1
|
Yu L, Gu X, Chen P, Yang R, Xu Y, Yang X. Effects of PTPN6 Gene Knockdown in SKM-1 Cells on Apoptosis, Erythroid Differentiation and Inflammations. Curr Issues Mol Biol 2024; 46:12061-12074. [PMID: 39590309 PMCID: PMC11593023 DOI: 10.3390/cimb46110715] [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: 08/20/2024] [Revised: 10/06/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Objective: Protein tyrosine phosphatase non-receptor type 6 (PTPN6) is a cytoplasmic phosphatase that acts as a key regulatory protein in cell signaling to control inflammation and cell death. In order to investigate the role of PTPN6 in hematologic tumor myelodysplastic syndrome (MDS), this study infected SKM-1 cell line (MDS cell line) with packaged H_PTPN6-shRNA lentivirus to obtain H_PTPN6-shRNA SKM-1 stable strain. The effect of PTPN6 knockdown on apoptosis, erythroid differentiation, and inflammations in SKM-1 cell line was examined. Methods: The stable knockdown SKM-1 cell line was validated using qPCR and Western blot assays. The proliferation activity, apoptosi, erythroid differentiation, and inflammatory cytokines in SKM-1 cells were assessed before and after transfection. Results: qPCR confirmed that the expression level of H_PTPN6-shRNA in SKM-1 cells was significantly reduced, and Western blot showed that the protein expression level of H_PTPN6-shRNA in SKM-1 cells was also significantly reduced. The CCK-8 cell viability assay confirmed that stable gene knockdown did not affect cell viability. Flow cytometry revealed that the apoptosis rate of cells in the PTPN6 knockdown group was 0.8%, lower than the 2.7% observed in the empty plasmid group; the expression rate of the erythroid differentiation marker CD235a was 13.2%, lower than the 25.0% observed in the empty plasmid group. The expression levels of the proinflammatory factors IL-6 and IL-8 increased, and the expression levels of the inhibitor factor IL-4 decreased. Conclusions: The PTPN6 gene was successfully knocked down using lentivirus-mediated transduction, and the constructed cell line was validated using PCR and Western blot. The CCK-8 cell viability assay confirmed that stable gene knockdown did not affect cell proliferation viability. Flow cytometry analysis of apoptosis and erythroid differentiation indicated that PTPN6 knockdown inhibits apoptosis and erythroid differentiation in SKM-1 cells and also alters the level of inflammations in the bone marrow microenvironment. It suggests that the PTPN6 gene acts as a tumor suppressor in myelodysplastic syndrome cells, influencing hematopoietic cell apoptosis, erythroid differentiation, and inflammations. This provides a reliable experimental basis for further in-depth studies on the mechanism of PTPN6 in MDS and related pharmacological research.
Collapse
Affiliation(s)
| | | | | | | | - Yonggang Xu
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (L.Y.); (X.G.); (P.C.); (R.Y.)
| | - Xiupeng Yang
- Department of Hematology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing 100091, China; (L.Y.); (X.G.); (P.C.); (R.Y.)
| |
Collapse
|
2
|
Tao Z, Bai S, Wu G, Zhai S, Zhang P, Fu C, Yu L. Therapeutic effect of ginkgetin on smoke-induced airway inflammation by down-regulating the c/EBPβ signaling pathway and CCL2 expression. JOURNAL OF ETHNOPHARMACOLOGY 2024; 331:118284. [PMID: 38735420 DOI: 10.1016/j.jep.2024.118284] [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: 03/11/2024] [Revised: 04/26/2024] [Accepted: 05/01/2024] [Indexed: 05/14/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginkgo biloba leaf and seed have been traditionally used in ancient China for the treatment of cough and asthma. However, there is limited literature available on the anti-COPD effects and mechanisms of Ginkgo biloba. AIMS OF THE STUDY The aim of this study was to comprehensively investigate the therapeutic potential of ginkgo extracts in COPD through a combination of in vivo and in vitro functional experiments. Transcriptomic analyses were also employed to uncover novel molecular mechanisms underlying the therapeutic effects of ginkgetin in COPD. MATERIALS AND METHODS The therapeutic efficacy of ginkgo extracts was assessed in a COPD model. The anti-inflammatory effects of ginkgetin and its underlying molecular mechanisms were examined in A549 cells treated with cigarette smoke extract (CSE). Additionally, transcriptomic analyses were conducted to identify novel molecular pathways influenced by ginkgetin. These findings were further validated using quantitative real-time polymerase chain reaction (qPCR) and Western blot techniques. RESULTS The ethyl acetate extract of Ginkgo biloba L. seeds and ginkgetin treatment significantly reduced cytokine production in COPD mice. Following drug administration, lung function improved in different groups. The transcriptome data strongly supports the inhibitory effect of ginkgetin on CSE-induced inflammation through the downregulation of the c/EBPβ signaling pathway and subsequent inhibition of CCL2 expression. CONCLUSION Our results demonstrate that ginkgetin, one of the biflavones found in Ginkgo biloba, exhibits inhibitory effects on smoke-induced airway inflammation. This effect is achieved through the downregulation of the c/EBPβ signaling pathway and the reduction of CCL2 expression.
Collapse
Affiliation(s)
- Zhu Tao
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; College of Life Science and Technology, Hubei Engineering University, Xiaogan 432000, China; Hubei Engineering Research Centre for Dual-use Resource Development of Food and Medicine, Wuhan 430074, China
| | - Shaoliang Bai
- Wuhan Aimin Pharmaceutical Co., Ltd Ezhou 436032, China
| | - Guodong Wu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shengbing Zhai
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Pei Zhang
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chunhua Fu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Engineering Research Centre for Dual-use Resource Development of Food and Medicine, Wuhan 430074, China.
| | - Longjiang Yu
- Institute of Resource Biology and Biotechnology, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Engineering Research Centre for Dual-use Resource Development of Food and Medicine, Wuhan 430074, China.
| |
Collapse
|
3
|
Liu H, Sun L, Zhao H, Zhao Z, Zhang S, Jiang S, Cheng T, Wang X, Wang T, Shao Y, Zhu H, Han H, Cao Y, Jiang E, Cao Y, Xu Y. Proteinase 3 depletion attenuates leukemia by promoting myeloid differentiation. Cell Death Differ 2024; 31:697-710. [PMID: 38589495 PMCID: PMC11165011 DOI: 10.1038/s41418-024-01288-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Revised: 03/19/2024] [Accepted: 03/22/2024] [Indexed: 04/10/2024] Open
Abstract
Hematopoietic stem and progenitor cells (HSPCs) that have impaired differentiation can transform into leukemic blasts. However, the mechanism that controls differentiation remains elusive. Here, we show that the genetic elimination of Proteinase 3 (PRTN3) in mice led to spontaneous myeloid differentiation. Mechanistically, our findings indicate that PRTN3 interacts with the N-terminal of STAT3, serving as a negative regulator of STAT3-dependent myeloid differentiation. Specifically, PRTN3 promotes STAT3 ubiquitination and degradation, while simultaneously reducing STAT3 phosphorylation and nuclear translocation during G-CSF-stimulated myeloid differentiation. Strikingly, pharmacological inhibition of STAT3 (Stattic) partially counteracted the effects of PRTN3 deficiency on myeloid differentiation. Moreover, the deficiency of PRTN3 in primary AML blasts promotes the differentiation of those cells into functional neutrophils capable of chemotaxis and phagocytosis, ultimately resulting in improved overall survival rates for recipients. These findings indicate PRTN3 exerts an inhibitory effect on STAT3-dependent myeloid differentiation and could be a promising therapeutic target for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Huan Liu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Lu Sun
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Hongfei Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Zihan Zhao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shiyue Zhang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Shan Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Clinical Laboratory, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Tianran Cheng
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Xiaohan Wang
- The Second School of Clinical Medicine, Guangdong Medical University, Dongguan, 523808, China
| | - Tong Wang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Ya Shao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
| | - Haiyan Zhu
- Department of Clinical Lab, Weihai Municipal Hospital, Weihai, 264200, China
| | - Huijuan Han
- Department of Medical Laboratory, School of Clinical Medicine, Ningxia Medical University; Ningxia Key Laboratory of Clinical and Pathogenic Microbiology, General Hospital of Ningxia Medical University, Yinchuan, 750001, China
| | - Yigeng Cao
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Erlie Jiang
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China
- Tianjin Institutes of Health Science, Tianjin, 301600, China
- Hematopoietic Stem Cell Transplantation Center, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, 300020, China
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Solna, 17165, Sweden.
| | - Yuanfu Xu
- State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, 300020, China.
- Tianjin Institutes of Health Science, Tianjin, 301600, China.
| |
Collapse
|
4
|
Hu D, Wang HJ, Yu LH, Guan ZR, Jiang YP, Hu JH, Yan YX, Zhou ZH, Lou JS. The role of Ginkgo Folium on antitumor: Bioactive constituents and the potential mechanism. JOURNAL OF ETHNOPHARMACOLOGY 2024; 321:117202. [PMID: 37742878 DOI: 10.1016/j.jep.2023.117202] [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: 07/17/2023] [Revised: 09/16/2023] [Accepted: 09/16/2023] [Indexed: 09/26/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Ginkgo biloba L. is a well-known and highly regarded resource in Chinese traditional medicine due to its effectiveness and safety. Ginkgo Folium, the leaf of Ginkgo biloba L., contains biologically active constituents with diverse pharmacological activities. Recent studies have shown promising antitumor effects of the bioactive constituents found in Ginkgo Folium against various types of cancer cells, highlighting its potential as a natural source of antitumor agents. Further research is needed to elucidate the underlying mechanisms and optimize its therapeutic potential. AIM OF THE REVIEW To provide a detailed understanding of the pharmacological activities of Ginkgo Folium and its potential therapeutic benefits for cancer patients. MATERIALS AND METHODS In this study, we conducted a thorough and systematic search of multiple online databases, including PubMed, Web of Science, Medline, using relevant keywords such as "Ginkgo Folium," "flavonoids," "terpenoids," "Ginkgo Folium extracts," and "antitumor" to cover a broad range of studies that could inform our review. Additionally, we followed a rigorous selection process to ensure that the studies included in our review met the predetermined inclusion criteria. RESULTS The active constituents of Ginkgo Folium primarily consist of flavonoids and terpenoids, with quercetin, kaempferol, isorhamnetin, ginkgolides, and bilobalide being the major compounds. These active constituents exert their antitumor effects through crucial biological events such as apoptosis, cell cycle arrest, autophagy, and inhibition of invasion and metastasis via modulating diverse signaling pathways. During the process of apoptosis, active constituents primarily exert their effects by modulating the caspase-8 mediated death receptor pathway and caspase-9 mediated mitochondrial pathway via regulating specific signaling pathways. Furthermore, by modulating multiple signaling pathways, active constituents effectively induce G1, G0/G1, G2, and G2/M phase arrest. Among these, the pathways associated with G2/M phase arrest are particularly extensive, with the cyclin-dependent kinases (CDKs) being most involved. Moreover, active constituents primarily mediate autophagy by modulating certain inflammatory factors and stressors, facilitating the fusion stage between autophagosomes and lysosomes. Additionally, through the modulation of specific chemokines and matrix metalloproteinases, active constituents effectively inhibit the processes of epithelial-mesenchymal transition (EMT) and angiogenesis, exerting a significant impact on cellular invasion and migration. Synergistic effects are observed among the active constituents, particularly quercetin and kaempferol. CONCLUSION Active components derived from Ginkgo Folium demonstrate a comprehensive antitumor effect across various levels and pathways, presenting compelling evidence for their potential in new drug development. However, in order to facilitate their broad and adaptable clinical application, further extensive experimental investigations are required to thoroughly explore their efficacy, safety, and underlying mechanisms of action.
Collapse
Affiliation(s)
- Die Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Hao-Jie Wang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Li-Hua Yu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zheng-Rong Guan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Ya-Ping Jiang
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jun-Hu Hu
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Ya-Xin Yan
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Zhao-Huang Zhou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China
| | - Jian-Shu Lou
- School of Pharmacy, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines, Engineering Laboratory of Development and Application of Traditional Chinese Medicines, Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang, 311121, China.
| |
Collapse
|
5
|
Wang C, Bai Y, Li T, Liu J, Wang Y, Ju S, Yao W, Xiong B. Ginkgetin exhibits antifibrotic effects by inducing hepatic stellate cell apoptosis via STAT1 activation. Phytother Res 2024; 38:1367-1380. [PMID: 38217097 DOI: 10.1002/ptr.8106] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 12/12/2023] [Accepted: 12/16/2023] [Indexed: 01/15/2024]
Abstract
Liver fibrosis affects approximately 800 million patients worldwide, with over 2 million deaths each year. Nevertheless, there are no approved medications for treating liver fibrosis. In this study, we investigated the impacts of ginkgetin on liver fibrosis and the underlying mechanisms. The impacts of ginkgetin on liver fibrosis were assessed in mouse models induced by thioacetamide or bile duct ligation. Experiments on human LX-2 cells and primary mouse hepatic stellate cells (HSCs) were performed to explore the underlying mechanisms, which were also validated in the mouse models. Ginkgetin significantly decreased hepatic extracellular matrix deposition and HSC activation in the fibrotic models induced by thioacetamide (TAA) and bile duct ligation (BDL). Beneficial effects also existed in inhibiting hepatic inflammation and improving liver function. In vitro experiments showed that ginkgetin markedly inhibited HSC viability and induced HSC apoptosis dose-dependently. Mechanistic studies revealed that the antifibrotic effects of ginkgetin depend on STAT1 activation, as the effects were abolished in vitro after STAT1 silencing and in vivo after inhibiting STAT1 activation by fludarabine. Moreover, we observed a meaningful cross-talk between HSCs and hepatocytes, in which IL-6, released by ginkgetin-induced apoptotic HSCs, enhanced hepatocyte proliferation by activating STAT3 signaling. Ginkgetin exhibits antifibrotic effects by inducing HSC apoptosis via STAT1 activation and enhances hepatocyte proliferation secondary to HSC apoptosis via the IL-6/STAT3 pathway.
Collapse
Affiliation(s)
- Chaoyang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tongqiang Li
- Department of Interventional Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Jiacheng Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yingliang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shuguang Ju
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Yao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bin Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Interventional Radiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
6
|
Ahn CR, Baek SH. Enhancing Gastric Cancer Therapeutic Efficacy through Synergistic Cotreatment of Linderae Radix and Hyperthermia in AGS Cells. Biomedicines 2023; 11:2710. [PMID: 37893084 PMCID: PMC10604735 DOI: 10.3390/biomedicines11102710] [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: 06/26/2023] [Revised: 09/21/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Gastric cancer remains a global health threat, particularly in Asian countries. Current treatment methods include surgery, chemotherapy, and radiation therapy. However, they all have limitations, such as adverse side effects, tumor resistance, and patient tolerance. Hyperthermia therapy uses heat to selectively target and destroy cancer cells, but it has limited efficacy when used alone. Linderae Radix (LR), a natural compound with thermogenic effects, has the potential to enhance the therapeutic efficacy of hyperthermia treatment. In this study, we investigated the synergistic anticancer effects of cotreatment with LR and 43 °C hyperthermia in AGS gastric cancer cells. The cotreatment inhibited AGS cell proliferation, induced apoptosis, caused cell cycle arrest, suppressed heat-induced heat shock responses, increased reactive oxygen species (ROS) generation, and promoted mitogen-activated protein kinase phosphorylation. N-acetylcysteine pretreatment abolished the apoptotic effect of LR and hyperthermia cotreatment, indicating the crucial role of ROS in mediating the observed anticancer effects. These findings highlight the potential of LR as an adjuvant to hyperthermia therapy for gastric cancer. Further research is needed to validate these findings in vivo, explore the underlying molecular pathways, and optimize treatment protocols for the development of novel and effective therapeutic strategies for patients with gastric cancer.
Collapse
Affiliation(s)
- Chae-Ryeong Ahn
- Department of Science in Korean Medicine, Graduate School, Kyung Hee University, Seoul 02447, Republic of Korea;
| | - Seung-Ho Baek
- College of Korean Medicine, Dongguk University, 32 Dongguk-ro, Goyang-si 10326, Republic of Korea
| |
Collapse
|
7
|
Zhang S, Sun Y, Yao F, Li H, Yang Y, Li X, Bai Z, Hu Y, Wang P, Xu X. Ginkgo Biflavones Cause p53 Wild-Type Dependent Cell Death in a Transcription-Independent Manner of p53. JOURNAL OF NATURAL PRODUCTS 2023; 86:346-356. [PMID: 36700552 DOI: 10.1021/acs.jnatprod.2c00959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Ginkgo biloba, as a medicinal plant in both traditional and western medicine, emerged as a potential therapeutic agent for the management of a variety of diseases, but ginkgo biflavones (bilobetin, isoginkgetin, and ginkgetin) application in cancer therapy and underlying mechanisms of action remained elusive. In the present study, we identified ginkgo biflavones as potential p53 activators that could enhance p53 protein expression level by inhibiting MDM2 protein expression. At the same time, they induced cell death independent of p53 transcriptional activity. Moreover, ginkgetin was a standout among ginkgo biflavones that reduced the survival of HCT-116 cells by induction of apoptosis and G2/M phase arrest. Furthermore, ginkgo biflavones induced ROS generation significantly, which resulted in ferroptosis. Finally, we provide evidence that ginkgetin strengthened the antitumor effect of fluorouracil (5-FU) in the HCT-116 colon cancer xenograft model. To sum up, ginkgo biflavones represent a new class of p53 activator that depends on the p53 wild-type status and warrants further exploration as potential anticancer agents.
Collapse
Affiliation(s)
- Siyu Zhang
- Institute of Burns, Tongren Hospital of Wuhan University (Wuhan Third Hospital), Wuhan 430060, P. R. China
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
| | - Yujie Sun
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, Shandong, P. R. China
| | - Fengli Yao
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- College of Food Science and Engineering, Ocean University of China, Qingdao 266071, P. R. China
| | - Hongju Li
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- College of Food Science and Engineering, Ocean University of China, Qingdao 266071, P. R. China
| | - Yacong Yang
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, Shandong, P. R. China
| | - Xionghao Li
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, Shandong, P. R. China
| | - Zhongyue Bai
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, Shandong, P. R. China
| | - Yu Hu
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, Shandong, P. R. China
| | - Peng Wang
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- College of Food Science and Engineering, Ocean University of China, Qingdao 266071, P. R. China
| | - Ximing Xu
- Center for Innovation Marine Drug Screening & Evaluation, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266071, P. R. China
- Marine Drug Screening and Evaluation Platform (QNLM), School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, P. R. China
- School of Medicine and Pharmacy, Ocean University of China, Qingdao 266071, Shandong, P. R. China
| |
Collapse
|
8
|
Tian Y, Liu H, Wang M, Wang R, Yi G, Zhang M, Chen R. Role of STAT3 and NRF2 in Tumors: Potential Targets for Antitumor Therapy. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248768. [PMID: 36557902 PMCID: PMC9781355 DOI: 10.3390/molecules27248768] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/02/2022] [Accepted: 12/08/2022] [Indexed: 12/14/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) and nuclear factor erythroid-derived 2-like 2 (NRF2, also known as NFE2L2), are two of the most complicated transcription regulators, which participate in a variety of physiological processes. Numerous studies have shown that they are overactivated in multiple types of tumors. Interestingly, STAT3 and NRF2 can also interact with each other to regulate tumor progression. Hence, these two important transcription factors are considered key targets for developing a new class of antitumor drugs. This review summarizes the pivotal roles of the two transcription regulators and their interactions in the tumor microenvironment to identify potential antitumor drug targets and, ultimately, improve patients' health and survival.
Collapse
Affiliation(s)
- Yanjun Tian
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining 272067, China
| | - Haiqing Liu
- Department of Physiology, School of Basic Medical Sciences (Institute of Basic Medical Sciences), Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250024, China
| | - Mengwei Wang
- School of Stomatology, Jining Medical University, Jining 272067, China
| | - Ruihao Wang
- School of Mental Health, Jining Medical University, Jining 272067, China
| | - Guandong Yi
- School of Nursing, Jining Medical University, Jining 272067, China
| | - Meng Zhang
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining 272067, China
| | - Ruijiao Chen
- Medical Laboratory of Jining Medical University, Jining Medical University, Jining 272067, China
- Correspondence: ; Tel.: +86-537-361-6216
| |
Collapse
|
9
|
Yang MH, Baek SH, Hwang ST, Um JY, Ahn KS. Corilagin exhibits differential anticancer effects through the modulation of STAT3/5 and MAPKs in human gastric cancer cells. Phytother Res 2022; 36:2449-2462. [PMID: 35234310 DOI: 10.1002/ptr.7419] [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: 06/15/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/24/2022]
Abstract
Corilagin (CLG) is a hydrolyzable tannin and possesses various pharmacological activities. Here, we investigated the impact of CLG as an anti-tumor agent against human gastric tumor cells. We observed that CLG could cause negative regulation of JAKs-Src-STAT3/5 signaling axis in SNU-1 cells, but did not affect these pathways in SNU-16 cells. Interestingly, CLG promoted the induction of mitogen-activated protein kinases (MAPKs) signaling pathways in only SNU-16 cells, but not in the SNU-1 cells. CLG exhibited apoptotic effects that caused an increased accumulation of the cells in sub-G1 phase and caspase-3 activation in both SNU-1 and SNU-16 cell lines. We also noticed that CLG and docetaxel co-treatment could exhibit significantly enhanced apoptotic effects against SNU-1 cells. Moreover, the combinations treatment of CLG and docetaxel markedly inhibited cell growth, phosphorylation of JAK-Src-STAT3 and induced substantial apoptosis. Additionally, pharmacological inhibition of JNK, p38, and ERK substantially blocked CLG-induced activation of MAPKs, cell viability, and apoptosis, thereby implicating the pivotal role of MAPKs in the observed anti-cancer effects of CLG. Taken together, our data suggest that CLG could effectively block constitutive STAT3/5 activation in SNU-1 cells but induce sustained MAPKs activation in SNU-16 cells.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea.,Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Seung Ho Baek
- College of Korean Medicine, Dongguk University, Goyang-si, South Korea
| | - Sun Tae Hwang
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea.,Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
10
|
Liu Q, Chen L, Yin W, Nie Y, Zeng P, Yang X. Anti-tumor effect of ginkgetin on human hepatocellular carcinoma cell lines by inducing cell cycle arrest and promoting cell apoptosis. Cell Cycle 2021; 21:74-85. [PMID: 34878966 DOI: 10.1080/15384101.2021.1995684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
This study explored the anti-tumor effect of ginkgetin, an extract from ginkgo biloba, on human hepatocellular carcinoma cell lines and the underlying mechanisms. Cell viability was measured by MTT assay. Apoptotic cell morphology was observed under an inverted microscope after Hoechst 33,258 staining, and the ratio of apoptotic and necrotic cells was examined by flow cytometry after FITC/PI staining. Cell cycle changes were analyzed using flow cytometry. Cytochrome c release and caspase 3 and 8 activities were monitored using the relevant reagent kits. The levels of cell cycle-related proteins were detected by Western blot. MTT results indicated that ginkgetin significantly reduced HepG2 cell viability in a dose-dependent manner. Cellular morphology observation revealed that ginkgetin induced typical apoptotic morphological features of HepG2 cells, such as increased apoptotic bodies and cell shrinkage. Cell cycle analysis showed that ginkgetin increased the proportion of cells in the S phase. S-phase cell accumulation could be attributed to the decreased expression of cell cycle regulatory factors. Similarly, ginkgetin also induced the apoptosis and S phase cell accumulation of another human HCC cell line SK-HEP-1. Furthermore, ginkgetin treatment increased caspase-3 activity and cytochrome c release but not caspase-8 activity, implying that ginkgetin might mediate cell apoptosis through the mitochondrial pathway. In addition, the tumor formation experiment in nude mice showed that ginkgetin administration inhibited tumor growth. These results suggest that ginkgetin could be a cell apoptosis stimulator by affecting the balance between cell proliferation and apoptosis, suggesting that ginkgetin might be suitable for human HCC treatment.
Collapse
Affiliation(s)
- Qiong Liu
- The First Affiliated Hospital, Department of Infectious Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Lingying Chen
- The First Affiliated Hospital, Department of blood transfusion, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Wenjun Yin
- The First Affiliated Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Yuehua Nie
- The First Affiliated Hospital, Department of Radiation Oncology, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Penghui Zeng
- The First Affiliated Hospital, Department of Clinical Laboratory, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xin Yang
- The First Affiliated Hospital, Department of Infectious Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
11
|
Yang MH, Ha IJ, Lee SG, Um JY, Ahn KS. Abrogation of STAT3 activation cascade by Ginkgolide C mitigates tumourigenesis in lung cancer preclinical model. J Pharm Pharmacol 2021; 73:1630-1642. [PMID: 34559878 DOI: 10.1093/jpp/rgab114] [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: 10/14/2020] [Accepted: 07/23/2021] [Indexed: 12/21/2022]
Abstract
OBJECTIVES Ginkgolide C (GGC) isolated from Ginkgo biloba (Ginkgoaceae) leaf can demonstrate pleiotropic pharmacological actions. However, its anti-oncogenic impact in non-small cell lung cancer (NSCLC) model has not been reconnoitered. As signal transducer and activator of transcription 3 (STAT3) cascade can promote tumour growth and survival, we contemplated that GGC may interrupt this signalling cascade to expend its anti-cancer actions in NSCLC. METHODS The effect of GGC on STAT3 activation, associated protein kinases, STAT3-regulated gene products, cellular proliferation and apoptosis was examined. The in-vivo effect of GGC on the growth of human NSCLC xenograft tumours in athymic nu/nu female mice was also investigated. KEY FINDINGS GGC attenuated the phosphorylation of STAT3 and STAT3 upstream kinases effectively. Exposure to pervanadate modulated GGC-induced down-regulation of STAT3 activation and promoted an elevation in the level of PTPε protein. Indeed, silencing of the PTPε gene reversed the GGC-promoted abrogation of STAT3 activation and apoptosis. Moreover, GGC exposure significantly reduced NSCLC tumour growth without demonstrating significant adverse effects via decreasing levels of p-STAT3 in mice tissues. CONCLUSIONS Overall, the findings support that GGC may exhibit anti-neoplastic actions by mitigation of STAT3 signalling cascade in NSCLC.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Seok-Geun Lee
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul, Republic of Korea
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
12
|
Li F, Li SS, Chen H, Zhao JZ, Hao J, Liu JM, Zu XG, Cui W. miR-320 accelerates chronic heart failure with cardiac fibrosis through activation of the IL6/STAT3 axis. Aging (Albany NY) 2021; 13:22516-22527. [PMID: 34582362 PMCID: PMC8507257 DOI: 10.18632/aging.203562] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2020] [Accepted: 08/24/2021] [Indexed: 12/12/2022]
Abstract
Cardiac fibrosis could induce abnormal cardiac function and become a novel target for cardiac hypertrophy and chronic heart failure. MiRNA-320 is a crucial miRNA in cardiovascular disease, but it is poorly understood whether it plays a role in cardiac fibrosis pathogenesis. We aimed to identify the specific underlying mechanism of miR-320 in cardiac fibrosis and hypertrophic pathogenesis. In our study, the GEO datasets revealed that STAT3 was significantly highly expressed in cardiomyocyte lines. MiR-320 activation and STAT3 signaling pathways were statistically significantly connected. Furthermore, miR-320 was highly associated with cardiac fibrosis and hypertrophic disease. Interstitial fibrosis was observed in the mice subjected to TAC surgery, markedly enhanced in miR-320 mimics. Mechanistically, we revealed that miR-320 mimics aggravated the pressure overload and induced cardiac hypertrophy and fibrosis via the IL6/STAT3/PTEN axis. MiR-320 mimics accelerated cardiac hypertrophy and cardiac fibrosis via the IL6/STAT3/PTEN axis. These results suggest that targeting miR-320 may represent a potential therapeutic strategy for cardiac hypertrophy and fibrosis.
Collapse
Affiliation(s)
- Fang Li
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Shan-Shan Li
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Hui Chen
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Jian-Zhi Zhao
- Department of Biochemistry and Molecular Biology, The Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Jie Hao
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Jin-Ming Liu
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Xiu-Guang Zu
- Third Division, Department of Cardiology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei 050011, PR China
| | - Wei Cui
- Department of Cardiology, The Second Hospital of Hebei Medical University and Hebei Institute of Cardiovascular Research, Shijiazhuang, Hebei 050011, PR China
| |
Collapse
|
13
|
Guan X, Zhang Y, Gareev I, Beylerli O, Li X, Lu G, Lv L, Hai X. MiR-499a prevents astrocytes mediated inflammation in ischemic stroke by targeting PTEN. Noncoding RNA Res 2021; 6:146-152. [PMID: 34632168 PMCID: PMC8488463 DOI: 10.1016/j.ncrna.2021.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 09/24/2021] [Accepted: 09/24/2021] [Indexed: 01/29/2023] Open
Abstract
INTRODUCTION Ischemic stroke (IS) is a common and severe neurological disorder and is associated with high rates of mortality and morbidity. Inflammatory reaction in astrocytes is one of the important pathological factors of stroke. Improved understanding of the underlying molecular mechanisms should aid better treatment of the disease. This study aimed to test our hypothesis that a miR-499a played an important role in the inflammatory response in astrocytes induced by IS targeting phosphatase and tensin homologue deleted on chromosome 10 (PTEN). METHODS This study was comprised of two models: oxygen-glucose deprivation (OGD) and reoxygenation model. Quantitative real-time PCR (qRT-PCR) and Western blot were used to examine gene expression levels, and MTT assay analysis were used to examine cell states. The relationships between miR-499a and PTEN were confirmed by luciferase reporter assay. RESULTS MiR-499a was robustly downregulated with OGD induced injury in astrocytes. Forced transient expression of miR-499a in OGD astrocytes nearly completely reversed the inflammatory response. Knockdown of miR-499a by its specific inhibitor in healthy astrocytes induced the inflammatory response resembling those produced by OGD. On the other hand, PTEN was markedly upregulated in OGD astrocytes, which was reciprocal to the expression of miR-499a. PTEN was experimentally validated as a direct target gene for miR-499a. Overexpression of PTEN was able to induce an inflammatory response of astrocytes. Moreover, PTEN siRNA counteracted the inflammatory response induced by OGD. CONCLUSIONS Taken together, our findings indicate miR-499a as an important factor to prevent inflammatory response and suggest miR-499a as a new molecule for the treatment of IS. The present study also demonstrated the relationship between miR-499a and PTEN, with PTEN as a downstream signaling mediator of miR-499a in the inflammatory response of astrocytes induced by IS.
Collapse
Affiliation(s)
- Xiaoxiang Guan
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Yiwei Zhang
- Harbin Medical University, 157 Baojian Street, Harbin, Heilongjiang, 150001, PR China
| | - Ilgiz Gareev
- Bashkir State Medical University, Ufa, 450008, Russia
| | - Ozal Beylerli
- Bashkir State Medical University, Ufa, 450008, Russia
| | - Xinyuan Li
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Guitian Lu
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Lin Lv
- The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| | - Xin Hai
- Department of Pharmacy, The First Affiliated Hospital of Harbin Medical University, 23 Youzheng Street, Harbin, Heilongjiang, 150001, PR China
| |
Collapse
|
14
|
Regulatory effects of IL-1β in the interaction of GBM and tumor-associated monocyte through VCAM-1 and ICAM-1. Eur J Pharmacol 2021; 905:174216. [PMID: 34058204 DOI: 10.1016/j.ejphar.2021.174216] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/06/2021] [Accepted: 05/26/2021] [Indexed: 12/20/2022]
Abstract
Glioblastoma (GBM) is the most common and lethal brain tumor with high inflammation. GBM cells infiltrate microglia and macrophages and are surrounded by pro-inflammatory cytokines. Interleukin (IL)-1β, which is abundantly expressed in the tumor microenvironment, is involved in tumor progression. Intercellular adhesion molecule (ICAM)-1 and vascular cell adhesion molecule (VCAM)-1 mediate cell-cell interactions, and these cell adhesion molecules (CAMs) can be regulated by cytokines in immune cells or cancer cells in the inflammatory tumor microenvironment. In this study, we found that ICAM-1 and VCAM-1 expression was induced when GBM cells were treated with IL-1β, and that adhesive interaction between monocytes and GBM cells increased accordingly. The levels of soluble CAMs (sICAM-1 and sVCAM-1) were also increased in the supernatants induced by IL-1β. Furthermore, the conditioned media contained sICAM-1 and sVCAM-1, which further promoted IL-6 and CCL2 expression in differentiated macrophages. IL-1β downregulated Src homology 1 domain-containing protein tyrosine phosphatase (SHP-1) in GBM. The expression of ICAM-1 and VCAM-1 was regulated by p38, AKT, and NF-κB signaling pathways, which were modulated by SHP-1 signaling. The present study suggests that IL-1β-induced protein expression of ICAM-1 and VCAM-1 in GBM may modulate the adhesive interaction between GBM and monocytes. In addition, IL-1β also induced the soluble form of ICAM-1 and VCAM-1 in GBM, which plays a key role in the regulation of tumor-associated monocyte/macrophage polarization.
Collapse
|
15
|
Sun TX, Li MY, Zhang ZH, Wang JY, Xing Y, Ri M, Jin CH, Xu GH, Piao LX, Jin HL, Zuo HX, Ma J, Jin X. Usnic acid suppresses cervical cancer cell proliferation by inhibiting PD-L1 expression and enhancing T-lymphocyte tumor-killing activity. Phytother Res 2021; 35:3916-3935. [PMID: 33970512 DOI: 10.1002/ptr.7103] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 02/11/2021] [Accepted: 03/12/2021] [Indexed: 11/10/2022]
Abstract
The programmed cell death 1 (PD-1)/programmed death ligand 1 (PD-L1) pathway is abnormally expressed in cervical cancer cells. Moreover, PD-1/PD-L1 blockade reduces the apoptosis and exhaustion of T cells and inhibits the development of malignant tumors. Usnic acid is a dibenzofuran compound originating from Usnea diffracta Vain and has anti-inflammatory, antifungal, and anticancer activities. However, the molecular mechanism of its antitumor effects has not been fully elucidated. In this work, we first observed that usnic acid decreased the expression of PD-L1 in HeLa cells and enhanced the cytotoxicity of co-cultured T cells toward tumor cells. Usnic acid inhibited PD-L1 protein synthesis by reducing STAT3 and RAS pathways cooperatively. It was subsequently shown that usnic acid induced MiT/TFE nuclear translocation through the suppression of mTOR signaling pathways, and promoted the biogenesis of lysosomes and the translocation of PD-L1 to the lysosomes for proteolysis. Furthermore, usnic acid inhibited cell proliferation, angiogenesis, migration, and invasion, respectively, by downregulating PD-L1, thereby inhibiting tumor growth. Taken together, our results show that usnic acid is an effective inhibitor of PD-L1 and our study provide novel insights into the mechanism of its anticancer targeted therapy.
Collapse
Affiliation(s)
- Tong Xin Sun
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Ming Yue Li
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Zhi Hong Zhang
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Jing Ying Wang
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Yue Xing
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - MyongHak Ri
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Cheng Hua Jin
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Guang Hua Xu
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Lian Xun Piao
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Hong Lan Jin
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Hong Xiang Zuo
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Juan Ma
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| | - Xuejun Jin
- Molecular Medicine Research Center, College of Pharmacy, Yanbian University, Yanji, China
| |
Collapse
|
16
|
Yang MH, Ha IJ, Um JY, Ahn KS. Albendazole Exhibits Anti-Neoplastic Actions against Gastric Cancer Cells by Affecting STAT3 and STAT5 Activation by Pleiotropic Mechanism(s). Biomedicines 2021; 9:biomedicines9040362. [PMID: 33807326 PMCID: PMC8065911 DOI: 10.3390/biomedicines9040362] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 03/26/2021] [Accepted: 03/30/2021] [Indexed: 12/24/2022] Open
Abstract
Albendazole (ABZ) has been reported to display anti-tumoral actions against various maliganncies, but possible impact of ABZ on gastric cancer has not been deciphered. As aberrant phosphorylation of STAT3 and STAT5 proteins can regulate the growth and progression of gastric cancer, we postulated that ABZ may interrupt the activation of these oncogenic transcription factors. We found that ABZ exposure abrogated STAT3/5 activation, inhibited phosphorylation of Janus-activated kinases 1/2 and Src and enhanced the levels of SHP-1 protein. Silencing of SHP-1 gene by small interfering RNA (siRNA) reversed the ABZ-promoted attenuation of STAT3 as well as STAT5 activation and cellular apoptosis. In addition, these effects were noted to be driven by an augmented levels of reactive oxygen species caused by drug-induced GSH/GSSG imbalance. Thus, the data indicates that ABZ can modulate the activation of STAT3 and STAT5 by pleiotropic mechanisms in gastric cancer cells.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea;
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - In Jin Ha
- Korean Medicine Clinical Trial Center (K-CTC), Korean Medicine Hospital, Kyung Hee University, Seoul 02447, Korea;
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea;
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea;
- Correspondence: ; Tel.: +82-2-961-2316
| |
Collapse
|
17
|
Menezes JCJMDS, Diederich MF. Bioactivity of natural biflavonoids in metabolism-related disease and cancer therapies. Pharmacol Res 2021; 167:105525. [PMID: 33667686 DOI: 10.1016/j.phrs.2021.105525] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/06/2021] [Accepted: 02/27/2021] [Indexed: 12/17/2022]
Abstract
Natural biflavonoids, such as amentoflavone, bilobetin, ginkgetin, isoginkgetin, taiwaniaflavone, morelloflavone, delicaflavone, hinokiflavone, and other derivatives (~ 40 biflavonoids), are isolated from Selaginella sp., Ginkgo biloba, Garcinia sp., and several other species of plants. They are able to exert therapeutic benefits by regulating several proteins/enzymes (PPAR-γ, CCAAT/enhancer-binding protein α [C/EBPα], STAT5, pancreatic lipase, PTP1B, fatty acid synthase, α-glucosidase [AG]) and insulin signaling pathways (via PI3K-AKT), which are linked to metabolism, cell growth, and cell survival mechanisms. Deregulated insulin signaling can cause complications of obesity and diabetes, which can lead to cognitive disorders such as Alzheimer's, Parkinson's, and dementia; therefore, the therapeutic benefits of these biflavones in these areas are highlighted. Since biflavonoids have shown potential to regulate metabolism, growth- and survival-related protein/enzymes, their relation to tumor growth and metastasis of cancer associated with angiogenesis are highlighted. The translational role of biflavones in cancer with respect to the inhibition of metabolism-related processes/pathways, enzymes, or proteins, such as STAT3/SHP-1/PTEN, kinesins, tissue kallikreins, aromatase, estrogen, protein modifiers, antioxidant, autophagy, and apoptosis induction mechanisms, are discussed. Finally, considering their observed bioactivity potential, oral bioavailability studies of biflavones and related clinical trials are outlined.
Collapse
Affiliation(s)
- José C J M D S Menezes
- Faculty of Pharmaceutical Sciences, Nagasaki International University, 2825-7 Huis Ten Bosch, Sasebo, Nagasaki 859-3298, Japan
| | - Marc F Diederich
- Department of Pharmacy, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, South Korea.
| |
Collapse
|
18
|
Jung YY, Ko JH, Um JY, Chinnathambi A, Alharbi SA, Sethi G, Ahn KS. LDL cholesterol promotes the proliferation of prostate and pancreatic cancer cells by activating the STAT3 pathway. J Cell Physiol 2020; 236:5253-5264. [PMID: 33368314 DOI: 10.1002/jcp.30229] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2019] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 12/11/2022]
Abstract
Hypercholesterolemia has been found to be closely linked with a significant increase in both cancer incidence and mortality. However, the exact correlation between serum cholesterol levels and cancer has not been completely deciphered. Here we analyzed the effect of low-density lipoprotein (LDL) cholesterol on prostate and pancreatic cancer cells. We noted that LDL induced a substantial STAT3 activation and JAK1, JAK2, Src activation in diverse prostate and pancreatic tumor cells. Moreover, LDL promoted cancer cell proliferation, migration, and invasion as well as upregulated the expression of diverse oncogenic gene products. However, deletion of LDL-activated STAT3 in LNCaP and PANC-1 cells and reduced LDL-induced cell viability. Simvastatin (SV) treatment also alleviated LDL-induced cell viability and migration ability in both the prostate and pancreatic tumor cells. These results demonstrate that LDL-induced STAT3 activation may exert a profound effect on the proliferation and survival of tumor cells.
Collapse
Affiliation(s)
- Young Yun Jung
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jeong-Hyeon Ko
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | - Jae-Young Um
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| | | | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, King Saud University, Riyadh, Kingdom of Saudi Arabia
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kwang Seok Ahn
- Department of Science in Korean Medicine, Kyung Hee University, Seoul, Republic of Korea
| |
Collapse
|
19
|
Adnan M, Rasul A, Hussain G, Shah MA, Zahoor MK, Anwar H, Sarfraz I, Riaz A, Manzoor M, Adem Ş, Selamoglu Z. Ginkgetin: A natural biflavone with versatile pharmacological activities. Food Chem Toxicol 2020; 145:111642. [PMID: 32783998 DOI: 10.1016/j.fct.2020.111642] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/12/2020] [Accepted: 07/18/2020] [Indexed: 12/13/2022]
Abstract
Natural products, being richly endowed with curative powers, have become spotlight for biomedical and pharmaceutical research to develop novel therapeutics during recent years. Ginkgetin (GK), a natural non-toxic biflavone, has been shown to exhibit anti-cancer, anti-inflammatory, anti-microbial, anti-adipogenic, and neuroprotective activities. GK combats cancer progression by arresting cell cycle, inducing apoptosis, stimulating autophagy, and targeting many deregulated signaling pathways such as JAK/STAT and MAPKs. GKhalts inflammation mediators like interleukins, iNOS, COX-2, PGE2, NF-κB, and acts as an inhibitor of PLA2. GK shows strong neuroprotection against oxidative stress-promoted cell death, inhibits cerebral micro-hemorrhage, decreases neurologic deficits, and halts apoptosis of neurons. GK also acts as anti-fungal, anti-viral, anti-bacterial, leishmanicidal and anti-plasmodial agent. GK shows substantial preventive or therapeutic effects in in vivo models of many diseases including atherosclerosis, cancer, neurodegenerative, hepatic, influenza, and inflammatory diseases. Based on various computational, in vitro and in vivo evidences, this article demonstrates the potential of ginkgetin for development of therapeutics against various diseases. Although GK has been systematically studied from pharmacological point of view, a vast field of pharmacokinetics, pre-clinical and clinical studies is still open for the researchers to fully validate its potential for the treatment of various diseases.
Collapse
Affiliation(s)
- Muhammad Adnan
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan.
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Muhammad Ajmal Shah
- Department of Pharmacognosy, Faculty of Pharmaceutical Sciences, Government College University, Faisalabad, Pakistan.
| | - Muhammad Kashif Zahoor
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Haseeb Anwar
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Iqra Sarfraz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Ammara Riaz
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Maleeha Manzoor
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad, Pakistan
| | - Şevki Adem
- Department of Chemistry, Faculty of Sciences, Çankırı Karatekin University, Uluyazı Campus Çankırı, Turkey
| | - Zeliha Selamoglu
- Department of Medical Biology, Faculty of Medicine, Nigde Ömer Halisdemir University, Nigde Campus, 51240, Turkey
| |
Collapse
|
20
|
WANG X, GONG X, ZHANG H, ZHU W, JIANG Z, SHI Y, LI L. In vitro anti-aging activities of ginkgo biloba leaf extract and its chemical constituents. FOOD SCIENCE AND TECHNOLOGY 2020. [DOI: 10.1590/fst.02219] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Affiliation(s)
- Xiaoyue WANG
- Beijing Technology and Business University, China
| | - Xiaoyan GONG
- Beijing Technology and Business University, China
| | - Huina ZHANG
- Beijing Technology and Business University, China
| | | | | | - Yujing SHI
- China Academy of Chinese Medical Sciences, China
| | - Li LI
- Beijing Technology and Business University, China
| |
Collapse
|
21
|
Yang MH, Jung SH, Chinnathambi A, Alahmadi TA, Alharbi SA, Sethi G, Ahn KS. Attenuation of STAT3 Signaling Cascade by Daidzin Can Enhance the Apoptotic Potential of Bortezomib against Multiple Myeloma. Biomolecules 2019; 10:biom10010023. [PMID: 31878046 PMCID: PMC7022648 DOI: 10.3390/biom10010023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 12/19/2019] [Indexed: 12/21/2022] Open
Abstract
Daidzin (DDZ) extracted from Pueraria lobate (Fabaceae) is a widely known phytoestrogen. DDZ can display anti-cancer activities against breast and prostate cancers, but its anti-oncogenic actions in multiple myeloma (MM) cells have not been studied. The signal transducer and activator of transcription 3 (STAT3) can control key processes including proliferation, differentiation, and survival in MM cells. Here, we noted that DDZ abrogated STAT3 activation (both constitutive as well as inducible) at Tyr705 and Ser727 in MM cells. Additionally, DDZ mitigated the phosphorylation of STAT3 upstream Janus-activated kinases (JAK1/2) and c-Src kinases. Pervanadate (tyrosine phosphatase blocker) exposure altered the DDZ-induced inhibition of STAT3 activation, thus affecting the action of this phytoestrogen on apoptosis. Moreover, DDZ impeded proliferation and augmented the apoptotic effects of bortezomib (Bor) in MM cells. Overall, the data indicate that DDZ may act as a potent suppressor of STAT3 signaling cascade, and the co-treatment of DDZ and Bor could be a promising therapeutic strategy, specifically in MM.
Collapse
Affiliation(s)
- Min Hee Yang
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea; (M.H.Y.); (S.H.J.)
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
| | - Sang Hoon Jung
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea; (M.H.Y.); (S.H.J.)
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.C.); (S.A.A.)
| | - Tahani Awad Alahmadi
- Department of Pediatrics, College of Medicine and King Khalid University Hospital, King Saud University Medical City, Riyadh 11461, Saudi Arabia;
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia; (A.C.); (S.A.A.)
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Correspondence: (G.S.); (K.S.A.)
| | - Kwang Seok Ahn
- KHU-KIST Department of Converging Science and Technology, Kyung Hee University, Seoul 02447, Korea; (M.H.Y.); (S.H.J.)
- Department of Science in Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea
- Correspondence: (G.S.); (K.S.A.)
| |
Collapse
|
22
|
Hu WH, Chan GKL, Duan R, Wang HY, Kong XP, Dong TTX, Tsim KWK. Synergy of Ginkgetin and Resveratrol in Suppressing VEGF-Induced Angiogenesis: A Therapy in Treating Colorectal Cancer. Cancers (Basel) 2019; 11:cancers11121828. [PMID: 31757048 PMCID: PMC6966653 DOI: 10.3390/cancers11121828] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 12/22/2022] Open
Abstract
Ginkgetin, a biflavone from Ginkgo biloba leaf, and resveratrol, a polyphenol found in grape and wine, are two phytochemicals being identified for its binding to vascular endothelial growth factor (VEGF): the binding, therefore, resulted in the alteration of the physiological roles of VEGF-mediated angiogenesis. The bindings of ginkgetin and resveratrol were proposed on different sites of VEGF, but both of them suppressed the angiogenic properties of VEGF. The suppressive activities of ginkgetin and resveratrol in VEGF-mediated angiogenesis were supported by several lines of evidence including (i) inhibiting the formation of sub-intestinal vessel in zebrafish embryos and microvascular sprouting in rat aortic ring; and (ii) suppressing the phosphorylations of VEGFR2, Akt, eNOS, and Erk as well as expressions of matrix metalloproteinases (MMPs), MMP-2, and MMP-9 in human umbilical vein endothelial cells (HUVECs). Here, we showed the synergy of ginkgetin and resveratrol in suppressing the VEGF-induced endothelial cell proliferation, migration, invasion, and tube formation. The synergy of ginkgetin and resveratrol was further illustrated in HT-29 colon cancer xenograft nude mice. Ginkgetin and resveratrol, when applied together, exerted a synergistic anti-tumor effect of 5-fluorouracil with decreasing microvessel density of tumors. In parallel, the combination of ginkgetin and resveratrol synergistically relieved the 5-fluorouracil-induced inflammatory response by suppressing expressions of COX-2 and inflammatory cytokines. Thus, the anti-angiogenic roles of ginkgetin and/or resveratrol could provide effective therapeutic strategy in cancer, similar to that of Avastin, in suppressing the VEGF-mediated angiogenesis during cancer development.
Collapse
Affiliation(s)
- Wei-Hui Hu
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Gallant Kar-Lun Chan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Ran Duan
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Huai-You Wang
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Xiang-Peng Kong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Tina Ting-Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
| | - Karl Wah-Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal Bioresources, HKUST Shenzhen Research Institute, Hi-Tech Park, Nanshan, Shenzhen 518057, China; (W.-H.H.); (G.K.-L.C.); (R.D.); (H.-Y.W.); (X.-P.K.); (T.T.-X.D.)
- Division of Life Science and Center for Chinese Medicine, The Hong Kong University of Science and Technology, Clear Water Bay Road, Hong Kong 999077, China
- Correspondence: ; Tel.: +852-2358-7332; Fax: +852-2358-1552
| |
Collapse
|
23
|
Wang HY, Zhang YQ. The main active constituents and detoxification process of Ginkgo biloba seeds and their potential use in functional health foods. J Food Compost Anal 2019. [DOI: 10.1016/j.jfca.2019.103247] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
|
24
|
Ko JH, Um JY, Lee SG, Yang WM, Sethi G, Ahn KS. Conditioned media from adipocytes promote proliferation, migration, and invasion in melanoma and colorectal cancer cells. J Cell Physiol 2019; 234:18249-18261. [PMID: 30851074 DOI: 10.1002/jcp.28456] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/10/2018] [Accepted: 12/14/2018] [Indexed: 12/31/2022]
Abstract
Epidemiological evidence suggests that obesity can significantly increase the risk of various cancers, although the mechanisms underlying this link are completely unknown. Here, we analyzed the effect of adipocytes on melanoma and colon cancer cells proliferation, migration, and invasion. The potential effects of conditioned media (CM) obtained from differentiated mouse 3T3-L1 cells and human adipose tissue-derived mesenchymal stem cells (hAMSC) on the proliferation, migration, and invasion of B16BL6 melanoma and colon 26-L5 cancer cells were investigated. The 3T3-L1 and hAMSC CM increased cell proliferation, migration, and invasion in both the cell lines. In addition, adipocytes CM increased matrix metalloproteinase 9 (MMP-9) and MMP-2 activity in both B16BL6 and colon 26-L5 cells. These effects were found to be associated with an increased expression of various oncogenic proteins in B16BL6 and colon 26-L5 cells. Also, adipocyte CM induced Akt and mTOR activation in both tumor cell lines, and the pharmacological inhibition of Akt and mTOR blocked the CM induced Akt as well as mTOR activation and CM-stimulated melanoma and colon cancer cell proliferation, migration, and invasion. These data suggest that adipocyte promotes melanoma and colon cancer progression through modulating the expression of diverse proteins associated with cancer growth and metastasis as well as modulation of the Akt/mTOR signaling.
Collapse
Affiliation(s)
- Jeong-Hyeon Ko
- Department of Korean Pathology, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea.,Department of Korean Medicine, Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Jae-Young Um
- Department of Korean Pathology, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea.,Department of Korean Medicine, Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Seok-Geun Lee
- Department of Korean Pathology, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea.,Department of Korean Medicine, Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Woong Mo Yang
- Department of Korean Pathology, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea.,Department of Korean Medicine, Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Kwang Seok Ahn
- Department of Korean Pathology, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea.,Department of Korean Medicine, Comorbidity Research Institute, College of Korean Medicine, Kyung Hee University, Kyungheedae-ro, Dongdaemun-gu, Seoul, Republic of Korea
| |
Collapse
|
25
|
Ginkgetin ameliorates experimental atherosclerosis in rats. Biomed Pharmacother 2018; 102:510-516. [PMID: 29579712 DOI: 10.1016/j.biopha.2018.03.107] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Revised: 03/17/2018] [Accepted: 03/17/2018] [Indexed: 11/20/2022] Open
Abstract
Atherosclerosis is a common disease seriously detrimental to human health. Natural products are important sources of therapeutic candidates for atherosclerosis. We here evaluated the effects of ginkgetin on experimental atherosclerosis in rats and explored the underlying mechanisms. Atherosclerosis was induced by high-fat diet for 12 weeks combined with single intraperitoneal injection of vitamin D3 in rats. The atherosclerotic rats were then treated with ginkgetin at 25, 50 and 100 mg/kg/d or simvastatin at 2 mg/kg/d for 8 weeks. Blood and thoracic aortas were collected for analyses of histopathology, lipid deposition, serum biochemistry, matrix metalloproteinases (MMPs), and nitric oxide (NO)/NO synthase (NOS) system. We found that ginkgetin improved thoracic aortic intima structure, reduced intima-media thickness and intima/media ratio, and attenuated lipid deposition in aorta of atherosclerotic rats. Ginkgetin also decreased the serum levels of total cholesterol, triglyceride and low-density lipoprotein cholesterol, but restored the serum levels of high-density lipoprotein cholesterol in atherosclerotic rats. Additionally, ginkgetin reduced the mRNA and protein expression of MMP-2 and MMP-9 in thoracic aortas of rats with atherosclerosis. Further examinations showed that ginkgetin increased the NO and NOS levels in serum and thoracic aortas. Ginkgetin also unregulated the expression of endothelial NOS and downregulated the expression of inducible NOS at both mRNA and protein levels in thoracic aortas of atherosclerotic rats. Altogether, ginkgetin showed therapeutic effects on experimental atherosclerosis associated with improving lipid profile and modulating the MMPs and NO/NOS systems in rats. Ginkgetin could be a promising candidate for the treatment of atherosclerosis.
Collapse
|
26
|
Pan LL, Wu WJ, Zheng GF, Han XY, He JS, Cai Z. Ginkgetin inhibits proliferation of human leukemia cells via the TNF-α signaling pathway. ACTA ACUST UNITED AC 2018; 72:441-447. [PMID: 28902633 DOI: 10.1515/znc-2016-0210] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 07/27/2017] [Indexed: 01/14/2023]
Abstract
Ginkgetin is known to be an anticancer agent in many studies. However, its effectiveness in treating chronic myeloid leukemia [corrected] remains unknown. The present study aimed to evaluate the effects of ginkgetin on the growth of the K562 cell line. The MTT assay was employed to examine the proliferation of K562, and a terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick end labeling (TUNEL) staining was conducted to detect the apoptotic rates. Furthermore, changes of tumor necrosis factor-α (TNF-α) were detected by Western blot analysis. Ginkgetin inhibited the proliferation of K562 cells in a dose- and time-dependent manner. Concentrations of ginkgetin required to induce 50% death of K562 at 24, 48 and 72 h were 38.9, 31.3 and 19.2 μM, respectively. Moreover, treatment of ginkgetin increased K562 apoptosis in vitro along with increased levels of TNF-α. Interestingly, anti-TNF-α antibody prevented ginkgetin-induced K562 cell apoptosis and growth inhibition via deactivation of caspase-8, caspase-9 and caspase-3. Concomitantly, downregulation of TNF-α by etanercept in vivo attenuated ginkgetin-induced inhibitory effects on the tumor growth in an xenograft mouse model. Our results indicate that ginkgetin effectively inhibits K562 cell proliferation, and TNF-α plays a key role in ginkgetin-induced cell apoptosis.
Collapse
|
27
|
Zeng H, Fu R, Yan L, Huang J. Lycorine Induces Apoptosis of A549 Cells via AMPK-Mammalian Target of Rapamycin (mTOR)-S6K Signaling Pathway. Med Sci Monit 2017; 23:2035-2041. [PMID: 28450693 PMCID: PMC5421746 DOI: 10.12659/msm.900742] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
BACKGROUND This study was designed to investigate the effect of lycorine (LY) on the AMPK-mTOR-S6K signaling pathway and to clarify its role in autophagy and apoptosis. MATERIAL AND METHODS Various concentrations of LY were used to treat non-small cell lung carcinoma A549 cells. The MTT assay was used to measure cell viability and acridine orange staining was used to detect cell morphology changes. Western blot analysis was used to test the effect of LY on the expression levels of LC3, caspase 3, and other proteins involved in the AMPK-mTOR-S6K signaling pathway. RESULTS The half maximal inhibitory concentration (IC50) of LY after 24-h treatment was 8.5 μM, with stronger inhibitory effect of 24-h LY treatment over 12-h LY treatment. Morphological observation showed that lower doses (4 μM and 8 μM) of LY treatment induced A549 cell death mainly caused by autophagy, whereas the higher dose (16 μM) of LY treatment induced A549 cell death, mainly caused by apoptosis. Furthermore, 8 μM LY caused the highest conversion of LC3-II from LC3-I. All LY treatments activated caspase-3. LY treatment also promoted AMPK phosphorylation (Thr172) and inhibited the phosphorylation of mTOR and S6K. CONCLUSIONS LY induced apoptosis of A549 cells by regulating the AMPK-mTOR-S6K signaling pathway. Lower levels (4~8 μM) of LY-induced autophagy contributed to LY-induced apoptosis.
Collapse
Affiliation(s)
- Hui Zeng
- Department of Medicine, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China (mainland)
| | - Rong Fu
- Department of Plastic Surgery, Sichuan Provincial People's Hospital, Chengdu, Sichuan, China (mainland)
| | - Linxia Yan
- Chengdu Lilai Biotechnology Co., Ltd., Keyuan Nanlu High-Tech Zone, Chengdu, Sichuan, China (mainland)
| | - Jian Huang
- Department of Emergency Medicine, Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, Sichuan, China (mainland)
| |
Collapse
|
28
|
Baek SH, Lee JH, Kim C, Ko JH, Ryu SH, Lee SG, Yang WM, Um JY, Chinnathambi A, Alharbi SA, Sethi G, Ahn KS. Ginkgolic Acid C 17:1, Derived from Ginkgo biloba Leaves, Suppresses Constitutive and Inducible STAT3 Activation through Induction of PTEN and SHP-1 Tyrosine Phosphatase. Molecules 2017; 22:molecules22020276. [PMID: 28208828 PMCID: PMC6155672 DOI: 10.3390/molecules22020276] [Citation(s) in RCA: 53] [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: 11/08/2016] [Revised: 01/25/2017] [Accepted: 02/07/2017] [Indexed: 11/16/2022] Open
Abstract
Ginkgolic acid C 17:1 (GAC 17:1) extracted from Ginkgo biloba leaves, has been previously reported to exhibit diverse antitumor effect(s) through modulation of several molecular targets in tumor cells, however the detailed mechanism(s) of its actions still remains to be elucidated. Signal transducer and activator of transcription 3 (STAT3) is an oncogenic transcription factor that regulates various critical functions involved in progression of diverse hematological malignancies, including multiple myeloma, therefore attenuating STAT3 activation may have a potential in cancer therapy. We determined the anti-tumor mechanism of GAC 17:1 with respect to its effect on STAT3 signaling pathway in multiple myeloma cell lines. We found that GAC 17:1 can inhibit constitutive activation of STAT3 through the abrogation of upstream JAK2, Src but not of JAK1 kinases in U266 cells and also found that GAC can suppress IL-6-induced STAT3 phosphorylation in MM.1S cells. Treatment of protein tyrosine phosphatase (PTP) inhibitor blocked suppression of STAT3 phosphorylation by GAC 17:1, thereby indicating a critical role for a PTP. We also demonstrate that GAC 17:1 can induce the substantial expression of PTEN and SHP-1 at both protein and mRNA level. Further, deletion of PTEN and SHP-1 genes by siRNA can repress the induction of PTEN and SHP-1, as well as abolished the inhibitory effect of drug on STAT3 phosphorylation. GAC 17:1 down-regulated the expression of STAT3 regulated gene products and induced apoptosis of tumor cells. Overall, GAC 17:1 was found to abrogate STAT3 signaling pathway and thus exert its anticancer effects against multiple myeloma cells.
Collapse
Affiliation(s)
- Seung Ho Baek
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
- College of Korean Medicine, Woosuk University, 46 Eoeun-ro, Wansan-gu, Jeonju-si, Jeollabuk-do 54987, Korea.
| | - Jong Hyun Lee
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Chulwon Kim
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Jeong-Hyeon Ko
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Seung-Hee Ryu
- Department of Radiation Oncology, University of Ulsan College of Medicine, Asan Medical Center, Seoul 05505, Korea.
| | - Seok-Geun Lee
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Woong Mo Yang
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Jae-Young Um
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
| | - Gautam Sethi
- Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia.
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, Perth, WA 6009, Australia.
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore.
| | - Kwang Seok Ahn
- College of Korean Medicine, Kyung Hee University, 24 Kyungheedae-ro, Dongdaemun-gu, Seoul 02447, Korea.
| |
Collapse
|
29
|
Ko JH, Ho Baek S, Nam D, Chung WS, Lee SG, Lee J, Mo Yang W, Um JY, Seok Ahn K. 3-Formylchromone inhibits proliferation and induces apoptosis of multiple myeloma cells by abrogating STAT3 signaling through the induction of PIAS3. Immunopharmacol Immunotoxicol 2016; 38:334-43. [DOI: 10.1080/08923973.2016.1203928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|