1
|
Zhang X, Miao J, Song Y, Zhang J, Miao M. Review on effects and mechanisms of plant-derived natural products against breast cancer bone metastasis. Heliyon 2024; 10:e37894. [PMID: 39318810 PMCID: PMC11420494 DOI: 10.1016/j.heliyon.2024.e37894] [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: 05/27/2024] [Revised: 08/16/2024] [Accepted: 09/12/2024] [Indexed: 09/26/2024] Open
Abstract
Bone metastasis is the prevalent form of metastasis in breast cancer, resulting in severe pain, pathological fractures, nerve compression, hypercalcemia, and other complications that significantly impair patients' quality of life. The infiltration and colonization of breast cancer (BC) cells in bone tissue disrupt the delicate balance between osteoblasts and osteoclasts within the bone microenvironment, initiating a vicious cycle of bone metastasis. Once bone metastasis occurs, conventional medical therapy with bone-modifying agents is commonly used to alleviate bone-related complications and improve patients' quality of life. However, the utilization of bone-modifying agents may cause severe drug-related adverse effects. Plant-derived natural products such as terpenoids, alkaloids, coumarins, and phenols have anti-tumor, anti-inflammatory, and anti-angiogenic pharmacological properties with minimal side effects. Certain natural products that exhibit both anti-breast cancer and anti-bone metastasis effects are potential therapeutic agents for breast cancer bone metastasis (BCBM). This article reviewed the effects of plant-derived natural products against BCBM and their mechanisms to provide a reference for the research and development of drugs related to BCBM.
Collapse
Affiliation(s)
- Xiaolei Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jinxin Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Yagang Song
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Jiawen Zhang
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| | - Mingsan Miao
- Academy of Chinese Medical Sciences, Henan University of Chinese Medicine, Zhengzhou, 450046, China
| |
Collapse
|
2
|
Yadav D, Yadav A, Bhattacharya S, Dagar A, Kumar V, Rani R. GLUT and HK: Two primary and essential key players in tumor glycolysis. Semin Cancer Biol 2024; 100:17-27. [PMID: 38494080 DOI: 10.1016/j.semcancer.2024.03.001] [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: 02/04/2024] [Revised: 03/02/2024] [Accepted: 03/09/2024] [Indexed: 03/19/2024]
Abstract
Cancer cells reprogram their metabolism to become "glycolysis-dominant," which enables them to meet their energy and macromolecule needs and enhancing their rate of survival. This glycolytic-dominancy is known as the "Warburg effect", a significant factor in the growth and invasion of malignant tumors. Many studies confirmed that members of the GLUT family, specifically HK-II from the HK family play a pivotal role in the Warburg effect, and are closely associated with glucose transportation followed by glucose metabolism in cancer cells. Overexpression of GLUTs and HK-II correlates with aggressive tumor behaviour and tumor microenvironment making them attractive therapeutic targets. Several studies have proven that the regulation of GLUTs and HK-II expression improves the treatment outcome for various tumors. Therefore, small molecule inhibitors targeting GLUT and HK-II show promise in sensitizing cancer cells to treatment, either alone or in combination with existing therapies including chemotherapy, radiotherapy, immunotherapy, and photodynamic therapy. Despite existing therapies, viable methods to target the glycolysis of cancer cells are currently lacking to increase the effectiveness of cancer treatment. This review explores the current understanding of GLUT and HK-II in cancer metabolism, recent inhibitor developments, and strategies for future drug development, offering insights into improving cancer treatment efficacy.
Collapse
Affiliation(s)
- Dhiraj Yadav
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India; Drug Discovery, Jubilant Biosys, Greater Noida, Noida, Uttar Pradesh, India
| | - Anubha Yadav
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Sujata Bhattacharya
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India
| | - Akansha Dagar
- Graduate School of Nanobioscience, Yokohama City University, 22-2 Seto, Kanazawa-Ku, Yokohama 236-0027, Japan
| | - Vinit Kumar
- Amity Institute of Molecular Medicine and Stem Cell Research, Amity University, Noida, Uttar Pradesh 201303, India.
| | - Reshma Rani
- Drug Discovery, Jubilant Biosys, Greater Noida, Noida, Uttar Pradesh, India.
| |
Collapse
|
3
|
Hou W, Huang L, Huang H, Liu S, Dai W, Tang J, Chen X, Lu X, Zheng Q, Zhou Z, Zhang Z, Lan J. Bioactivities and Mechanisms of Action of Sinomenine and Its Derivatives: A Comprehensive Review. Molecules 2024; 29:540. [PMID: 38276618 PMCID: PMC10818773 DOI: 10.3390/molecules29020540] [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: 12/14/2023] [Revised: 01/13/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024] Open
Abstract
Sinomenine, an isoquinoline alkaloid extracted from the roots and stems of Sinomenium acutum, has been extensively studied for its derivatives as bioactive agents. This review concentrates on the research advancements in the biological activities and action mechanisms of sinomenine-related compounds until November 2023. The findings indicate a broad spectrum of pharmacological effects, including antitumor, anti-inflammation, neuroprotection, and immunosuppressive properties. These compounds are notably effective against breast, lung, liver, and prostate cancers, exhibiting IC50 values of approximately 121.4 nM against PC-3 and DU-145 cells, primarily through the PI3K/Akt/mTOR, NF-κB, MAPK, and JAK/STAT signaling pathways. Additionally, they manifest anti-inflammatory and analgesic effects predominantly via the NF-κB, MAPK, and Nrf2 signaling pathways. Utilized in treating rheumatic arthritis, these alkaloids also play a significant role in cardiovascular and cerebrovascular protection, as well as organ protection through the NF-κB, Nrf2, MAPK, and PI3K/Akt/mTOR signaling pathways. This review concludes with perspectives and insights on this topic, highlighting the potential of sinomenine-related compounds in clinical applications and the development of medications derived from natural products.
Collapse
Affiliation(s)
- Wen Hou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Lejun Huang
- College of Rehabilitation, Gannan Medical University, Ganzhou 341000, China;
| | - Hao Huang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Shenglan Liu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Wei Dai
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jianhong Tang
- Laboratory Animal Engineering Research Center of Ganzhou, Gannan Medical University, Ganzhou 341000, China;
| | - Xiangzhao Chen
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Xiaolu Lu
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Qisheng Zheng
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Zhinuo Zhou
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Ziyun Zhang
- College of Pharmacy, Gannan Medical University, Ganzhou 341000, China; (W.H.); (H.H.); (S.L.); (W.D.); (X.C.); (X.L.); (Q.Z.); (Z.Z.); (Z.Z.)
| | - Jinxia Lan
- College of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
4
|
Yan J, Yang J, Shen H, Gao R, Lv S. Sinomenine regulates circTRPM7-related pathway to inhibit gastric cancer cell growth and metastasis. Chem Biol Drug Des 2023; 102:870-881. [PMID: 37495546 DOI: 10.1111/cbdd.14297] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/24/2023] [Accepted: 07/12/2023] [Indexed: 07/28/2023]
Abstract
Sinomenine has been found to have antitumor effects in a variety of cancers, including gastric cancer. Circular RNA (circRNA) is an important regulator of gastric cancer progression. However, it is not known whether Sinomenine mediates gastric cancer processes by regulating circRNA-related pathways. Quantitative real-time PCR was used to measure the expression of circTRPM7, microRNA-145-5p (miR-145-5p), and pre-B-cell leukemia homeobox 3 (PBX3). MTT assay, colony formation assay, EdU assay, transwell assay, wound-healing assay, and flow cytometry were used to detect cell proliferation, migration, invasion, and apoptosis. The expression of related proteins was detected by Western blot. Mechanically, the interaction of miR-145-5p with circTRPM7/PBX3 was validated by dual-luciferase reporter assay and RIP assay. Our study showed that circTRPM7 expression was reduced in Sinomenine-treated gastric cancer cells. Moreover, overexpression of circTRPM7 upregulated the growth and metastasis of Sinomenine-treated gastric cancer cells. CircTRPM7 could sponge miR-145-5p, and miR-145-5p reversed the effect of circTRPM7 on the growth and metastasis of Sinomenine-treated gastric cancer cells. PBX3 was the target of miR-145-5p, and knockdown of PBX3 could restore the in-miR-145-5p promotion effect on the malignant behavior of Sinomenine-treated gastric cancer cells. To sum up, our data indicated that Sinomenine played an antitumor role in gastric cancer cells via circTRPM7/miR-145-5p/PBX3 axis.
Collapse
Affiliation(s)
- Jingwei Yan
- Department of Traditional Chinese Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Jingqing Yang
- Department of Geriatric Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Huifen Shen
- Department of Traditional Chinese Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Rong Gao
- Department of Rheumatology and Immunology, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Shilong Lv
- Traditional Chinese Medicine, Weihai Health School, Weihai, China
| |
Collapse
|
5
|
Ghatak S, Khanna S, Roy S, Thirunavukkarasu M, Pradeep SR, Wulff BC, El Masry MS, Sharma A, Palakurti R, Ghosh N, Xuan Y, Wilgus TA, Maulik N, Yoder MC, Sen CK. Driving adult tissue repair via re-engagement of a pathway required for fetal healing. Mol Ther 2023; 31:454-470. [PMID: 36114673 PMCID: PMC9931555 DOI: 10.1016/j.ymthe.2022.09.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 08/16/2022] [Accepted: 09/06/2022] [Indexed: 02/07/2023] Open
Abstract
Fetal cutaneous wound closure and repair differ from that in adulthood. In this work, we identify an oxidant stress sensor protein, nonselenocysteine-containing phospholipid hydroperoxide glutathione peroxidase (NPGPx), that is abundantly expressed in normal fetal epidermis (and required for fetal wound closure), though not in adult epidermis, but is variably re-induced upon adult tissue wounding. NPGPx is a direct target of the miR-29 family. Following injury, abundance of miR-29 is lowered, permitting a prompt increase in NPGPx transcripts and protein expression in adult wound-edge tissue. NPGPx expression was required to mediate increased keratinocyte migration induced by miR-29 inhibition in vitro and in vivo. Increased NPGPx expression induced increased SOX2 expression and β-catenin nuclear localization in keratinocytes. Augmenting physiologic NPGPx expression via experimentally induced miR-29 suppression, using cutaneous tissue nanotransfection or targeted lipid nanoparticle delivery of anti-sense oligonucleotides, proved to be sufficient to overcome the deleterious effects of diabetes on this specific pathway to enhance tissue repair.
Collapse
Affiliation(s)
- Subhadip Ghatak
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Savita Khanna
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Sashwati Roy
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Mahesh Thirunavukkarasu
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, Farmington, CT 06030, USA
| | - Seetur R Pradeep
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, Farmington, CT 06030, USA
| | - Brian C Wulff
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mohamed S El Masry
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Plastic Surgery, Zagazig University, Zagazig 44519, Egypt
| | - Anu Sharma
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ravichand Palakurti
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Nandini Ghosh
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Yi Xuan
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA
| | - Traci A Wilgus
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nilanjana Maulik
- Department of Surgery, Molecular Cardiology and Angiogenesis Laboratory, University of Connecticut Health, Farmington, CT 06030, USA
| | - Mervin C Yoder
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chandan K Sen
- Indiana Center for Regenerative Medicine & Engineering, Indiana University Health Comprehensive Wound Center, Department of Surgery, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN 47907, USA.
| |
Collapse
|
6
|
Liu X, Chen L, Wang T. Overcoming cisplatin resistance of human lung cancer by sinomenine through targeting the miR-200a-3p-GLS axis. J Chemother 2022:1-10. [PMID: 36120905 DOI: 10.1080/1120009x.2022.2111490] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
Abstract
Lung cancer, a malignant disease, is one of the leading causes of patient death. Non-small cell lung cancer (NSCLC) is the most common type of lung cancer. Currently, chemotherapeutic agents such as cisplatin are widely used against lung cancer. However, development of chemoresistance, which led to poor prognosis and low survival rate greatly limited the clinical applications of cisplatin. Sinomenine (SIN) is a bioactive component of sinomenium acutum. Accumulating evidence revealed SIN exhibits potential anti-tumor activities in various types of cancers. However, the precise molecular mechanisms for the sinomenine-induced anti-cancer effects have not been fully elucidated. Here, we assessed the effects of sinomenine on cisplatin sensitivity in NSCLC cells. The combination of SIN with cisplatin showed synergistically inhibitory effects on lung cancer cells by calculating the combination index (CI value) using the Calcusyn 2.0 software. Moreover, we detected that the glutamine metabolism was significantly suppressed by sinomenine treatments in lung cancer cells. Under low glutamine supply, A549 cells showed less sensitivity to sinomenine treatments. Meanwhile, miR-200a-3p was found to be significantly induced by SIN treatments. We demonstrated a suppressive role of miR-200a-3p on glutamine metabolism. Furthermore, miR-200a-3p was downregulated but the glutamine metabolism was significantly hyperactive in A549 cisplatin resistant cells compared with parental cells. Bioinformatical analysis and luciferase assay demonstrated the glutaminase (GLS), a key enzyme of glutamine metabolism, is the direct target of miR-200a-3p in lung cancer cells. Finally, rescue experiments demonstrated that recovery of GLS in miR-200a-3p overexpressing-cisplatin resistant cells successfully overrode the sinomenine-mediated cisplatin sensitization. In summary, this study revealed a new molecular mechanism for the sinomenine-promoted cisplatin sensitization, contributing to investigating the sinomenine-based therapeutic agents against chemoresistant NSCLC.
Collapse
Affiliation(s)
- Xi Liu
- Department of Thoracic Surgery, People's Liberation Army of China General Hospital, Beijing, P. R. China
| | - Lei Chen
- Department of Thoracic Surgery, People's Liberation Army of China General Hospital, Beijing, P. R. China
| | - Tao Wang
- Department of Thoracic Surgery, People's Liberation Army of China General Hospital, Beijing, P. R. China
| |
Collapse
|
7
|
Cai Q, Yang HS, Li YC, Zhu J. Dissecting the Roles of PDCD4 in Breast Cancer. Front Oncol 2022; 12:855807. [PMID: 35795053 PMCID: PMC9251513 DOI: 10.3389/fonc.2022.855807] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 05/12/2022] [Indexed: 11/29/2022] Open
Abstract
The human programmed cell death 4 (PDCD4) gene was mapped at chromosome 10q24 and encodes the PDCD4 protein comprised of 469 amino acids. PDCD4 inhibits protein translation PDCD4 inhibits protein translation to suppress tumor progression, and its expression is frequently decreased in breast cancer. PDCD4 blocks translation initiation complex by binding eIF4A via MA-3 domains or by directly binding 5’ mRNA internal ribosome entry sites with an RNA binding domain to suppress breast cancer progression and proliferation. Numerous regulators and biological processes including non-coding RNAs, proteasomes, estrogen, natural compounds and inflammation control PDCD4 expression in breast cancer. Loss of PDCD4 expression is also responsible for drug resistance in breast cancer. HER2 activation downregulates PDCD4 expression by activating MAPK, AKT, and miR-21 in aromatase inhibitor-resistant breast cancer cells. Moreover, modulating the microRNA/PDCD4 axis maybe an effective strategy for overcoming chemoresistance in breast cancer. Down-regulation of PDCD4 is significantly associated with short overall survival of patients, which suggests that PDCD4 may be an independent prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Qian Cai
- Department of Geriatric Medicine, Qilu Hospital of Shandong University, Jinan, China
- Key Laboratory of Cardiovasular Proteomics of Shandong Province, Qilu Hospital of Shandong University, Jinan, China
| | - Hsin-Sheng Yang
- Department of Toxicology and Cancer Biology, Collage of Medicine, University of Kentucky, Lexington, KY, United States
| | - Yi-Chen Li
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
| | - Jiang Zhu
- Department of Breast Surgery, General Surgery, Qilu Hospital of Shandong University, Jinan, China
- *Correspondence: Jiang Zhu,
| |
Collapse
|
8
|
Zhu Z, Zhou H, Chen F, Deng J, Yin L, He B, Hu Q, Wang T. Synthesis, Antitumor of Sinomenine Derivatives and Apoptotic Induction via IL-6/PI3K/Akt/NF-κB Signaling Pathway in MCF-7 Cells. ChemMedChem 2022; 17:e202200234. [PMID: 35612514 DOI: 10.1002/cmdc.202200234] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/19/2022] [Indexed: 11/07/2022]
Abstract
Natural products have been widely considered as an important resource for new drugs or lead compounds. Sinomenine (SIN) and its derivatives exert antitumor activity via regulation of inflammatory mediators. For these reasons we synthesized three series of SIN derivatives (compounds 4a-i, 7a-c and 11a-c) as antitumor agents from this natural product. All compounds were prepared by the modification at the C1 and C4 positions of A ring, the C4 position of A ring and the C6 and C7 positions of C ring, respectively. All the derivatives were subjected to in vitro antitumor activity against HeLa, A549, HepG-2, MCF-7 and HT-29 cell lines. To observe the apoptotic induction of SIN derivatives and its mechanism, fluorescent staining and western bolt were carried out for active compound against MCF-7. Based on the screening results, most of SIN derivatives showed better antitumor activity than SIN. Some of them were found to possess broad spectrum antitumor activity. Most notably, 11c exhibited obvious antitumor activity in both cell lines with IC50 value less than 11 μM. Besides, 11c induced apoptosis of MCF-7 in a dose-dependent manner. Western blot assay demonstrated that 11c inhibited IL-6-mediated activation of PI3K/Akt pathway. A docking study revealed that 11c had stronger binding interaction with the residues of IL-6 than SIN. All these results indicate that 11c may be a potential anti-breast cancer agent by directly targeting IL-6.
Collapse
Affiliation(s)
- Zuchang Zhu
- Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, waihuandong Road #232, Guangzhou Higher Education Mega Center, Guangzhou, China, Guangdong, China, 510006, Guangzhou, CHINA
| | - Huixian Zhou
- Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, waihuandong Road #232, Guangzhou Higher Education Mega Center, Guangzhou, China, Guangdong, China, 510006, Guangzhou, CHINA
| | - Fenglian Chen
- Guangzhou University of Traditional Chinese Medicine: Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, waihuandong Road #232, Guangzhou Higher Education Mega Center, Guangzhou, China, Guangdong, China, 510006, Guangzhou, CHINA
| | - Jianxiong Deng
- Guangzhou University of Traditional Chinese Medicine: Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, waihuandong Road #232, Guangzhou Higher Education Mega Center, Guangzhou, China, Guangdong, China, 510006, Guangzhou, CHINA
| | - Lina Yin
- Guangzhou University of Traditional Chinese Medicine: Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, waihuandong Road #232, Guangzhou Higher Education Mega Center, Guangzhou, China, Guangdong, China, 510006, Guangzhou, CHINA
| | - Baoen He
- United Biotechnology, Department of Purification R&D, Anji Road #2428, Sanzao Town, Jinwan District, Zhuhai City, Guangdong, China, 519041, Zhuhai, CHINA
| | - Qingzhong Hu
- Guangzhou University of Traditional Chinese Medicine: Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, waihuandong Road #232, Guangzhou Higher Education Mega Center, Guangzhou, China, Guangdong, China, 510006, Guangzhou, CHINA
| | - Tao Wang
- Guangzhou University of Chinese Medicine, School of Pharmaceutical Sciences, 510006, Guangzhou, CHINA
| |
Collapse
|
9
|
Li X, Chen W, Huang L, Zhu M, Zhang H, Si Y, Li H, Luo Q, Yu B. Sinomenine hydrochloride suppresses the stemness of breast cancer stem cells by inhibiting Wnt signaling pathway through down-regulation of WNT10B. Pharmacol Res 2022; 179:106222. [DOI: 10.1016/j.phrs.2022.106222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/29/2022] [Accepted: 04/07/2022] [Indexed: 12/22/2022]
|
10
|
Zhu D, Xu X, Zhang M, Wang T. TPX2 regulated by miR‑29c‑3p induces cell proliferation in osteosarcoma via the AKT signaling pathway. Oncol Lett 2022; 23:143. [PMID: 35340555 PMCID: PMC8931832 DOI: 10.3892/ol.2022.13262] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Accepted: 02/25/2022] [Indexed: 12/02/2022] Open
Abstract
The present study aimed to investigate the significance of targeting protein for Xenopus kinesin-like protein 2 (TPX2) expression in osteosarcoma. First, the TPX2 expression and survival analysis data were evaluated from The Cancer Genome Atlas (TCGA) database. Next, reverse transcription-quantitative PCR was used to explore the expression of TPX2 in osteosarcoma tissues. The observed potential target relationship between TPX2 and microRNA (miR)-29c-3p was verified using TargetScan and luciferase reporter assays. Kaplan-Meier survival analysis was used to determine associations between TPX2 expression levels and survival prognosis. TPX2 small interfering RNA was successfully constructed and transfected into osteosarcoma cell lines. The effects of TPX2 on osteosarcoma cell proliferation were then detected by MTT assay. In addition, the expression levels of AKT signaling pathway-associated proteins were identified by western blot analysis. The expression of TPX2 was upregulated and the expression of miR-29c-3p was downregulated in osteosarcoma. High expression of TPX2 was linked to a poor prognosis. Using luciferase assay and the miRNA mimic and inhibitors, miR-29c-3p was able to target and repress TPX2, and siRNA knockdown of TPX2 resulted in the inhibition of osteosarcoma cell proliferation by affecting the AKT pathway. Overall, the study showed that miR-29c-3p could inhibit the proliferation of osteosarcoma cells via TPX2 downregulation, and that TPX2 and miR-29c-3p may serve as promising prognostic indicators.
Collapse
Affiliation(s)
- Dongsheng Zhu
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang Affiliated to Xuzhou Medical University, Lianyungang, Jiangsu 222000, P.R. China
| | - Xiangfei Xu
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang Affiliated to Xuzhou Medical University, Lianyungang, Jiangsu 222000, P.R. China
| | - Ming Zhang
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang Affiliated to Xuzhou Medical University, Lianyungang, Jiangsu 222000, P.R. China
| | - Tong Wang
- Department of Pediatric Surgery, The First People's Hospital of Lianyungang Affiliated to Xuzhou Medical University, Lianyungang, Jiangsu 222000, P.R. China
| |
Collapse
|
11
|
Sabo AA, Dudau M, Constantin GL, Pop TC, Geilfus CM, Naccarati A, Dragomir MP. Two Worlds Colliding: The Interplay Between Natural Compounds and Non-Coding Transcripts in Cancer Therapy. Front Pharmacol 2021; 12:652074. [PMID: 34295245 PMCID: PMC8290364 DOI: 10.3389/fphar.2021.652074] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 06/07/2021] [Indexed: 12/25/2022] Open
Abstract
Cancer is a devastating disease and has recently become the leading cause of death in western countries, representing an immense public health burden. When it comes to cancer treatment, chemotherapy is one of the main pillars, especially for advanced stage tumors. Over the years, natural compounds have emerged as one of the most valuable resources for new chemotherapies. It is estimated that more than half of the currently used chemotherapeutic agents are derived from natural compounds. Usually, natural compounds are discovered empirically and an important limitation of introducing new anti-cancer natural products is lack of knowledge with regard to their mechanism of action. Recent data has proven that several natural compounds may function via modulating the expression and function of non-coding RNAs (ncRNAs). NcRNAs are a heterogenous class of RNA molecules which are usually not translated into proteins but have an important role in gene expression regulation and are involved in multiple tumorigenic processes, including response/resistance to pharmacotherapy. In this review, we will discuss how natural compounds function via ncRNAs while summarizing the available data regarding their effects on over 15 types of cancer. Moreover, we will critically analyze the current advances and limitations in understanding the way natural compounds exert these health-promoting effects by acting on ncRNAs. Finally, we will propose several hypotheses that may open new avenues and perspectives regarding the interaction between natural compounds and ncRNAs, which could lead to improved natural compound-based therapeutic strategies in cancer.
Collapse
Affiliation(s)
- Alexandru A. Sabo
- Pediatrics 2 (General and Special Pediatrics), Klinikum Stuttgart, Olgahospital, Zentrum für Kinder, Jugend- und Frauenmedizin, Stuttgart, Germany
| | - Maria Dudau
- Biochemistry-Proteomics Department, Victor Babes National Institute of Pathology, Bucharest, Romania
- Department of Cellular and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| | - George L. Constantin
- Division of Soil Science and Site Science, Thaer-Institute of Agricultural and Horticultural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Tudor C. Pop
- Department of Pediatrics, Marie Curie Emergency Clinical Hospital for Children, Bucharest, Romania
| | - Christoph-M. Geilfus
- Division of Controlled Environment Horticulture, Thaer-Institute of Agricultural and Horticultural Sciences, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Alessio Naccarati
- IIGM Italian Institute for Genomic Medicine, Turin, Italy
- Candiolo Cancer Institute, FPO-IRCCS, Turin, Italy
| | - Mihnea P. Dragomir
- Department of Surgery, Fundeni Clinical Hospital, Carol Davila University of Medicine and Pharmacy, Bucharest, Romania
| |
Collapse
|
12
|
Yang W, Feng Q, Li M, Su J, Wang P, Wang X, Yin Y, Wang X, Zhao M. Sinomenine Suppresses Development of Hepatocellular Carcinoma Cells via Inhibiting MARCH1 and AMPK/STAT3 Signaling Pathway. Front Mol Biosci 2021; 8:684262. [PMID: 34179090 PMCID: PMC8222788 DOI: 10.3389/fmolb.2021.684262] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Accepted: 05/27/2021] [Indexed: 01/09/2023] Open
Abstract
Promotion of apoptosis and suppression of proliferation in tumor cells are popular strategies for developing anticancer drugs. Sinomenine (SIN), a plant-derived alkaloid, displays antitumor activity. However, the mechanism of action of SIN against hepatocellular carcinoma (HCC) is unclear. Herein, several molecular technologies, such as Western Blotting, qRT-PCR, flow cytometry, and gene knockdown were applied to explore the role and mechanism of action of SIN in the treatment of HCC. It was found that SIN arrests HCC cell cycle at G0/G1 phase, induces apoptosis, and suppresses proliferation of HCC cells via down-regulating the expression of membrane-associated RING-CH finger protein 1 (MARCH1). Moreover, SIN induces cell death and growth inhibition through AMPK/STAT3 signaling pathway. MARCH1 expression was silenced by siRNA to explore its involvement in the regulation of AMPK/STAT3 signaling pathway. Silencing MARCH1 caused down-regulation of phosphorylation of AMPK, STAT3 and decreased cell viability and function. Our results suggested that SIN inhibits proliferation and promotes apoptosis of HCC cells by MARCH1-mediated AMPK/STAT3 signaling pathway. This study provides new support for SIN as a clinical anticancer drug and illustrates that targeting MARCH1 could be a novel treatment strategy in developing anticancer therapeutics.
Collapse
Affiliation(s)
- Wei Yang
- Department of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Qihua Feng
- Department of Imaging, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Minjing Li
- Department of Chinese Medicine Prescription, Binzhou Medical University, Yantai, China
| | - Jiaqi Su
- Department of Medical Imaging, Binzhou Medical University, Yantai, China
| | - Peiyuan Wang
- Department of Imaging, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Xu Wang
- Department of Imaging, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, China
| | - Yancun Yin
- Department of Human Anatomy, Binzhou Medical University, Yantai, China
| | - Xia Wang
- Department of Oral Pathology, Binzhou Medical University, Yantai, China
| | - Mingdong Zhao
- Department of Medical Imaging, Binzhou Medical University, Yantai, China
| |
Collapse
|
13
|
Zhao F, Zhao Z, Han Y, Li S, Liu C, Jia K. Baicalin suppresses lung cancer growth phenotypes via miR-340-5p/NET1 axis. Bioengineered 2021; 12:1699-1707. [PMID: 33955315 PMCID: PMC8806212 DOI: 10.1080/21655979.2021.1922052] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
As a malignant disease, lung cancer has a high morbidity and mortality rate. Baicalin is derived from Radix Scutellariae and has anti-tumor effects, however, its role in lung cancer remains unknown. Here, functional assays suggested baicalin suppressed in vitro lung cancer phenotypes. We used micro (mi)RNA array analysis to explore baicalin effects on miRNA expression. We observed baicalin increased miR-340-5p expression, whereas inhibition of this expression abolished anti-tumor effects of baicalin. Furthermore, neuroepithelial cell transforming 1 (NET1) functioned as a miR-340-5p target, and acted in a baicalin-dependent manner to regulate lung cancer progression. Thus, baicalin elicited antitumor activities by affecting the miR-340-5p/NET1 axis, suggesting a new approach to lung cancer clinical management.
Collapse
Affiliation(s)
- Fucheng Zhao
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Zhenxia Zhao
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Yanru Han
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Sujuan Li
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Caili Liu
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| | - Kui Jia
- Department of Integrated Chinese and Western Medicine, The First Affiliated Hospital of Xinxiang Medical University, Weihui, Henan, China
| |
Collapse
|
14
|
Xia J, Li S, Ma D, Guo W, Long H, Yin W. MicroRNA‑29‑3p regulates the β‑catenin pathway by targeting IGF1 to inhibit the proliferation of prolactinoma cells. Mol Med Rep 2021; 23:432. [PMID: 33846792 PMCID: PMC8060803 DOI: 10.3892/mmr.2021.12071] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 03/16/2021] [Indexed: 02/06/2023] Open
Abstract
The present study aimed to analyze the effects and underlying mechanisms of microRNA (miR)-29-3p on the proliferation and secretory abilities of prolactinoma cells by targeting insulin-like growth factor (IGF)-1/β-catenin. The relationship between miR-29a-3p and the survival of prolactinoma cells was analyzed with the Kaplan-Meier method in reference to The Cancer Genome Atlas. The expression levels of miR-29a-3p and IGF-1 in MMQ and GH3 cells were detected. A dual-luciferase reporter gene assay was performed to verify the combination of miR-29a-3p and IGF-1. Cells were transfected with a miR-29a-3p mimic and/or IGF-1 pcDNA3.1 to analyze the effects on the proliferation, apoptosis and secretion of prolactin (PRL) and growth hormone (GH) of prolactinoma cells. The effects on β-catenin in the cytoplasm and nucleus were investigated by western blot analysis. The results showed that miR-29a-3p expression was low in MMQ and GH3 cells. Overexpression miR-29a-3p inhibited IGF-1 mRNA and protein expression. miR-29a-3p inhibited cell proliferation and PRL and GH expression, and promoted apoptosis by inhibiting IGF-1. Increasing the expression of miR-29a-3p increased β-catenin levels in the cytoplasm, whereas IGF-1 promoted β-catenin activation and entry into the nucleus, and reversed the inhibitory effects of miR-29a-3p on β-catenin. To conclude, miR-29a-3p inhibited the proliferation and secretory abilities of prolactinoma cells by inhibiting nuclear translocation of β-catenin via a molecular mechanism that is inseparable from IGF-1.
Collapse
Affiliation(s)
- Jie Xia
- Department of Pharmacy, Yunnan Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, P.R. China
| | - Songmei Li
- Department of Pharmacy, Yunnan Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, P.R. China
| | - Dianfei Ma
- Department of Pharmacy, Yunnan Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650021, P.R. China
| | - Wenyujie Guo
- Department of Pediatrics, Kunming Hospital of Traditional Chinese Medicine, Kunming, Yunnan 650011, P.R. China
| | - Hong Long
- Department of Pediatrics, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| | - Weiping Yin
- Department of Pediatrics, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
15
|
Tang LB, Ma SX, Chen ZH, Huang QY, Wu LY, Wang Y, Zhao RC, Xiong LX. Exosomal microRNAs: Pleiotropic Impacts on Breast Cancer Metastasis and Their Clinical Perspectives. BIOLOGY 2021; 10:biology10040307. [PMID: 33917233 PMCID: PMC8067993 DOI: 10.3390/biology10040307] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 03/28/2021] [Accepted: 04/03/2021] [Indexed: 01/07/2023]
Abstract
As a major threat factor for female health, breast cancer (BC) has garnered a lot of attention for its malignancy and diverse molecules participating in its carcinogenesis process. Among these complex carcinogenesis processes, cell proliferation, epithelial-to-mesenchymal transition (EMT), mesenchymal-to-epithelial transition (MET), and angiogenesis are the major causes for the occurrence of metastasis and chemoresistance which account for cancer malignancy. MicroRNAs packaged and secreted in exosomes are termed "exosomal microRNAs (miRNAs)". Nowadays, more researches have uncovered the roles of exosomal miRNAs played in BC metastasis. In this review, we recapitulated the dual actions of exosomal miRNAs exerted in the aggressiveness of BC by influencing migration, invasion, and distant metastasis. Next, we presented how exosomal miRNAs modify angiogenesis and stemness maintenance. Clinically, several exosomal miRNAs can govern the transformation between drug sensitivity and chemoresistance. Since the balance of the number and type of exosomal miRNAs is disturbed in pathological conditions, they are able to serve as instructive biomarkers for BC diagnosis and prognosis. More efforts are needed to connect the theoretical studies and clinical traits together. This review provides an outline of the pleiotropic impacts of exosomal miRNAs on BC metastasis and their clinical implications, paving the way for future personalized drugs.
Collapse
Affiliation(s)
- Li-Bo Tang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (L.-B.T.); (Q.-Y.H.); (L.-Y.W.); (Y.W.); (R.-C.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Shu-Xin Ma
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Zhuo-Hui Chen
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Qi-Yuan Huang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (L.-B.T.); (Q.-Y.H.); (L.-Y.W.); (Y.W.); (R.-C.Z.)
- Second Clinical Medical College, Nanchang University, Nanchang 330006, China;
| | - Long-Yuan Wu
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (L.-B.T.); (Q.-Y.H.); (L.-Y.W.); (Y.W.); (R.-C.Z.)
- First Clinical Medical College, Nanchang University, Nanchang 330006, China
| | - Yi Wang
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (L.-B.T.); (Q.-Y.H.); (L.-Y.W.); (Y.W.); (R.-C.Z.)
| | - Rui-Chen Zhao
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (L.-B.T.); (Q.-Y.H.); (L.-Y.W.); (Y.W.); (R.-C.Z.)
- Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330006, China;
| | - Li-Xia Xiong
- Department of Pathophysiology, Basic Medical College, Nanchang University, Nanchang 330006, China; (L.-B.T.); (Q.-Y.H.); (L.-Y.W.); (Y.W.); (R.-C.Z.)
- Jiangxi Province Key Laboratory of Tumor Pathogenesis and Molecular Pathology, Nanchang 330006, China
- Correspondence: ; Tel.: +86-791-8636-0556
| |
Collapse
|
16
|
Hsa_circ_0053063 inhibits breast cancer cell proliferation via hsa_circ_0053063/hsa-miR-330-3p/PDCD4 axis. Aging (Albany NY) 2021; 13:9627-9645. [PMID: 33744861 PMCID: PMC8064214 DOI: 10.18632/aging.202707] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Accepted: 02/13/2021] [Indexed: 01/22/2023]
Abstract
Breast cancer (BC) is one of the most common malignancies and its mortality is the highest among females. Circular RNAs (circRNAs), a novel group of non-coding RNAs, play an important regulatory role in angiogenesis and cancer progression. Hsa_circ_0053063 is a circRNA generated from several exons of HADHA. The potential role of hsa_circ_0053063 in BC remains unknown and needs to be explored. Hsa_circ_0053063 was mainly located in the cytoplasm and activated in BC tissues and cell lines. The binding position between hsa_circ_0053063 and miR-330-3p was confirmed by luciferase reporter assay. Moreover, hsa_circ_0053063 inhibited cell viability, proliferation, and progression of BC through the negative regulation of miR-330-3p. Programmed cell death 4 (PDCD4) is a direct target of miR-330-3p. Besides, the over-expression of miR-330-3p promoted cell progression by directly targeting and regulating PDCD4. Mechanistically, hsa_circ_0053063 activated PDCD4 by targeting miR-330-3p to inhibit BC progression. In conclusion, hsa_circ_0053063 inhibits breast cancer cell proliferation via hsa_circ_0053063/hsa-miR-330-3p/PDCD4 axis, which may provide a new therapeutic target for BC patients.
Collapse
|
17
|
Sinomenine-phenolic acid coamorphous drug systems: Solubilization, sustained release, and improved physical stability. Int J Pharm 2021; 598:120389. [PMID: 33609724 DOI: 10.1016/j.ijpharm.2021.120389] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/01/2021] [Accepted: 02/10/2021] [Indexed: 02/07/2023]
Abstract
Sinomenine (SIN), isolated from Caulis sinomenii, is a benzyltetrahydroisoquinoline-type alkaloid with potent anti-inflammatory and analgesic effects. SIN-HCl has been used in the forms of tablets or enteric-coated tablets in the treatment of rheumatoid arthritis in China for years, while its short half-life leads to attenuated therapeutic effects and serious side effects. In the current study, three phenolic acids, including salicylic acid (SAA), 2,3-dihydroxybenzoic acid (23DHB), and 2,4-dihydroxybenzoic acid (24DHB), were firstly employed as coamorphous coformers to prepare three binary SIN-phenolic acid coamorphous systems. These new coamorphous systems were characterized by powder X-ray diffraction (PXRD), modulated temperature differential scanning calorimetry (mDSC), and Fourier transform infrared spectroscopy (FTIR). The formation of SIN-phenolic acid coamorphous systems are supported by the absence of diffraction peaks in their PXRD spectra, as well as the single Tgs of three samples (i.e., SIN-SAA, SIN-23DHB, and SIN-24DHB) at 109.5 °C, 124.9 °C, and 135.3 °C. Importantly, the salt formation between SIN and phenolic acids was observed in FTIR. In three coamorphous systems, coamorphous SIN-24DHB shows superior physicochemical stability under both low humidity and accelerated storage conditions. They were also more soluble than crystalline SIN, while were released slower than the commercial SIN-HCl in dissolution experiments. Therefore, our study suggests that phenolic acids may be used as a new type of coformers in the preparation of coamorphous systems for active pharmaceutical ingredients.
Collapse
|
18
|
Li XM, Li MT, Jiang N, Si YC, Zhu MM, Wu QY, Shi DC, Shi H, Luo Q, Yu B. Network Pharmacology-Based Approach to Investigate the Molecular Targets of Sinomenine for Treating Breast Cancer. Cancer Manag Res 2021; 13:1189-1204. [PMID: 33603465 PMCID: PMC7881794 DOI: 10.2147/cmar.s282684] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2020] [Accepted: 01/18/2021] [Indexed: 12/24/2022] Open
Abstract
Purpose Sinomenine has been known to inhibit the proliferation of breast cancer cells. However, its targets have not been found yet. This study aimed to search for molecular targets of sinomenine for treating breast cancer via network pharmacology. Methods Potential targets of sinomenine or breast cancer were separately screened from indicated databases. The common targets of both sinomenine and breast cancer were considered as the targets of sinomenine for treating breast cancer. A sinomenine-target-pathway network was constructed based on the obtained results from Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. The putative targets of sinomenine were further determined by using protein–protein interaction (PPI) analysis and molecular docking. Finally, the putative targets were verified in vitro and in vivo. Results Twenty predicted targets were identified through network pharmacological analysis. Gene Ontology (GO) and KEGG pathway enrichment indicated that these predicted targets enriched in the process of MAP kinase activity, VEGF signaling pathway, Relaxin signaling pathway, Growth hormone synthesis, secretion and action. MAPK1, NOS3, NR3C1, NOS1 and NOS2 were further identified as the putative targets by using PPI and molecular docking analysis. Expression of MAPK1, NR3C1, NOS1, NOS2 and NOS3 genes were significantly regulated by sinomenine in both MCF-7 cells and MDA-MB-231 cells. Furthermore, the expression of NR3C1 in human breast cancer specimens was lower than that in para-tumor normal tissues. Meanwhile, the expression of NR3C1 in xenograft tumors was up-regulated after sinomenine treatment. Conclusion MAPK1, NR3C1, NOS1, NOS2 and NOS3 were identified as the putative targets of sinomenine for treating breast cancer. NR3C1 was preliminarily confirmed as a target of sinomenine in two breast cancer cell lines, xenograft tumor models and human breast cancer specimens. These data indicated that the network pharmacology-based prediction of sinomenine targets for treating breast cancer could be reliable.
Collapse
Affiliation(s)
- Xiao-Mei Li
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China.,Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Mao-Ting Li
- Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China.,Student Brigade, Second Military Medical University, Shanghai, People's Republic of China
| | - Ni Jiang
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Ya-Chen Si
- Student Brigade, Second Military Medical University, Shanghai, People's Republic of China
| | - Meng-Mei Zhu
- Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| | - Qiao-Yuan Wu
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Dong-Chen Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Shanghai, 200433, People's Republic of China
| | - Hui Shi
- Department of Respiratory and Critical Care Medicine, Shanghai Changhai Hospital, Shanghai, 200433, People's Republic of China
| | - Qing Luo
- Cancer Research Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, 563003, People's Republic of China
| | - Bing Yu
- Department of Cell Biology, Center for Stem Cell and Medicine, Navy Medical University (Second Military Medical University), Shanghai, 200433, People's Republic of China
| |
Collapse
|
19
|
Feng S, Luo S, Ji C, Shi J. miR-29c-3p regulates proliferation and migration in ovarian cancer by targeting KIF4A. World J Surg Oncol 2020; 18:315. [PMID: 33261630 PMCID: PMC7709319 DOI: 10.1186/s12957-020-02088-z] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 11/16/2020] [Indexed: 12/27/2022] Open
Abstract
Background Increasing evidence suggested that microRNA and kinesin superfamily proteins play an essential role in ovarian cancer. The association between KIF4A and ovarian cancer (OC) was investigated in this study. Methods We performed bioinformatics analysis in the GEO database to screen out the differentially expressed miRNAs (DEmiRNAs) associated with ovarian cancer prognosis. Upstream targeting prediction for KIF4A was acquired by using the mirDIP database. The potential regulatory factor miR-29c-3p for KIF4A was obtained from the intersection of the above all miRNAs. The prognosis of KIF4A and target-miRNA in OC was obtained in the subsequent analysis. qRT-PCR and Western blot detected KIF4A expression level in IOSE80 (human normal ovarian epithelial cell line). In the meantime, the gene expression level was detected in A2780, HO-8910PM, COC1, and SKOV3 cell lines (human ovarian carcinoma cell line). MTT and colony formation assays were used to detect cell proliferation of SKOV3 cell line. The following assays detected cell migration through the use of transwell and wound heal assays. Targeted binding relationship between KIF4A and miRNA was detected by using the dual-luciferase reporter assay. Results Both high expression of KIF4A and lower expression of miR-29c-3p could be used as biomarkers indicating poor prognosis in OC patients. Cellular function tests confirmed that when KIF4A was silenced, it inhibited the proliferation and migration of OC cells. In addition, 3′-UTR of KIF4A had a direct binding site with miR-29c-3p, which indicated that the expression of KIF4A could be regulated by miR-29c-3p. In subsequent assays, the proliferation and migration of OC cells were inhibited by the overexpression of miR-29c-3p. At the same time, rescue experiments also confirmed that the promotion of KIF4A could be reversed by miR-29c-3p. Conclusion In a word, our data revealed a new mechanism for the role of KIF4A in the occurrence and development of OC.
Collapse
Affiliation(s)
- Songwei Feng
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Shanhui Luo
- Department of Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, People's Republic of China
| | - Chenchen Ji
- Orthopedic Institute, Soochow University, Suzhou, People's Republic of China
| | - Jia Shi
- Department of Laboratory, The Affiliated Wuxi Maternity and Child Health Care Hospital of Nanjing Medical University, 48 Huaishuxiang, Wuxi, 214002, Jiangsu Province, People's Republic of China.
| |
Collapse
|
20
|
Lu K, Chen Q, Li M, He L, Riaz F, Zhang T, Li D. Programmed cell death factor 4 (PDCD4), a novel therapy target for metabolic diseases besides cancer. Free Radic Biol Med 2020; 159:150-163. [PMID: 32745771 DOI: 10.1016/j.freeradbiomed.2020.06.016] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 02/06/2023]
Abstract
Programmed cell death factor 4 (PDCD4) is originally described as a tumor suppressor gene that exerts antineoplastic effects by promoting apoptosis and inhibiting tumor cell proliferation, invasion, and metastasis. Several investigations have probed the aberrant expression of PDCD4 with the progression of metabolic diseases, such as polycystic ovary syndrome (PCOS), obesity, diabetes, and atherosclerosis. It has been ascertained that PDCD4 causes glucose and lipid metabolism disorders, insulin resistance, oxidative stress, chronic inflammatory response, and gut flora disorders to regulate the progression of metabolic diseases. This review aims to summarize the latest researches to uncover the structure, expression regulation, and biological functions of PDCD4 and to elucidate the regulatory mechanism of the development of tumors and metabolic diseases. This review has emphasized the understanding of the PDCD4 role and to provide new ideas for the research, diagnosis, and treatment of tumors and metabolic diseases.
Collapse
Affiliation(s)
- Kaikai Lu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Qian Chen
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Mengda Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Lei He
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Farooq Riaz
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Tianyun Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China
| | - Dongmin Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi, 710061, PR China; Key Laboratory of Environment and Genes Related to Diseases, Ministry of Education, Xi'an, Shaanxi, 710061, PR China.
| |
Collapse
|
21
|
Zheng Q, Zhu Q, Li C, Hao S, Li J, Yu X, Qi D, Pan Y. Sinomenine Can Inhibit the Growth and Invasion Ability of Retinoblastoma Cell through Regulating PI3K/AKT Signaling Pathway. Biol Pharm Bull 2020; 43:1551-1555. [PMID: 32759601 DOI: 10.1248/bpb.b20-00387] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sinomenine was found to play anti-cancer functions in different type of cancers, while the mechanisms underlying the anticancer effects of sinomenine in retinoblastoma (RB) remains unclear. The present study was designed to explore the impacts of sinomenine on cell proliferation and invasion ability of RB cells and the related mechanism. Human retinoblastoma cell line WERI-RB-1 and Y79 cells were cultured and treated by different concentration of sinomenine, and then the proliferation ability of the cells was determined via performing the 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) proliferation assay. The cell apoptosis was examined via performing the flow cytometry assay. Then scratch wound healing analysis as well as and transwell invasion analysis have been performed to determine the effect of sinomenine on cell migration ability as well as invasion ability. The proteins level of phosphatidylinositol 3-kinase (PI3K)/AKT signaling molecules were determined with Western blot assay. We found that sinomenine was able to decrease the proliferation and promote the apoptosis of RB cells in a dose-dependent manner; moreover, sinomenine also significantly suppressed the migration as well as invasion ability of WERI-RB-1 and Y79 cells in vitro. Furthermore, sinomenine also de-activated PI3K/AKT signaling in WERI-RB-1 cells via inhibited the phosphorylation of PI3K and AKT proteins. Sinomenine can exert anti-tumor function on RB cells in vitro, therefore sinomenine might be a potential alterative medication for the treatment for RB.
Collapse
Affiliation(s)
- Qian Zheng
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Qin Zhu
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Cuiping Li
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Shuang Hao
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Jianguo Li
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Xin Yu
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Dengmei Qi
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| | - Yu Pan
- Department of Ophthalmology, Zibo Maternal and Child Health Hospital
| |
Collapse
|
22
|
Wang Y, Shang G, Wang W, Qiu E, Pei Y, Zhang X. Magnoflorine inhibits the malignant phenotypes and increases cisplatin sensitivity of osteosarcoma cells via regulating miR-410-3p/HMGB1/NF-κB pathway. Life Sci 2020; 256:117967. [PMID: 32553931 DOI: 10.1016/j.lfs.2020.117967] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Revised: 05/29/2020] [Accepted: 06/11/2020] [Indexed: 12/18/2022]
Abstract
AIMS Magnoflorine is an essential type of alkaloid and possesses anti-tumor activity in multiple cancers. Recent studies have demonstrated that magnoflorine plays tumor-suppressive roles in gastric and breast cancers. However, its role in osteosarcoma (OS) tumorigenesis is enigmatic. This study aimed to investigate the role and mechanism of magnoflorine in OS. MATERIALS AND METHODS Two human OS cells (MG-63 and U-2 OS) were treated with different concentrations of magnoflorine. Cell viability and invasion were then detected by Cell Counting Kit-8 and Transwell assay, respectively. And the effects of magnoflorine on the epithelial-mesenchymal transition (EMT) and cisplatin sensitivity were also measured. To explore the potential mechanism, we assayed the influence of magnoflorine on the miR-410-3p/HMGB1/NF-κB signaling pathway. Additionally, rescue experiments were performed to further confirm the regulation mechanism of magnoflorine. KEY FINDINGS Magnoflorine inhibited the viability, invasion, and EMT of OS cells in a dose-dependent manner. And it increased the sensitivity of OS cells to cisplatin. Magnoflorine significantly suppressed HMGB1 expression and NF-κB activation, but upregulated miR-410-3p level. Overexpression of HMGB1 promoted NF-κB activation and reversed the effects of magnoflorine on the viability, invasion, EMT and cisplatin sensitivity of OS cells. miR-410-3p mimic inhibited the EMT of OS cells, which was restored by HMGB1 upregulation. And miR-410-3p inhibitor abrogated the influence of magnoflorine on HMGB1 expression in OS cells. SIGNIFICANCE Magnoflorine inhibited the malignant phenotypes and increased cisplatin sensitivity of OS cells via modulating miR-410-3p/HMGB1/NF-κB pathway. These results indicated that magnoflorine might be a novel drug for the treatment of OS.
Collapse
Affiliation(s)
- Yuming Wang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Guanning Shang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Wei Wang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Enduo Qiu
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Yi Pei
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China
| | - Xiaojing Zhang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Shenyang 110042, Liaoning Province, PR China.
| |
Collapse
|
23
|
Sinomenine Inhibits Migration and Invasion of Human Lung Cancer Cell through Downregulating Expression of miR-21 and MMPs. Int J Mol Sci 2020; 21:ijms21093080. [PMID: 32349289 PMCID: PMC7247699 DOI: 10.3390/ijms21093080] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Accepted: 04/24/2020] [Indexed: 02/06/2023] Open
Abstract
Sinomenine is an alkaloid derived from Sinomenium acutum. Recent studies have found that sinomenine can inhibit various cancers by inhibiting the proliferation, migration and invasion of tumors and inducing apoptosis. This study aims to investigate the effect and mechanism of sinomenine on inhibiting the migration and invasion of human lung adenocarcinoma cells in vitro. The results demonstrate that viabilities of A549 and H1299 cells were inhibited by sinomenine in a dose-dependent manner. When treated with sub-toxic doses of sinomenine, cell migration and invasion are markedly suppressed. Sinomenine decreases the mRNA level of matrix metalloproteinase-2 (MMP-2), MMP-9, and the extracellular inducer of matrix metalloproteinase (EMMPRIN/CD147), but elevates the expression of reversion-inducing cysteine-rich proteins with kazal motifs (RECK) and the tissue inhibitor of metalloproteinase-1 (TIMP-1) and TIMP-2. In addition, sinomenine significantly increases the expression of the epithelial marker E-cadherin but concomitantly decreases the expression of the mesenchymal marker vimentin, suggesting that it suppresses epithelial–mesenchymal transition (EMT). Moreover, sinomenine downregulates oncogenic microRNA-21 (miR-21), which has been known to target RECK. The downregulation of miR-21 decreases cell invasion, while the upregulation of miR-21 increases cell invasion. Furthermore, the downregulation of miR-21 stimulates the expression of RECK, TIMP-1/-2, and E-cadherin, but reduces the expression of MMP-2/-9, EMMPRIN/CD147, and vimentin. Taken together, the results reveal that the inhibition of A549 cell invasion by sinomenine may, at least in part, be through the downregulating expression of MMPs and miR-21. These findings demonstrate an attractive therapeutic potential for sinomenine in lung cancer anti-metastatic therapy.
Collapse
|
24
|
Liu W, Yu X, Zhou L, Li J, Li M, Li W, Gao F. Sinomenine Inhibits Non-Small Cell Lung Cancer via Downregulation of Hexokinases II-Mediated Aerobic Glycolysis. Onco Targets Ther 2020; 13:3209-3221. [PMID: 32368080 PMCID: PMC7176511 DOI: 10.2147/ott.s243212] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 03/28/2020] [Indexed: 12/16/2022] Open
Abstract
Background Addiction to aerobic glycolysis is a common metabolic phenotype in human non-small cell lung cancer (NSCLC). The natural product Sinomenine (Sin) exhibits significant anti-tumor effects in various human cancers. However, the underlying mechanism remains elusive. Methods The inhibitory effect of Sin on NSCLC cells was determined by MTS and soft agar assays. The glycolysis efficacy of NSCLC cells was examined by glucose uptake and lactate production. The activation of Akt signaling and the protein level of hexokinases II (HK2) were examined by immunoblot (IB), qRT-PCR, and immunohistochemical staining (IHC). The in vivo anti-tumor effect of Sin was validated by the xenograft mouse model. Results We showed that HK2 is highly expressed in NSCLC tissues and cell lines. Depletion of HK2 suppressed cell viability, anchorage-independent colony formation, and xenograft tumor growth. Sinomenine exhibited a profound inhibitory effect on NSCLC cells by reducing HK2-mediated glycolysis both in vitro and in vivo. Ectopic overexpression of HK2 compromised these anti-tumor efficacies in sinomenine-treated NSCLC cells. Moreover, we revealed that sinomenine decreased Akt activity, which caused the down-regulation of HK2-mediated glycolysis. Knockdown of Akt reduced HK2 protein level and impaired glycolysis. In contrast, overexpression of constitutively activated Akt1 reversed this phenotype. Conclusion This study suggests that targeting HK2-mediated aerobic glycolysis is required for sinomenine-mediated anti-tumor activity.
Collapse
Affiliation(s)
- Wenbin Liu
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, People's Republic of China
| | - Xinfang Yu
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio 44195, USA
| | - Li Zhou
- Department of Pathology, Xiangya Hospital of Central South University, Changsha, Hunan 410008, People's Republic of China
| | - Jigang Li
- Department of Pathology, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410006, People's Republic of China
| | - Ming Li
- School of Stomatology, Hunan University of Chinese Medicine, Changsha, Hunan 410208, People's Republic of China.,Changsha Stomatological Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, 410004, People's Republic of China
| | - Wei Li
- Department of Radiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Feng Gao
- Department of Ultrasonography, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, People's Republic of China
| |
Collapse
|
25
|
Varghese E, Liskova A, Kubatka P, Samuel SM, Büsselberg D. Anti-Angiogenic Effects of Phytochemicals on miRNA Regulating Breast Cancer Progression. Biomolecules 2020; 10:biom10020191. [PMID: 32012744 PMCID: PMC7072640 DOI: 10.3390/biom10020191] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 01/19/2020] [Accepted: 01/25/2020] [Indexed: 12/16/2022] Open
Abstract
Several phytochemicals have been identified for their role in modifying miRNA regulating tumor progression. miRNAs modulate the expression of several oncogenes and tumor suppressor genes including the genes that regulate tumor angiogenesis. Hypoxia inducible factor-1 alpha (HIF-1α) signaling is a central axis that activates oncogenic signaling and acts as a metabolic switch in endothelial cell (EC) driven tumor angiogenesis. Tumor angiogenesis driven by metabolic reprogramming of EC is crucial for tumor progression and metastasis in many different cancers, including breast cancers, and has been linked to aberrant miRNA expression profiles. In the current article, we identify different miRNAs that regulate tumor angiogenesis in the context of oncogenic signaling and metabolic reprogramming in ECs and review how selected phytochemicals could modulate miRNA levels to induce an anti-angiogenic action in breast cancer. Studies involving genistein, epigallocatechin gallate (EGCG) and resveratrol demonstrate the regulation of miRNA-21, miRNA-221/222 and miRNA-27, which are prognostic markers in triple negative breast cancers (TNBCs). Modulating the metabolic pathway is a novel strategy for controlling tumor angiogenesis and tumor growth. Cardamonin, curcumin and resveratrol exhibit their anti-angiogenic property by targeting the miRNAs that regulate EC metabolism. Here we suggest that using phytochemicals to target miRNAs, which in turn suppresses tumor angiogenesis, should have the potential to inhibit tumor growth, progression, invasion and metastasis and may be developed into an effective therapeutic strategy for the treatment of many different cancers where tumor angiogenesis plays a significant role in tumor growth and progression.
Collapse
Affiliation(s)
- Elizabeth Varghese
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (E.V.); (S.M.S.)
| | - Alena Liskova
- Clinic of Obstetrics and Gynecology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Peter Kubatka
- Department of Medical Biology, Jessenius Faculty of Medicine, Comenius University in Bratislava, 03601 Martin, Slovakia;
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (E.V.); (S.M.S.)
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha P.O. Box 24144, Qatar; (E.V.); (S.M.S.)
- Correspondence: ; Tel.: +974-4492-8334; Fax: +974-4492-8333
| |
Collapse
|