1
|
Varghese S, Jisha M, Rajeshkumar K, Gajbhiye V, Alrefaei AF, Jeewon R. Endophytic fungi: A future prospect for breast cancer therapeutics and drug development. Heliyon 2024; 10:e33995. [PMID: 39091955 PMCID: PMC11292557 DOI: 10.1016/j.heliyon.2024.e33995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/26/2024] [Accepted: 07/02/2024] [Indexed: 08/04/2024] Open
Abstract
Globally, breast cancer is a primary contributor to cancer-related fatalities and illnesses among women. Consequently, there is a pressing need for safe and effective treatments for breast cancer. Bioactive compounds from endophytic fungi that live in symbiosis with medicinal plants have garnered significant interest in pharmaceutical research due to their extensive chemical composition and prospective medicinal attributes. This review underscores the potentiality of fungal endophytes as a promising resource for the development of innovative anticancer agents specifically tailored for breast cancer therapy. The diversity of endophytic fungi residing in medicinal plants, success stories of key endophytic bioactive metabolites tested against breast cancer and the current progress with regards to in vivo studies and clinical trials on endophytic fungal metabolites in breast cancer research forms the underlying theme of this article. A thorough compilation of putative anticancer compounds sourced from endophytic fungi that have demonstrated therapeutic potential against breast cancer, spanning the period from 1990 to 2022, has been presented. This review article also outlines the latest trends in endophyte-based drug discovery, including the use of artificial intelligence, machine learning, multi-omics approaches, and high-throughput strategies. The challenges and future prospects associated with fungal endophytes as substitutive sources for developing anticancer drugs targeting breast cancer are also being highlighted.
Collapse
Affiliation(s)
- Sherin Varghese
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India
| | - M.S. Jisha
- School of Biosciences, Mahatma Gandhi University, Kottayam, Kerala, 686560, India
| | - K.C. Rajeshkumar
- National Fungal Culture Collection of India (NFCCI), Biodiversity and Palaeobiology (Fungi) Gr., Agharkar Research Institute, G.G. Agharkar Road, Pune, 411 004, Maharashtra, India
| | - Virendra Gajbhiye
- Nanobioscience Group, Agharkar Research Institute, G.G. Agharkar Road, Pune, 411 004, Maharashtra, India
| | - Abdulwahed Fahad Alrefaei
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
| | - Rajesh Jeewon
- Department of Zoology, College of Science, King Saud University, P.O. Box 2455, Riyadh, 11451, Saudi Arabia
- Department of Health Sciences, Faculty of Medicine and Health Sciences, University of Mauritius, Reduit, Mauritius
| |
Collapse
|
2
|
Dang W, Guo P, Song X, Zhang Y, Li N, Yu C, Xing B, Liu R, Jia X, Zhang Q, Feng X, Liu Z. Nuclear Targeted Peptide Combined With Gambogic Acid for Synergistic Treatment of Breast Cancer. Front Chem 2022; 9:821426. [PMID: 35155383 PMCID: PMC8832139 DOI: 10.3389/fchem.2021.821426] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 12/14/2021] [Indexed: 12/24/2022] Open
Abstract
As a natural compound, gambogic acid (GA) emerged a shining multi-target antitumor activity in a variety of tumors. Whereas its poor solubility and non-specific effect to tumor blocked the clinical application of this drug. Herein, we reported a simple and effective strategy to construct liposome modified with nuclear targeted peptide CB5005N (VQRKRQKLMPC) via polyethylene glycol (PEG) linker to decrease the inherent limitations of GA and promote its anti-tumor activity. In this study, liposomes were prepared by thin film hydration method. The characterization of formulations contained particle size, Zeta potential, morphology and encapsulation efficiency. Further, in vitro cytotoxicity and uptake tests were investigated by 4T1 and MDA-MB-231 cells, and nuclear targeting capability was performed on MDA-MB-231 cells. In addition, the in vivo antitumor effect and biological distribution of formulations were tested in BALB/c female mice. The GA-loaded liposome modified by CB5005N showed small size, good uniformity, better targeting, higher anti-tumor efficiency, better tumor inhibition rate and lower toxicity to normal tissues than other groups. In vitro and in vivo research proved that CB5005N-GA-liposome exhibited excellent anti-tumor activity and significantly reduced toxicities. As a result, CB5005N-GA-liposome nano drug delivery system enhanced the tumor targeting and antitumor effects of GA, which provided a basis for its clinical application.
Collapse
Affiliation(s)
- Wenli Dang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pan Guo
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xunan Song
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Ying Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Nan Li
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Changxiang Yu
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Bin Xing
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Rui Liu
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xintao Jia
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Qingqing Zhang
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Xiaojiao Feng
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhidong Liu
- Tianjin State Key Laboratory of Component-based Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Heihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Engineering Research Center of Modern Chinese Medicine Discovery and Preparation Technique, Ministry of Education, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- *Correspondence: Zhidong Liu,
| |
Collapse
|
3
|
Amewu RK, Sakyi PO, Osei-Safo D, Addae-Mensah I. Synthetic and Naturally Occurring Heterocyclic Anticancer Compounds with Multiple Biological Targets. Molecules 2021; 26:7134. [PMID: 34885716 PMCID: PMC8658833 DOI: 10.3390/molecules26237134] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/21/2021] [Accepted: 11/22/2021] [Indexed: 01/09/2023] Open
Abstract
Cancer is a complex group of diseases initiated by abnormal cell division with the potential of spreading to other parts of the body. The advancement in the discoveries of omics and bio- and cheminformatics has led to the identification of drugs inhibiting putative targets including vascular endothelial growth factor (VEGF) family receptors, fibroblast growth factors (FGF), platelet derived growth factors (PDGF), epidermal growth factor (EGF), thymidine phosphorylase (TP), and neuropeptide Y4 (NY4), amongst others. Drug resistance, systemic toxicity, and drug ineffectiveness for various cancer chemo-treatments are widespread. Due to this, efficient therapeutic agents targeting two or more of the putative targets in different cancer cells are proposed as cutting edge treatments. Heterocyclic compounds, both synthetic and natural products, have, however, contributed immensely to chemotherapeutics for treatments of various diseases, but little is known about such compounds and their multimodal anticancer properties. A compendium of heterocyclic synthetic and natural product multitarget anticancer compounds, their IC50, and biological targets of inhibition are therefore presented in this review.
Collapse
Affiliation(s)
- Richard Kwamla Amewu
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
| | - Patrick Opare Sakyi
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
- Department of Chemical Sciences, School of Sciences, University of Energy and Natural Resources, Sunyani P.O. Box 214, Ghana
| | - Dorcas Osei-Safo
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
| | - Ivan Addae-Mensah
- Department of Chemistry, School of Physical and Mathematical Sciences, College of Basic and Applied Sciences, University of Ghana, Legon, Accra P.O. Box LG 56, Ghana; (R.K.A.); (P.O.S.); (D.O.-S.)
| |
Collapse
|
4
|
Pu Y, Jin P, Liu L, Pu Q, Wu F. Dysosma versipellis Extract Inhibits Esophageal Cancer Progression through the Wnt Signaling Pathway. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2021; 2021:1221899. [PMID: 34729077 PMCID: PMC8557981 DOI: 10.1155/2021/1221899] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022]
Abstract
OBJECTIVE In this study, we aim to investigate the effect of Dysosma versipellis extract on biological behavior of esophageal cancer cells and its underlying mechanisms. METHODS A total of 30 BALB/C nude mice (class SPF) were equally and randomly divided into the control group, model group, and Dysosma versipellis group. CP-C cell of esophageal cancer was subcutaneously injected into the model group as well as the Dysosma versipellis group, and the same amount of normal saline into the control group, in order to compare the tumorigenesis of nude mice of three groups. Wnt, β-catenin, and p-GSK3β/GSK3β expression in tumor tissues was detected using Western blot. CP-C cells in logarithmic growth were selected and divided into 4 groups, including the control group, podophyllotoxin group, Wnt activator group, and combined group (mixture of podophyllotoxin and Wnt activator). The cell viability, apoptosis, and invasion ability, Wnt, β-catenin, and p-GSK3β/GSK3β expression level of CP-C cells in each group were detected via MTT assay, flow cytometry, transwell, and Western blot, respectively. RESULTS The tumorigenesis rates of the control group, model group, and Dysosma versipellis group were 0%, 90% (1 tumor-free mouse), and 80% (2 tumor-free mice), respectively. The tumor mass in the Dysosma versipellis group was significant less than that in the model group. Based on the results of Western blot, Wnt, ß-catenin, and p-GSK3β/GSK3β expression of the Dysosma versipellis group was lower than that of the control group. The in vitro viability test indicated that there was a significant difference in cell viability exhibited among four groups. Cell viability level in the 3 groups, including the combined group, blank group, and Wnt activator group, was higher than the podophyllotoxin group at each time point. In vitro apoptosis assay revealed that significant differences in cell apoptosis exhibited among four groups. Cell apoptosis rate was higher in the podophyllotoxin group compared to the remaining three groups. The Wnt activator group showed the lowest cell apoptosis rate. The in vitro invasion assay demonstrated that numbers of transmembrane cell in the 3 groups, involving the combined group, blank group, and Wnt activator group, showed a higher level than the podophyllotoxin group. The results of Western blot manifested that the podophyllotoxin group showed lower level of Wnt, ß-catenin, and p-GSK3β/GSK3β expression compared to the other 3 groups. CONCLUSION Podophyllotoxin in Dysosma versipellis has an excellent antiesophageal cancer effect and is able to inhibit cell viability as well as invasion ability and promote apoptosis of esophageal cancer cells by inhibiting the Wnt signaling pathway, which could be potentially used in future clinical treatment of esophageal cancer.
Collapse
Affiliation(s)
- Yanchun Pu
- School of Pharmaceutical Sciences, Hunan University of Medicine, No. 492, Jinxi South Road, Huaihua, Hunan Province 418099, China
| | - Ping Jin
- School of Pharmaceutical Sciences, Hunan University of Medicine, No. 492, Jinxi South Road, Huaihua, Hunan Province 418099, China
| | - Lianghong Liu
- School of Pharmaceutical Sciences, Hunan University of Medicine, No. 492, Jinxi South Road, Huaihua, Hunan Province 418099, China
| | - Qinlin Pu
- School of Pharmaceutical Sciences, Hunan University of Medicine, No. 492, Jinxi South Road, Huaihua, Hunan Province 418099, China
| | - Fangping Wu
- School of Pharmaceutical Sciences, Hunan University of Medicine, No. 492, Jinxi South Road, Huaihua, Hunan Province 418099, China
| |
Collapse
|
5
|
Fan HY, Zhu ZL, Xian HC, Wang HF, Chen BJ, Tang YJ, Tang YL, Liang XH. Insight Into the Molecular Mechanism of Podophyllotoxin Derivatives as Anticancer Drugs. Front Cell Dev Biol 2021; 9:709075. [PMID: 34447752 PMCID: PMC8383743 DOI: 10.3389/fcell.2021.709075] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Accepted: 07/22/2021] [Indexed: 02/05/2023] Open
Abstract
Podophyllotoxin (PTOX) is a biologically active compound derived from the podophyllum plant, and both it and its derivatives possess excellent antitumor activity. The PTOX derivatives etoposide (VP-16) and teniposide (VM-26) have been approved by the U.S. Food and Drug Administration (FDA) for cancer treatment, but are far from perfect. Hence, numerous PTOX derivatives have been developed to address the major limitations of PTOX, such as systemic toxicity, drug resistance, and low bioavailability. Regarding their anticancer mechanism, extensive studies have revealed that PTOX derivatives can induce cell cycle G2/M arrest and DNA/RNA breaks by targeting tubulin and topoisomerase II, respectively. However, few studies are dedicated to exploring the interactions between PTOX derivatives and downstream cancer-related signaling pathways, which is reasonably important for gaining insight into the role of PTOX. This review provides a comprehensive analysis of the role of PTOX derivatives in the biological behavior of tumors and potential molecular signaling pathways, aiming to help researchers design and develop better PTOX derivatives.
Collapse
Affiliation(s)
- Hua-yang Fan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Zhuo-li Zhu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Hong-chun Xian
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Hao-fan Wang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Bing-jun Chen
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Ya-Jie Tang
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao, China
| | - Ya-ling Tang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| | - Xin-hua Liang
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology (Sichuan University), Chengdu, China
| |
Collapse
|
6
|
Yang Y, Liu L, Sun J, Wang S, Yang Z, Li H, Huang N, Zhao W. Deoxypodophyllotoxin Inhibits Non-Small Cell Lung Cancer Cell Growth by Reducing HIF-1α-Mediated Glycolysis. Front Oncol 2021; 11:629543. [PMID: 33732648 PMCID: PMC7959795 DOI: 10.3389/fonc.2021.629543] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 01/11/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer cell proliferation is a metabolically demanding process that requires high rate of glycolysis to support anabolic growth. Deoxypodophyllotoxin (DPT) is a natural flavonolignan with various pharmacological activities, including antitumor effect. However, whether DPT affects the metabolic reprogramming of cancer cells is unknown. The purpose of this study is to investigate the role of DPT on non-small cell lung cancer (NSCLC) and to explore whether HIF-1α-mediated glycolysis is involved in its mechanism of action.The level of HIF-1α mRNA and protein in NSCLC cells following DPT treatment was detected using qRT-PCR and western blotting, respectively. Cell Counting Kit-8 (CCK-8) and caspase-3 activity assays were performed to analyze cell proliferation and apoptosis. The underlying molecular mechanism was identified by dual luciferase assay, Western blotting, qRT-PCR, glucose consumption, lactate production, and immunoprecipitation. A murine NSCLC model was used to clarify the effect of DPT treatment on tumor cell proliferation. Our findings showed that DPT treatment inhibited NSCLC cell growth in a dose- and time-dependent manner. Further analysis suggested that DPT treatment inhibited HIF-1α signaling pathway by Parkin-mediated protein degradation in NSCLC cells. DPT treatment significantly decreased glucose consumption and lactate production. In addition, DPT treatment reduced the expression of HIF-1α target genes, including GLUT1, HK2 and LDHA, resulting in reduction in glycolysis. We further revealed that DPT-induced cell growth inhibition and increased glucose and lactate levels could be reversed by overexpressing HIF-1α. Additionally, we found that DPT repressed NSCLC growth and GLUT1, HK2 and LDHA expression in vivo. Overall, this study suggested that DPT inhibited NSCLC growth by preventing HIF-1α-mediated glycolysis.
Collapse
Affiliation(s)
- Yuping Yang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Lingling Liu
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, China.,Development and Regeneration Key Laboratory of Sichuan Province, Chengdu Medical College, Chengdu, China
| | - Jinghui Sun
- School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, China
| | - Shu Wang
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, Nanjing, China
| | | | - Honghui Li
- Department of Refractive Surgery, Chengdu Aier Eye Hospital, Chengdu, China
| | - Na Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Wei Zhao
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Chengdu Medical College, Chengdu, China.,School of Laboratory Medicine/Sichuan Provincial Engineering Laboratory for Prevention and Control Technology of Veterinary Drug Residue in Animal-origin Food, Chengdu Medical College, Chengdu, China
| |
Collapse
|
7
|
Zhang J, Lu R, Zhang Y, Matuszek Ż, Zhang W, Xia Y, Pan T, Sun J. tRNA Queuosine Modification Enzyme Modulates the Growth and Microbiome Recruitment to Breast Tumors. Cancers (Basel) 2020; 12:E628. [PMID: 32182756 PMCID: PMC7139606 DOI: 10.3390/cancers12030628] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Transfer RNA (tRNA) queuosine (Q)-modifications occur specifically in 4 cellular tRNAs at the wobble anticodon position. tRNA Q-modification in human cells depends on the gut microbiome because the microbiome product queuine is required for its installation by the enzyme Q tRNA ribosyltransferase catalytic subunit 1 (QTRT1) encoded in the human genome. Queuine is a micronutrient from diet and microbiome. Although tRNA Q-modification has been studied for a long time regarding its properties in decoding and tRNA fragment generation, how QTRT1 affects tumorigenesis and the microbiome is still poorly understood. RESULTS We generated single clones of QTRT1-knockout breast cancer MCF7 cells using Double Nickase Plasmid. We also established a QTRT1-knockdown breast MDA-MB-231 cell line. The impacts of QTRT1 deletion or reduction on cell proliferation and migration in vitro were evaluated using cell culture, while the regulations on tumor growth in vivo were evaluated using a xenograft BALB/c nude mouse model. We found that QTRT1 deficiency in human breast cancer cells could change the functions of regulation genes, which are critical in cell proliferation, tight junction formation, and migration in human breast cancer cells in vitro and a breast tumor mouse model in vivo. We identified that several core bacteria, such as Lachnospiraceae, Lactobacillus, and Alistipes, were markedly changed in mice post injection with breast cancer cells. The relative abundance of bacteria in tumors induced from wildtype cells was significantly higher than those of QTRT1 deficiency cells. CONCLUSIONS Our results demonstrate that the QTRT1 gene and tRNA Q-modification altered cell proliferation, junctions, and microbiome in tumors and the intestine, thus playing a critical role in breast cancer development.
Collapse
Affiliation(s)
- Jilei Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (J.Z.); (R.L.); (Y.Z.); (Y.X.)
| | - Rong Lu
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (J.Z.); (R.L.); (Y.Z.); (Y.X.)
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (J.Z.); (R.L.); (Y.Z.); (Y.X.)
| | - Żaneta Matuszek
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL 60637, USA; (Ż.M.); (T.P.)
| | - Wen Zhang
- Department of Chemistry, University of Chicago, Chicago, IL 60637, USA;
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (J.Z.); (R.L.); (Y.Z.); (Y.X.)
| | - Tao Pan
- Department of Biochemistry & Molecular Biology, University of Chicago, Chicago, IL 60637, USA; (Ż.M.); (T.P.)
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Illinois at Chicago, Chicago, IL 60612, USA; (J.Z.); (R.L.); (Y.Z.); (Y.X.)
- University of Illinois Cancer Center, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
8
|
Zhang J, Li K, Zhang Y, Lu R, Wu S, Tang J, Xia Y, Sun J. Deletion of sorting nexin 27 suppresses proliferation in highly aggressive breast cancer MDA-MB-231 cells in vitro and in vivo. BMC Cancer 2019; 19:555. [PMID: 31182056 PMCID: PMC6558813 DOI: 10.1186/s12885-019-5769-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Accepted: 05/29/2019] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sorting Nexin 27 (SNX27) belongs to a family of sortin nexins and possesses a unique binding domain at the C-terminus which mediates protein-protein interaction in intracellular trafficking, membrane remodeling, organelle motility, and tight junctions. However, its role in cancer development, especially in vivo, remains largely unknown. METHODS We have generated a stable SNX27 knockdown clone in a highly aggressive breast cancer cell line MDA-MB-231 using an inducible lentiviral shRNA system. Cell migration and proliferation of SNX27 knockdown (KD) cells were compared with wild-type (WT) cells by MTT and wound healing assay, respectively. The differences in colony formation between SNX27-KD and WT cells were detected by soft agar culture and matrigel 3D culture. Furthermore, tumor growth was examined in a xenograft nude mouse model using SNX27-KD and WT MDA-MB-231 cells. The critical EMT (epithelial-mesenchymal transition) regulators were examined in vitro and in vivo. RESULTS The wound healing assay showed that SNX27 knockdown significantly decreased cell motility and proliferation. Colony formation in soft agar showed that the SNX27 knockdown cells formed significantly fewer and smaller colonies than the parental MDA-MB-231 cells. Western blots and immunostaining showed that knockdown of SNX27 led to increased expression of E-cadherin and β-catenin proteins, which facilitate adhesion formation and reverse EMT. EMT is a cellular program that allows polarized, immotile epithelial cells to convert to motile mesenchymal cells, promoting carcinoma invasion. The expression levels of Vimentin, the transcription factor of EMT, and tight junction protein Claudin-5, were significantly diminished in the SNX27 knockdown cells. The expression of PCNA, the cell proliferation marker, was increased in SNX27-KD cells transfected with E-cadherin siRNA. In a xenograft nude mouse model, we found that knockdown of SNX27 significantly inhibited tumor growth. The tumors from mice with SNX27-KD cells showed less proliferation compared to tumors from mice injected with wildtype cells. The increase in E-cadherin and β-catenin and decrease in Vimentin and Claudin-5 were observed in tumors of mice injected with SNX27-KD cells. CONCLUSIONS Our data have demonstrated that SNX27 plays a crucial role in tumor growth in vitro and in vivo.
Collapse
Affiliation(s)
- Jilei Zhang
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Kendy Li
- Liberal Arts & Human Sciences, Virginia Polytechnic Institute and State University, Blacksburg, Virginia, USA
| | - Yongguo Zhang
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Rong Lu
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Shaoping Wu
- Department of Biochemistry, Rush University, Chicago, IL, USA
| | - Jingrong Tang
- Section of Inflammation and Cardiometabolic Diseases, National Heart, Lung, and Blood Institute (NHLBI), National Institutes of Health, Bethesda, MD, USA
| | - Yinglin Xia
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA
| | - Jun Sun
- Division of Gastroenterology and Hepatology, Medicine, University of Illinois at Chicago, 840 S Wood Street, Room 704 CSB, MC716, Chicago, IL, 60612, USA.
| |
Collapse
|
9
|
Gamage CDB, Park SY, Yang Y, Zhou R, Taş İ, Bae WK, Kim KK, Shim JH, Kim E, Yoon G, Kim H. Deoxypodophyllotoxin Exerts Anti-Cancer Effects on Colorectal Cancer Cells Through Induction of Apoptosis and Suppression of Tumorigenesis. Int J Mol Sci 2019; 20:E2612. [PMID: 31141929 PMCID: PMC6601030 DOI: 10.3390/ijms20112612] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Revised: 05/24/2019] [Accepted: 05/24/2019] [Indexed: 02/01/2023] Open
Abstract
Deoxypodophyllotoxin (DPT) is a cyclolignan compound that exerts anti-cancer effects against various types of cancers. DPT induces apoptosis and inhibits the growth of breast, brain, prostate, gastric, lung, and cervical tumors. In this study, we sought to determine the effect of DPT on cell proliferation, apoptosis, motility, and tumorigenesis of three colorectal cancer (CRC) cell lines: HT29, DLD1, and Caco2. DPT inhibited the proliferation of these cells. Specifically, the compound-induced mitotic arrest in CRC cells by destabilizing microtubules and activating the mitochondrial apoptotic pathway via regulation of B-cell lymphoma 2 (Bcl-2) family proteins (increasing Bcl-2 associated X (BAX) and decreasing B-cell lymphoma-extra-large (Bcl-xL)) ultimately led to caspase-mediated apoptosis. In addition, DPT inhibited tumorigenesis in vitro, and in vivo skin xenograft experiments revealed that DPT significantly decreased tumor size and tumor weight. Taken together, our results suggest DPT to be a potent compound that is suitable for further exploration as a novel chemotherapeutic for human CRC.
Collapse
Affiliation(s)
- Chathurika D B Gamage
- College of Pharmacy and Research Institute of Life and Pharmaeutical Sciences, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Korea.
| | - So-Yeon Park
- College of Pharmacy and Research Institute of Life and Pharmaeutical Sciences, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Korea.
| | - Yi Yang
- College of Pharmacy and Research Institute of Life and Pharmaeutical Sciences, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Korea.
| | - Rui Zhou
- College of Pharmacy and Research Institute of Life and Pharmaeutical Sciences, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Korea.
| | - İsa Taş
- College of Pharmacy and Research Institute of Life and Pharmaeutical Sciences, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Korea.
| | - Woo Kyun Bae
- Department of Internal Medicine, Chonnam National University Medical School, 160 Baekseo-ro, Dong-gu, Gwangju 61469, Korea.
| | - Kyung Keun Kim
- Department of Pharmacology, Chonnam National University Medical School, 160 Baekseo-ro, Dong-gu, Gwangju 61469, Korea.
| | - Jung-Hyun Shim
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-ro, muan, Jeonnam 58554, Korea.
| | - Eunae Kim
- College of Pharmacy, Chosun University, 309 Philmun-daero, Dong-gu, Gwangju 61452, Korea.
| | - Goo Yoon
- College of Pharmacy and Natural Medicine Research Institute, Mokpo National University, 1666 Yeongsan-ro, muan, Jeonnam 58554, Korea.
| | - Hangun Kim
- College of Pharmacy and Research Institute of Life and Pharmaeutical Sciences, Sunchon National University, 255 Jungang-ro, Sunchon, Jeonnam 57922, Korea.
| |
Collapse
|
10
|
Zálešák F, Bon DJYD, Pospíšil J. Lignans and Neolignans: Plant secondary metabolites as a reservoir of biologically active substances. Pharmacol Res 2019; 146:104284. [PMID: 31136813 DOI: 10.1016/j.phrs.2019.104284] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 05/21/2019] [Accepted: 05/22/2019] [Indexed: 12/19/2022]
Abstract
Lignans and neolignans are plant secondary metabolites derived from the oxidative coupling of phenylpropanoids. Biological activity of these phenolic compounds ranges from antioxidant, antitumor (terminaloside P, IC50 = 10 nM), anti-inflammatory, anti-neurodegenerative (schibitubin B, IC50 = 3.2 nM) and antiviral (patentiflorin A, IC50 = 14-23 nM) to antimicrobial. In addition, it was observed that several members of this group, namely enterolactone and its biochemical precursors also known as phytoestrogens, possess important protective properties. Most of these lignans and neolignans are presented in reasonable amounts in one's diet and thus the protection they provide against the colon and breast cancer, to name a few, is even more important to note. Similarly, neuroprotective properties were observed (schisanwilsonin G, IC50 = 3.2 nM) These structural motives also serve as an important starting point in the development of anticancer drugs. Presumably the most famous members of this family, etoposide and teniposide, synthetic derivatives of podophyllotoxin, are used in the clinical treatment of lymphocytic leukemia, certain brain tumors, and lung tumors already for nearly 20 years. This review describes 413 lignans and neolignans which have been isolated between 2016 and mid-2018 being reported in more than 300 peer-reviewed articles. It covers their source, structure elucidation, and bioactivity. Within the review, the structure-based overview of compounds as well as the bioactivity-based overview of compounds are described.
Collapse
Affiliation(s)
- František Zálešák
- Department of Organic Chemistry, Faculty of Science, Palacky University, tř. 17. listopadu 1192/12, CZ-771 46 Olomouc, Czech Republic.
| | - David Jean-Yves Denis Bon
- Department of Organic Chemistry, Faculty of Science, Palacky University, tř. 17. listopadu 1192/12, CZ-771 46 Olomouc, Czech Republic.
| | - Jiří Pospíšil
- Department of Organic Chemistry, Faculty of Science, Palacky University, tř. 17. listopadu 1192/12, CZ-771 46 Olomouc, Czech Republic; Laboratory of Growth Regulators, The Czech Academy of Sciences, Institute of Experimental Botany & Palacký University, Šlechtitelů 27, CZ-78371 Olomouc, Czech Republic.
| |
Collapse
|
11
|
Scandenolone from Cudrania tricuspidata fruit extract suppresses the viability of breast cancer cells (MCF-7) in vitro and in vivo. Food Chem Toxicol 2019; 126:56-66. [PMID: 30753858 DOI: 10.1016/j.fct.2019.02.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 01/31/2019] [Accepted: 02/08/2019] [Indexed: 12/28/2022]
Abstract
Scandenolone, an isoflavone, has shown anti-cancer potential. In this study, we extracted scandenolone from Cudrania tricuspidata fruit and evaluated its anti-breast cancer effects as well as toxicity in cell and animal models. In cell model, scandenolone suppressed the breast cancer MCF-7 cells viability, ceased mitotic cell cycle, decreased mitochondrial membrane potential, up-regulated cleaved caspase-3 and promoted the phosphorylation of p53. Additionally, this isoflavone promoted cell apoptosis and induced a sustained activation of the phosphorylation of p38 and ERK, but not JNK and Akt. The effects were further verified in a human MCF-7 breast cancer xenograft model, where scandenolone efficiently suppressed the cancer growth and increased apoptotic cells in tumor tissue. However scandenolone has also shown certain toxicity to normal hepatocytes and breast epithelial cells. It could be concluded that scandenolone suppressed the growth of breast cancer cells, but its toxicity towards normal cells might limit its potential clinical use.
Collapse
|
12
|
Tavares WR, Seca AML. The Current Status of the Pharmaceutical Potential of Juniperus L. Metabolites. MEDICINES 2018; 5:medicines5030081. [PMID: 30065158 PMCID: PMC6165314 DOI: 10.3390/medicines5030081] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/16/2018] [Accepted: 07/20/2018] [Indexed: 12/27/2022]
Abstract
Background: Plants and their derived natural compounds possess various biological and therapeutic properties, which turns them into an increasing topic of interest and research. Juniperus genus is diverse in species, with several traditional medicines reported, and rich in natural compounds with potential for development of new drugs. Methods: The research for this review were based in the Scopus and Web of Science databases using terms combining Juniperus, secondary metabolites names, and biological activities. This is not an exhaustive review of Juniperus compounds with biological activities, but rather a critical selection taking into account the following criteria: (i) studies involving the most recent methodologies for quantitative evaluation of biological activities; and (ii) the compounds with the highest number of studies published in the last four years. Results: From Juniperus species, several diterpenes, flavonoids, and one lignan were emphasized taking into account their level of activity against several targets. Antitumor activity is by far the most studied, being followed by antibacterial and antiviral activities. Deoxypodophyllotoxin and one dehydroabietic acid derivative appears to be the most promising lead compounds. Conclusions: This review demonstrates the Juniperus species value as a source of secondary metabolites with relevant pharmaceutical potential.
Collapse
Affiliation(s)
- Wilson R Tavares
- Faculty of Sciences and Technology, University of Azores, 9501-801 Ponta Delgada, Portugal.
| | - Ana M L Seca
- Department of Chemistry & QOPNA-Organic Chemistry, Natural Products and Food Stuffs, University of Aveiro, Campus de Santiago, 3810-193 Aveiro, Portugal.
- cE3c-Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group & Faculty of Sciences and Technology, University of Azores, Rua Mãe de Deus, 9501-321 Ponta Delgada, Portugal.
| |
Collapse
|
13
|
Chen Y, Zhao K, Liu F, Li Y, Zhong Z, Hong S, Liu X, Liu L. Predicting Antitumor Effect of Deoxypodophyllotoxin in NCI-H460 Tumor-Bearing Mice on the Basis of In Vitro Pharmacodynamics and a Physiologically Based Pharmacokinetic-Pharmacodynamic Model. Drug Metab Dispos 2018; 46:897-907. [DOI: 10.1124/dmd.117.079830] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2017] [Accepted: 04/02/2018] [Indexed: 11/22/2022] Open
|
14
|
Zang X, Wang G, Cai Q, Zheng X, Zhang J, Chen Q, Wu B, Zhu X, Hao H, Zhou F. A Promising Microtubule Inhibitor Deoxypodophyllotoxin Exhibits Better Efficacy to Multidrug-Resistant Breast Cancer than Paclitaxel via Avoiding Efflux Transport. Drug Metab Dispos 2018. [DOI: 10.1124/dmd.117.079442] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
|