQ exhibited antitumor activity via the modulation of multiple pathways and molecular targets, including Akt, ERK1/2, STAT3, and NF-κB. The present review highlighted the anticancer potential of TQ . We summarize the anti-cancer, anti-oxidant, and anti-inflammatory properties of TQ, focusing on its molecular targets and its promising action in cancer therapy. We further described the molecular mechanisms by which TQ prevents signaling pathways that mediate cancer progression, invasion, and metastasis.
Collapse
Affiliation(s)
- Manzar Alam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India
| | - Gulam Mustafa Hasan
- Department of Biochemistry, College of Medicine, Prince Sattam Bin Abdulaziz University, P.O. Box 173, Al-Kharj, 11942, Saudi Arabia
| | - Md Meraj Ansari
- Centre for Pharmaceutical Nanotechnology, Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, SAS Nagar, Mohali, Punjab, 160062, India
| | - Rishi Sharma
- Department of Forensic Medicine and Toxicology, All India Institute of Medical Sciences, Rishikesh, Uttarakhand, 249203, India
| | - Dharmendra Kumar Yadav
- College of Pharmacy, Gachon University of Medicine and Science, Hambakmoeiro, Yeonsu-gu, Incheon City, 21924, South Korea.
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, New Delhi, 110025, India.
| |
Collapse
|
19
|
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
|
20
|
Alnuqaydan AM, Rah B. Tamarix articulata Inhibits Cell Proliferation, Promotes Cell Death Mechanisms and Triggers G 0/G 1 Cell Cycle Arrest in Hepatocellular Carcinoma Cells. Food Technol Biotechnol 2021; 59:162-173. [PMID: 34316277 PMCID: PMC8284106 DOI: 10.17113/ftb.59.02.21.6904] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 03/21/2021] [Indexed: 01/10/2023] Open
Abstract
RESEARCH BACKGROUND From ancient times plants have been used for medicinal purposes against various ailments. In the modern era, plants are a major source of drugs and are an appealing drug candidate for the anticancer therapeutics against various molecular targets. Here we tested methanolic extract of dry leaves of Tamarix articulata for anticancer activity against a panel of hepatocellular carcinoma cells. EXPERIMENTAL APPROACH Cell viability of hepatocellular carcinoma cells was determined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay after a dose-dependent treatment with the extract of T. articulata. Phase-contrast microscopy and 4՛,6-diamidino-2-phenylindole (DAPI) staining served to analyse cellular and nuclear morphology. Immunoblotting was performed to determine the expression of proteins associated with autophagy, apoptosis and cell cycle. However, flow cytometry was used for the quantification of apoptotic cells and the analysis of cells in different phases of the cycle after the treatment with various doses of T. articulata. Additionally, acridine orange staining and 2՛,7՛-dichlorofluorescein diacetate (DCFH-DA) dye were used to analyse the quantification of autophagosomes and reactive oxygen species. RESULTS AND CONCLUSION Our results demonstrate that T. articulata methanolic extract exhibits promising antiproliferative activity with IC50 values (271.1±4.4), (298.3±7.1) and (336.7±6.1) µg/mL against hepatocellular carcinoma HepG2, Huh7D12 and Hep3B cell lines, respectively. Mechanistically, we found that T. articulata methanolic extract induces cell death by activating apoptosis and autophagy pathways. First, T. articulata methanolic extract promoted autophagy, which was confirmed by acridine orange staining. The immunoblotting analysis further confirmed that the extract at higher doses consistently induced the conversion of LC3I to LC3II form with a gradual decrease in the expression of autophagy substrate protein p62. Second, T. articulata methanolic extract promoted reactive oxygen species production in hepatocellular carcinoma cells and activated reactive oxygen species-mediated apoptosis. Flow cytometry and immunoblotting analysis showed that the plant methanolic extract induced dose-dependent apoptosis and activated proapoptotic proteins caspase-3 and PARP1. Additionally, the extract triggered the arrest of the G0/G1 phase of the cell cycle and upregulated the protein expression of p27/Kip and p21/Cip, with a decrease in cyclin D1 expression in hepatocellular carcinoma cells. NOVELTY AND SCIENTIFIC CONTRIBUTION The current study demonstrates that T. articulata methanolic extract exhibits promising anticancer potential to kill tumour cells by programmed cell death type I and II mechanisms and could be explored for potential drug candidate molecules to curtail cancer in the future.
Collapse
Affiliation(s)
| | - Bilal Rah
- Corresponding authors: Phone: +966558764066, +966506166275, E-mail: ,
| |
Collapse
|
21
|
Lee HK, Park SH, Nam MJ. Proteasome inhibitor MG132 induces apoptosis in human osteosarcoma U2OS cells. Hum Exp Toxicol 2021; 40:1985-1997. [PMID: 34002651 DOI: 10.1177/09603271211017972] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
MG132 is a potent, reversible, and cell-permeable 20S proteasome inhibitor and it is derived from a Chinese medicinal plant. The purpose of this study is to investigate the anticancer effects of MG132 against human osteosarcoma U2OS cells. We first performed MTT and colony formation assays to investigate the anti-proliferative effects of MG132. The results demonstrated that MG132 suppressed the proliferation of U2OS cells. Furthermore, we found that treatment with MG132 increased apoptosis and induced DNA damage in U2OS cells. Additionally, zymography, wound healing, and invasion assays showed that MG132 suppressed the enzymatic activity of matrix metalloproteinases, cell migration, and invasion, respectively of U2OS cells. Furthermore, western blotting assay was performed to investigate the apoptotic signaling pathways in MG132-treated U2OS cells. Our results showed that MG132 downregulated the expression of antiapoptotic proteins, including CDK2, CDK4, Bcl-xL, and Bcl-2, whereas it upregulated the expression of proapoptotic proteins, including p21, p27, p53, p-p53 (ser15, ser20, and ser46), cleaved forms of caspase-3, caspase-7, caspase-9, and PARP, and FOXO3 in U2OS cells. These results demonstrated that MG132 activated apoptotic signaling pathways in U2OS cells. Interestingly, MG132 downregulated the phosphorylation of Akt and Erk. Taken together, our results suggest that MG132 has anticancer effects in U2OS cells. Therefore, MG132 may be a potential therapeutic agent for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Han Ki Lee
- Department of Biological Science, 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 Jin Nam
- Department of Biological Science, Gachon University, Seongnam-si, Gyeonggi-do, Republic of Korea
| |
Collapse
|
22
|
Choi YJ, Lee J, Ha SH, Lee HK, Lim HM, Yu SH, Lee CM, Nam MJ, Yang YH, Park K, Choi YS, Jang KY, Park SH. 6,8-Diprenylorobol induces apoptosis in human colon cancer cells via activation of intracellular reactive oxygen species and p53. ENVIRONMENTAL TOXICOLOGY 2021; 36:914-925. [PMID: 33382531 DOI: 10.1002/tox.23093] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 12/19/2020] [Indexed: 06/12/2023]
Abstract
6,8-Diprenylorobol is a natural compound mainly found in Glycyrrhiza uralensis fisch and Maclura tricuspidata, which has been used traditionally as food and medicine in Asia. So far, the antiproliferative effect of 6,8-diprenylorobol has not been studied yet in colon cancer. In this study, we aimed to evaluate the antiproliferative effects of 6,8-diprenylorobol in LoVo and HCT15, two kinds of human colon cancer cells. 6,8-Diprenylorobol inhibited the proliferation of LoVo and HCT15 cells in a dose- and time-dependent manner. A 40 μM of 6,8-diprenylorobol for 72 h reduced both of cell viability under 50%. After treatment of 6,8-diprenylorobol (40 and 60 μM) for 72 h, late apoptotic cell portion in LoVo and HCT15 cells were 24, 70% and 13, 90%, respectively, which was confirmed by checking DNA fragmentation in both cells. Mechanistically, 6,8-diprenylorobol activated p53 and its phosphorylated form (Ser15, Ser20, and Ser46) expression but suppressed Akt and mitogen-activated protein kinases (MAPKs) phosphorylation in LoVo and HCT15 cells. Interestingly, 6,8-diprenylorobol induced the generation of intracellular reactive oxygen species (ROS), which was attenuated with N-acetyl cysteine (NAC) treatment. Compared to the control, 60 μM of 6,8-diprenylorobol caused to increase ROS level to 210% in LoVo and HCT15, which was reduced into 161% and 124%, respectively with NAC. Furthermore, cell viability and apoptotic cell portion by 6,8-diprenylorobol was recovered by incubation with NAC. Taken together, these results indicate that 6,8-diprenylorobol has the potential antiproliferative effect against LoVo and HCT15 colon cancer cells through activation of p53 and generation of ROS.
Collapse
Affiliation(s)
- Yong Jun Choi
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Jongsung Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, South Korea
| | - Sang Hoon Ha
- Division of Biotechnology, Jeonbuk National University, Iksan, South Korea
| | - Han Ki Lee
- Department of Biological Science, Gachon University, Seongnam, South Korea
| | - Heui Min Lim
- Department of Biological Science, Gachon University, Seongnam, South Korea
| | - Seon-Hak Yu
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Chang Min Lee
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Myeong Jin Nam
- Department of Biological Science, Gachon University, Seongnam, South Korea
| | - Yung-Hun Yang
- Department of Biological Engineering, Konkuk University, Seoul, South Korea
| | - Kyungmoon Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| | - Youn Soo Choi
- Department of Biomedical Sciences, Seoul National University, Graduate School, Seoul, South Korea
- Department of Medicine, College of Medicine, Seoul National University, Seoul, South Korea
| | - Kyu Yun Jang
- Department of Pathology, Jeonbuk National University Medical School, Jeonju, South Korea
- Research Institute of Clinical Medicine of Jeonbuk National University, Jeonju, South Korea
- Biomedical Research Institute of Jeonbuk National University Hospital, Jeonju, South Korea
| | - See-Hyoung Park
- Department of Bio and Chemical Engineering, Hongik University, Sejong, South Korea
| |
Collapse
|
23
|
Türk E, Ozan Tekeli I, Özkan H, Uyar A, Cellat M, Kuzu M, Yavas I, Alizadeh Yegani A, Yaman T, Güvenç M. The protective effect of esculetin against aluminium chloride-induced reproductive toxicity in rats. Andrologia 2021; 53:e13930. [PMID: 33368464 DOI: 10.1111/and.13930] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Revised: 11/17/2020] [Accepted: 11/18/2020] [Indexed: 11/28/2022] Open
Abstract
One of the prominent health problems caused by Aluminium was the decrease in male fertility rates. In the study, the protective effect of Esculetin (ESC) against the reproductive toxicity induced by Aluminium chloride (AlCl3 ) was investigated. For this purpose, AlCl3 was administrated to Wistar Albino rats at a dose of 34 mg/kg and ESC was administrated at a dose of 50 mg/kg for 70 days. It was determined that AlCl3 treatment reduced sperm motility and concentration, increased dead/live rate and abnormal sperm rate. It decreased serum testosterone level, and co-treatment of ESC significantly regulated these values. In the AlCl3 -treated group, MDA level increased and GSH level, GPx and CAT activities decreased compared with those of the control group. However, co-treatment of ESC showed an amelioratory effect on the values except for CAT activity. It was observed that the expression level of NRF-2 increased in the ESC and AlCl3 + ESC groups, and NF-κB increased in the AlCl3 group with the control group. It was determined that Caspase-3 expression decreased, and Bcl-2 expression increased in AlCl3 + ESC group compared to AlCl3 group. It was also determined that AlCl3 -induced tissue injury was significantly prevented by ESC co-treatment.
Collapse
Affiliation(s)
- Erdinç Türk
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Mustafa Kemal University, Antakya-Hatay, Turkey
| | - Ibrahim Ozan Tekeli
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Mustafa Kemal University, Antakya-Hatay, Turkey
| | - Hüseyin Özkan
- Department of Genetics, Faculty of Veterinary Medicine, Mustafa Kemal University, Antakya-Hatay, Turkey
| | - Ahmet Uyar
- Department of Pathology, Faculty of Veterinary Medicine, Mustafa Kemal University, Antakya-Hatay, Turkey
| | - Mustafa Cellat
- Department of Physiology, Faculty of Veterinary Medicine, Mustafa Kemal University Antakya-Hatay, Turkey
| | - Müslüm Kuzu
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Karabuk University, Karabuk, Turkey
| | - Ilker Yavas
- Department of Reproduction and Artificial Insemination, Faculty of Veterinary Medicine, University of Mustafa Kemal, Antakya-Hatay, Turkey
| | - Arash Alizadeh Yegani
- Department of Pharmacology and Toxicology, Faculty of Veterinary Medicine, Mustafa Kemal University, Antakya-Hatay, Turkey
| | - Turan Yaman
- Department of Pathology, Faculty of Veterinary Medicine, Yuzuncu Yil University, Van, Turkey
| | - Mehmet Güvenç
- Department of Physiology, Faculty of Veterinary Medicine, Mustafa Kemal University Antakya-Hatay, Turkey
| |
Collapse
|
24
|
Antitumor Effect of the Ethanolic Extract from Seeds of Euphorbia lathyris in Colorectal Cancer. Nutrients 2021; 13:nu13020566. [PMID: 33572111 PMCID: PMC7915714 DOI: 10.3390/nu13020566] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 02/01/2021] [Accepted: 02/05/2021] [Indexed: 01/01/2023] Open
Abstract
The seeds of Euphorbia lathyris have been used in traditional medicine to treat various medical conditions. However, neither all of their active biocompounds nor the molecular mechanisms underlying their therapeutic effects have been described. A new ethanolic extract of defatted flour from mature seeds of Euphorbia lathyris showed a high total polyphenol content and significant antioxidant activity. Chromatographic analysis showed that esculetin, euphorbetin, gaultherin, and kaempferol-3-rutinoside were the most abundant polyphenolic bioactive compounds. Antiproliferative assays showed a high and selective antitumor activity against colon cancer cell lines (T84 and HCT-15). In addition, a significant antiproliferative activity against glioblastoma multiforme cells was also demonstrated. Its mechanism of action to induce cell death was mediated by the overexpression of caspases 9, 3, and 8, and by activation of autophagy. Interestingly, a reduction in the migration capacity of colon cancer cells and a significant antiangiogenic effect on human umbilical vein endothelial cells were also demonstrated. Finally, the extract significantly reduced the subpopulations of cancer stem cells. This extract could be the basis to develop new therapeutic strategies for the treatment of colon cancer, although further experiments will be necessary to determine its in vivo effects.
Collapse
|
25
|
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
|
26
|
Ren S, Xing Y, Wang C, Jiang F, Liu G, Li Z, Jiang T, Zhu Y, Piao D. Fraxetin inhibits the growth of colon adenocarcinoma cells via the Janus kinase 2/signal transducer and activator of transcription 3 signalling pathway. Int J Biochem Cell Biol 2020; 125:105777. [PMID: 32504672 DOI: 10.1016/j.biocel.2020.105777] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Revised: 05/31/2020] [Accepted: 06/01/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Fraxetin, extracted from the bark of Fraxinus rhynchophylla, has been shown to exhibit antitumour and anti-inflammatory pharmacological properties. However, the mechanism underlying its anticancer activity towards colon adenocarcinoma (COAD) is not well understood. We aimed to determine the antitumour effect of fraxetin on COAD cell lines and elucidate its biochemical and molecular targets. METHODS The cell lines HCT116 and DLD-1 were used to evaluate the in vitro antitumour efficacy of fraxetin. Cytotoxicity and viability were assessed by CCK-8 and plate colony formation assays. Flow cytometry was used to assess apoptosis and cell cycle progression in fraxetin-treated COAD cells. Western blot, RT-qPCR, molecular docking, immunohistochemical, and immunofluorescence analyses were used to gain insights into cellular and molecular mechanisms. Preclinical curative effects were evaluated in nude mouse xenograft models. RESULTS Fraxetin significantly inhibited COAD cell proliferation in both dose- and time-dependent manners, specifically by inducing S-phase cell cycle arrest and triggering intrinsic apoptosis. Additionally, the level of p-JAK2 was decreased by fraxetin via the Janus kinase 2/signal transducer and activator of transcription 3 (JAK2/STAT3) signalling pathway. Interestingly, in COAD cells, fraxetin directly targeted the Y1007 and Y1008 residues of JAK2 to suppress its auto- or transphosphorylation, leading to decreased activation of its downstream effector STAT3 and blocking its nuclear translocation. Finally, fraxetin exhibited good tumour growth suppression activity and low toxicity. CONCLUSIONS Fraxetin inhibits the proliferation of COAD cells by regulating the JAK2/STAT3 signalling pathway, providing evidence that targeting JAK2 with fraxetin may offer a novel potential auxiliary therapy for COAD treatment.
Collapse
Affiliation(s)
- Shuo Ren
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yanwei Xing
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Chengbo Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Fengqi Jiang
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Guangyu Liu
- Department of Anorectal Surgery, The Shenzhen Hospital of Southern Medical University, Southern Medical University, Shenzhen, China
| | - Ziyi Li
- The Hepatosplenic Surgery Center, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Tao Jiang
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Yuekun Zhu
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Daxun Piao
- Department of Colorectal Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China.
| |
Collapse
|
27
|
Huang SX, Mou JF, Luo Q, Mo QH, Zhou XL, Huang X, Xu Q, Tan XD, Chen X, Liang CQ. Anti-Hepatitis B Virus Activity of Esculetin from Microsorium fortunei In Vitro and In Vivo. Molecules 2019; 24:E3475. [PMID: 31557836 PMCID: PMC6803987 DOI: 10.3390/molecules24193475] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/14/2019] [Accepted: 09/24/2019] [Indexed: 02/07/2023] Open
Abstract
Coumarins are widely present in a variety of plants and have a variety of pharmacological activities. In this study, we isolated a coumarin compound from Microsorium fortunei (Moore) Ching; the compound was identified as esculetin by hydrogen and carbon spectroscopy. Its anti-hepatitis B virus (HBV) activity was investigated in vitro and in vivo. In the human hepatocellular liver carcinoma 2.2.15 cell line (HepG2.2.15) transfected with HBV, esculetin effecting inhibited the expression of the HBV antigens and HBV DNA in vitro. Esculetin inhibited the expression of Hepatitis B virus X (HBx) protein in a dose-dependent manner. In the ducklings infected with duck hepatitis B virus (DHBV), the levels of DHBV DNA, duck hepatitis B surface antigen (DHBsAg), duck hepatitis B e-antigen (DHBeAg), alanine aminotransferase (ALT) and aspartate aminotransferase (AST) decreased significantly after esculetin treatment. Summing up the above, the results suggest that esculetin efficiently inhibits HBV replication both in vitro and in vivo, which provides an opportunity for further development of esculetin as antiviral drug.
Collapse
Affiliation(s)
- Si-Xin Huang
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Jun-Fei Mou
- Biotechnology Institute, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Qin Luo
- Science Experiment Center, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Qing-Hu Mo
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Xian-Li Zhou
- Biotechnology Institute, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Xiao Huang
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Qing Xu
- Guangxi Key Laboratory of Molecular Medicine in Liver Injury and Repair, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Xiang-Duan Tan
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Xu Chen
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China.
| | - Cheng-Qin Liang
- College of Pharmacy, Guilin Medical University, Guilin 541004, Guangxi, China.
| |
Collapse
|