1
|
Khwaza V, Aderibigbe BA. Potential Pharmacological Properties of Triterpene Derivatives of Ursolic Acid. Molecules 2024; 29:3884. [PMID: 39202963 PMCID: PMC11356970 DOI: 10.3390/molecules29163884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 08/13/2024] [Accepted: 08/14/2024] [Indexed: 09/03/2024] Open
Abstract
Ursolic acid (UA) and its derivatives have garnered significant attention due to their extensive pharmacological activity. UA is a pentacyclic triterpenoid found in a variety of plants, such as apples, rosemary, thyme, etc., and it possesses a range of pharmacological properties. Researchers have synthesized various derivatives of UA through structural modifications to enhance its potential pharmacological properties. Various in vitro and in vivo studies have indicated that UA and its derivatives possess diverse biological activities, such as anticancer, antifungal, antidiabetic, antioxidant, antibacterial, anti-inflammatory and antiviral properties. This review article provides a review of the biological activities of UA and its derivatives to show their valuable therapeutic properties useful in the treatment of different diseases, mainly focusing on the relevant structure-activity relationships (SARs), the underlying molecular targets/pathways, and modes of action.
Collapse
Affiliation(s)
- Vuyolwethu Khwaza
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice 5700, Eastern Cape, South Africa
| | - Blessing A. Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice 5700, Eastern Cape, South Africa
| |
Collapse
|
2
|
Medeiros M, Guenka S, Bastos D, Oliveira KL, Brassesco MS. Amicis Omnia Sunt Communia: NF-κB Inhibition as an Alternative to Overcome Osteosarcoma Heterogeneity. Pharmaceuticals (Basel) 2024; 17:734. [PMID: 38931401 PMCID: PMC11206879 DOI: 10.3390/ph17060734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/28/2024] Open
Abstract
Tumor heterogeneity poses a significant challenge in osteosarcoma (OS) treatment. In this regard, the "omics" era has constantly expanded our understanding of biomarkers and altered signaling pathways (i.e., PI3K/AKT/mTOR, WNT/β-catenin, NOTCH, SHH/GLI, among others) involved in OS pathophysiology. Despite different players and complexities, many commonalities have been described, among which the nuclear factor kappa B (NF-κB) stands out. Its altered activation is pervasive in cancer, with pleiotropic action on many disease-relevant traits. Thus, in the scope of this article, we highlight the evidence of NF-κB dysregulation in OS and its integration with other cancer-related pathways while we summarize the repertoire of compounds that have been described to interfere with its action. In silico strategies were used to demonstrate that NF-κB is closely coordinated with other commonly dysregulated signaling pathways not only by functionally interacting with several of their members but also by actively participating in the regulation of their transcription. While existing inhibitors lack selectivity or act indirectly, the therapeutic potential of targeting NF-κB is indisputable, first for its multifunctionality on most cancer hallmarks, and secondly, because, as a common downstream effector of the many dysregulated pathways influencing OS aggressiveness, it turns complex regulatory networks into a simpler picture underneath molecular heterogeneity.
Collapse
Affiliation(s)
- Mariana Medeiros
- Cell Biology Department, Ribeirão Preto Medical School, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil;
| | - Sophia Guenka
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - David Bastos
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| | - Karla Laissa Oliveira
- Regional Blood Center, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14051-140, São Paulo, Brazil;
| | - María Sol Brassesco
- Biology Department, Faculty of Philosophy, Sciences and Letters at Ribeirão Preto, University of São Paulo, Avenida Bandeirantes, 3900-Vila Monte Alegre, Ribeirão Preto 14040-900, São Paulo, Brazil; (S.G.); (D.B.)
| |
Collapse
|
3
|
Wani AK, Singh R, Akhtar N, Prakash A, Nepovimova E, Oleksak P, Chrienova Z, Alomar S, Chopra C, Kuca K. Targeted Inhibition of the PI3K/Akt/mTOR Signaling Axis: Potential for Sarcoma Therapy. Mini Rev Med Chem 2024; 24:1496-1520. [PMID: 38265369 DOI: 10.2174/0113895575270904231129062137] [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: 07/06/2023] [Revised: 10/15/2023] [Accepted: 10/23/2023] [Indexed: 01/25/2024]
Abstract
Sarcoma is a heterogeneous group of malignancies often resistant to conventional chemotherapy and radiation therapy. The phosphatidylinositol-3-kinase/ protein kinase B /mammalian target of rapamycin (PI3K/Akt/mTOR) signaling pathway has emerged as a critical cancer target due to its central role in regulating key cellular processes such as cell growth, proliferation, survival, and metabolism. Dysregulation of this pathway has been implicated in the development and progression of bone sarcomas (BS) and soft tissue sarcomas (STS). PI3K/Akt/mTOR inhibitors have shown promising preclinical and clinical activity in various cancers. These agents can inhibit the activation of PI3K, Akt, and mTOR, thereby reducing the downstream signaling events that promote tumor growth and survival. In addition, PI3K/Akt/mTOR inhibitors have been shown to enhance the efficacy of other anticancer therapies, such as chemotherapy and radiation therapy. The different types of PI3K/Akt/mTOR inhibitors vary in their specificity, potency, and side effect profiles and may be effective depending on the specific sarcoma type and stage. The molecular targeting of PI3K/Akt/mToR pathway using drugs, phytochemicals, nanomaterials (NMs), and microbe-derived molecules as Pan-PI3K inhibitors, selective PI3K inhibitors, and dual PI3K/mTOR inhibitors have been delineated. While there are still challenges to be addressed, the preclinical and clinical evidence suggests that these inhibitors may significantly improve patient outcomes. Further research is needed to understand the potential of these inhibitors as sarcoma therapeutics and to continue developing more selective and effective agents to meet the clinical needs of sarcoma patients.
Collapse
Affiliation(s)
- Atif Khurshid Wani
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Reena Singh
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Nahid Akhtar
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Ajit Prakash
- Department of Biochemistry and Biophysics, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Eugenie Nepovimova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Patrik Oleksak
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Zofia Chrienova
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
| | - Suliman Alomar
- King Saud University, Zoology Department, College of Science, Riyadh, 11451, Saudi Arabia
| | - Chirag Chopra
- School of Bioengineering and Biosciences, Lovely Professional University, Jalandhar (144411), Punjab, India
| | - Kamil Kuca
- Department of Chemistry, Faculty of Science, University of Hradec Králové, Hradec Králové, Czechia
- Biomedical Research Center, University Hospital Hradec Kralove, Hradec Králové, Czechia
| |
Collapse
|
4
|
Zhao H, Tang S, Tao Q, Ming T, Lei J, Liang Y, Peng Y, Wang M, Liu M, Yang H, Ren S, Xu H. Ursolic Acid Suppresses Colorectal Cancer by Down-Regulation of Wnt/β-Catenin Signaling Pathway Activity. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:3981-3993. [PMID: 36826439 DOI: 10.1021/acs.jafc.2c06775] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
Overwhelming evidence points to an abnormally active Wnt/β-catenin signaling as a key player in colorectal cancer (CRC) pathogenesis. Ursolic acid (UA) is a pentacyclic triterpenoid that has been found in a broad variety of fruits, spices, and medicinal plants. UA has been shown to have potent bioactivity against a variety of cancers, including CRC, with the action mechanism obscure. Our study tried to learn more about the efficacy of UA on CRC and its functional mechanism amid the Wnt/β-catenin signaling cascade. We determined the efficacy of UA on CRC SW620 cells with respect to the proliferation, migration, clonality, apoptosis, cell cycle, and Wnt/β-catenin signaling cascade, with assessment of the effect of UA on normal colonic NCM460 cells. Also, the effects of UA on the tumor development, apoptosis, cell cycle, and Wnt/β-catenin signaling axis were evaluated after a subcutaneous SW620 xenograft tumor model was established in mice. In this work, we showed that UA drastically suppressed proliferation, migration, and clonality; induced apoptosis; and arrested the cell cycle at the G0/G1 phase of SW620 cells, without the influence on NCM460 cells, accompanied by weakened activity of the Wnt/β-catenin signaling pathway. Besides, UA markedly deterred the growth of the xenograft tumor, ameliorated pathological features, triggered apoptosis, and arrested the cell cycle in xenograft CRC tissue, by lessening the Wnt/β-catenin signaling cascade. Overall, UA may inhibit the malignant phenotype, induce apoptosis, and arrest the cell cycle of CRC, potentially by attenuating the Wnt/β-catenin signaling axis, providing insights into the mechanism for the potency of UA on CRC.
Collapse
Affiliation(s)
- Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shun Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Qiu Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Jiarong Lei
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuanjing Liang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Yuhui Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Minmin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| |
Collapse
|
5
|
Liao WL, Liu YF, Ying TH, Shieh JC, Hung YT, Lee HJ, Shen CY, Cheng CW. Inhibitory Effects of Ursolic Acid on the Stemness and Progression of Human Breast Cancer Cells by Modulating Argonaute-2. Int J Mol Sci 2022; 24:ijms24010366. [PMID: 36613808 PMCID: PMC9820512 DOI: 10.3390/ijms24010366] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/28/2022] Open
Abstract
The stemness and metastasis of cancer cells are crucial features in determining cancer progression. Argonaute-2 (AGO2) overexpression was reported to be associated with microRNA (miRNA) biogenesis, supporting the self-renewal and differentiation characteristics of cancer stem cells (CSCs). Ursolic acid (UA), a triterpene compound, has multiple biological functions, including anticancer activity. In this study, we find that UA inhibits the proliferation of MDA-MB-231 and MCF-7 breast cancer cell lines using the CCK-8 assay. UA induced a significant decrease in the fraction of CSC in which it was examined by changes in the expression of stemness biomarkers, including the Nanog and Oct4 genes. UA altered invasion and migration capacities by significant decreases in the levels of epithelial-to-mesenchymal transition (EMT) proteins of slug and vimentin. Furthermore, the co-reduction in oncogenic miRNA levels (miR-9 and miR-221) was a result of the down-modulation in AGO2 in breast cancer cells in vitro. Mechanically, UA increases PTEN expression to inactivate the FAK/PI3K/Akt/mTOR signaling pathway and the decreased level of c-Myc in quantitative RT-PCR and Western blot imaging analyses. Our current understanding of the anticancer potential of UA in interrupting between EMT programming and the state of CSC suggests that UA can contribute to improvements in the clinical practice of breast cancer.
Collapse
Affiliation(s)
- Wen-Ling Liao
- Graduate Institute of Integrated Medicine, China Medical University, Taichung 40433, Taiwan
- Center for Personalized Medicine, China Medical University Hospital, Taichung 40433, Taiwan
| | - Yu-Fan Liu
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Tsung-Ho Ying
- Department of Obstetrics and Gynecology, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
| | - Jia-Ching Shieh
- Department of Biomedical Sciences, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Yueh-Tzu Hung
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Huei-Jane Lee
- Department of Biochemistry, School of Medicine, College of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
| | - Chen-Yang Shen
- Institute of Biomedical Sciences, Academia Sinica, Taipei 11529, Taiwan
- Graduate Institute of Environmental Science, China Medical University, Taichung 40433, Taiwan
- Correspondence: (C.-Y.S.); (C.-W.C.); Tel.: +886-2-2789-9036 (C.-Y.S.); +886-4-2473-0022 (ext. 11677) (C.-W.C.); Fax: +886-2-2782-3047 (C.-Y.S.); +886-4-2372-3229 (C.-W.C.)
| | - Chun-Wen Cheng
- Institute of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 40201, Taiwan
- Correspondence: (C.-Y.S.); (C.-W.C.); Tel.: +886-2-2789-9036 (C.-Y.S.); +886-4-2473-0022 (ext. 11677) (C.-W.C.); Fax: +886-2-2782-3047 (C.-Y.S.); +886-4-2372-3229 (C.-W.C.)
| |
Collapse
|
6
|
Ursolic Acid Analogs as Potential Therapeutics for Cancer. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27248981. [PMID: 36558113 PMCID: PMC9785537 DOI: 10.3390/molecules27248981] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/13/2022] [Accepted: 12/14/2022] [Indexed: 12/23/2022]
Abstract
Ursolic acid (UA) is a pentacyclic triterpene isolated from a large variety of vegetables, fruits and many traditional medicinal plants. It is a structural isomer of Oleanolic Acid. The medicinal application of UA has been explored extensively over the last two decades. The diverse pharmacological properties of UA include anti-inflammatory, antimicrobial, antiviral, antioxidant, anti-proliferative, etc. Especially, UA holds a promising position, potentially, as a cancer preventive and therapeutic agent due to its relatively non-toxic properties against normal cells but its antioxidant and antiproliferative activities against cancer cells. Cell culture studies have shown interference of UA with multiple pharmacological and molecular targets that play a critical role in many cells signaling pathways. Although UA is considered a privileged natural product, its clinical applications are limited due to its low absorption through the gastro-intestinal track and rapid elimination. The low bioavailability of UA limits its use as a therapeutic drug. To overcome these drawbacks and utilize the importance of the scaffold, many researchers have been engaged in designing and developing synthetic analogs of UA via structural modifications. This present review summarizes the synthetic UA analogs and their cytotoxic antiproliferative properties reported in the last two decades.
Collapse
|
7
|
Chen L, Liu M, Yang H, Ren S, Sun Q, Zhao H, Ming T, Tang S, Tao Q, Zeng S, Meng X, Xu H. Ursolic acid inhibits the activation of smoothened-independent non-canonical hedgehog pathway in colorectal cancer by suppressing AKT signaling cascade. Phytother Res 2022; 36:3555-3570. [PMID: 35708264 DOI: 10.1002/ptr.7523] [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: 12/13/2021] [Revised: 04/19/2022] [Accepted: 04/23/2022] [Indexed: 11/07/2022]
Abstract
It is being brought to light that smoothened (SMO)-independent non-canonical Hedgehog signaling is associated with the pathogenesis of various cancers. Ursolic acid (UA), a pentacyclic triterpenoid present in many medicinal herbs, manifests potent effectiveness against multiple malignancies including colorectal cancer (CRC). In our previous study, UA was found to protect against CRC in vitro by suppression of canonical Hedgehog signaling cascade. Here, the influence of UA on SMO-independent non-canonical Hedgehog signaling in CRC was investigated in the present study, which demonstrated that UA hampered the proliferation and migration, induced the apoptosis of HCT-116hSMO- cells with SMO gene knockdown, accompanied by the augmented expression of the suppressor of fused (SUFU), and lessened levels of MYC (c-Myc), glioma-associated oncogene (GLI1) and Sonic Hedgehog (SHH), and lowered phosphorylation of protein kinase B (PKB, AKT), suggesting that UA diminished non-canonical Hedgehog signal transduction in CRC. In HCT-116hSMO- xenograft tumor, UA ameliorated the symptoms, impeded the growth and caused the apoptosis of CRC, with heightened SUFU expression, and abated levels of MYC, GLI1, and SHH, and mitigated phosphorylation of AKT, indicating that UA down-regulated non-canonical Hedgehog signaling cascade in CRC. Taken together, UA may alleviate CRC by suppressing AKT signaling-dependent activation of SMO-independent non-canonical Hedgehog pathway.
Collapse
Affiliation(s)
- Li Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Maolun Liu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Han Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shan Ren
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiang Sun
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hui Zhao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Tianqi Ming
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Shun Tang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Qiu Tao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Sha Zeng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xianli Meng
- State Key Laboratory of Southwestern Chinese Medicine Resources, Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Haibo Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Department of Pharmacology, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Li S, Wu R, Wang L, Dina Kuo HC, Sargsyan D, Zheng X, Wang Y, Su X, Kong AN. Triterpenoid ursolic acid drives metabolic rewiring and epigenetic reprogramming in treatment/prevention of human prostate cancer. Mol Carcinog 2022; 61:111-121. [PMID: 34727410 PMCID: PMC8665082 DOI: 10.1002/mc.23365] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/11/2021] [Accepted: 10/18/2021] [Indexed: 01/03/2023]
Abstract
Ursolic acid (UA) is a triterpenoid phytochemical with a strong anticancer effect. The metabolic rewiring, epigenetic reprogramming, and chemopreventive effect of UA in prostate cancer (PCa) remain unknown. Herein, we investigated the efficacy of UA in PCa xenograft, and its biological effects on cellular metabolism, DNA methylation, and transcriptomic using multi-omics approaches. The metabolomics was quantified by liquid-chromatography-mass spectrometry (LC-MS) while epigenomic CpG methylation in parallel with transcriptomic gene expression was studied by next-generation sequencing technologies. UA administration attenuated the growth of transplanted human VCaP-Luc cells in immunodeficient mice. UA regulated several cellular metabolites and metabolism-related signaling pathways including S-adenosylmethionine (SAM), methionine, glucose 6-phosphate, CDP-choline, phosphatidylcholine biosynthesis, glycolysis, and nucleotide sugars metabolism. RNA-seq analyses revealed UA regulated several signaling pathways, including CXCR4 signaling, cancer metastasis signaling, and NRF2-mediated oxidative stress response. Epigenetic reprogramming study with DNA Methyl-seq uncovered a list of differentially methylated regions (DMRs) associated with UA treatment. Transcriptome-DNA methylome correlative analysis uncovered a list of genes, of which changes in gene expression correlated with the promoter CpG methylation status. Altogether, our results suggest that UA regulates metabolic rewiring of metabolism including SAM potentially driving epigenetic CpG methylation reprogramming, and transcriptomic signaling resulting in the overall anticancer chemopreventive effect.
Collapse
Affiliation(s)
- Shanyi Li
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- International Center for Aging and Cancer, Hainan Medical University, Haikou, Hainan, China
| | - Renyi Wu
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Lujing Wang
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Graduate Program in Pharmaceutical Science, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Hsiao-Chen Dina Kuo
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Graduate Program in Pharmaceutical Science, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Davit Sargsyan
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
- Graduate Program in Pharmaceutical Science, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Xi Zheng
- Department of Chemical Biology, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Yujue Wang
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Xiaoyang Su
- Metabolomics Shared Resource, Rutgers Cancer Institute of New Jersey, New Brunswick, New Jersey, USA
- Department of Medicine, Rutgers-Robert Wood Johnson Medical School, New Brunswick, New Jersey, USA
| | - Ah-Ng Kong
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| |
Collapse
|
9
|
Tang Z, Dong H, Li T, Wang N, Wei X, Wu H, Liu Y, Wang W, Guo Z, Xiao X. The Synergistic Reducing Drug Resistance Effect of Cisplatin and Ursolic Acid on Osteosarcoma through a Multistep Mechanism Involving Ferritinophagy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:5192271. [PMID: 34970416 PMCID: PMC8714329 DOI: 10.1155/2021/5192271] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 11/03/2021] [Accepted: 11/29/2021] [Indexed: 12/13/2022]
Abstract
Increasing evidence suggests that traditional Chinese medicine strategies are obviously beneficial for cancer treatment, but scientific research on the underlying molecular mechanisms is lacking. We report that ursolic acid, a bioactive ingredient isolated from Radix Actinidiae chinensis, has strong antitumour effects on osteosarcoma cells. Functional studies showed that ursolic acid inhibited tumour cell proliferation and promoted the apoptosis of a variety of osteosarcoma cells. Ursolic acid had a synergistic cytotoxic effect with cisplatin on osteosarcoma cells. In a mouse osteosarcoma xenograft model, low-dose cisplatin combined with ursolic acid significantly reduced tumour growth. Notably, ursolic acid reversed weight loss in mice treated with cisplatin. Mechanistic studies showed that ursolic acid degraded ferritin by activating autophagy and induced intracellular overload of ferrous ions, leading to ferroptosis. In addition, ursolic acid enhanced the DNA-damaging effect of cisplatin on osteosarcoma cells. Taken together, these findings suggest that ursolic acid is a nontoxic adjuvant that may enhance the effectiveness of chemotherapy in osteosarcoma.
Collapse
Affiliation(s)
- Zhen Tang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hui Dong
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an, China
| | - Ning Wang
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Xinghui Wei
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Hao Wu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yichao Liu
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wei Wang
- Department of Immunology, State Key Laboratory of Cancer Biology, Fourth Military Medical University, Xi'an, China
| | - Zheng Guo
- Department of Orthopaedics, Tangdu Hospital, Fourth Military Medical University, Xi'an, China
| | - Xin Xiao
- Department of Orthopedics, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| |
Collapse
|
10
|
Fu D, Ni Z, Wu K, Cheng P, Ji X, Li G, Shang X. A novel redox-responsive ursolic acid polymeric prodrug delivery system for osteosarcoma therapy. Drug Deliv 2021; 28:195-205. [PMID: 33438472 PMCID: PMC7808744 DOI: 10.1080/10717544.2020.1870583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/29/2022] Open
Abstract
Ursolic acid (UA), found widely in nature, exerts effective anti-tumoral activity against various malignant tumors. However, the low water solubility and poor bioavailability of UA have greatly hindered its translation to the clinic. To overcome these drawbacks, a simple redox-sensitive UA polymeric prodrug was synthesized by conjugating UA to polyethylene glycol using a disulfide bond. This formulation can self-assemble into micelles (U-SS-M) in aqueous solutions to produce small size micelles (∼62.5 nm in diameter) with high drug loading efficiency (∼16.7%) that exhibit pH and reduction dual-sensitivity. The cell and animal studies performed using the osteosarcoma MG-63 cell line and MG-63 cancer xenograft mice as the model systems consistently confirmed that the U-SS-M formulation could significantly prolong the circulation in blood and favor accumulation in tumor tissue. Targeted accumulation allows the U-SS-M to be effectively internalized by cancer cells, where the rapid release of UA is favored by a glutathione-rich and acidic intracellular environment, and ultimately achieves potent antitumor efficacy.
Collapse
Affiliation(s)
- Daijie Fu
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Zhe Ni
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Kerong Wu
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Peng Cheng
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Xiaofeng Ji
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Guoyuan Li
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| | - Xifu Shang
- Department of Orthopedics, The First Affiliated Hospital of University of Science and Technology of China, Hefei, China
| |
Collapse
|
11
|
Song GR, Choi YJ, Park SJ, Shin S, Lee G, Choi HJ, Lee DY, Song GY, Oh S. Root Bark of Morus alba L. and Its Bioactive Ingredient, Ursolic Acid, Suppress the Proliferation of Multiple Myeloma Cells by Inhibiting Wnt/β-Catenin Pathway. J Microbiol Biotechnol 2021; 31:1559-1567. [PMID: 34584036 PMCID: PMC9706038 DOI: 10.4014/jmb.2109.09002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/22/2021] [Accepted: 09/23/2021] [Indexed: 12/15/2022]
Abstract
The root bark of Morus alba L. has cytotoxic activity against several types of cancer cells. However, little is known about its chemopreventive mechanisms and bioactive metabolites. In this study, we showed that M. alba L. root bark extracts (MRBE) suppressed β-catenin response transcription (CRT), which is aberrantly activated in various cancers, by promoting the degradation of β-catenin. In addition, MRBE repressed the expression of the β-catenin/T-cell factor (TCF)-dependent genes, cmyc and cyclin D1, thus inhibiting the proliferation of RPMI-8226 multiple myeloma (MM) cells. MRBE induced apoptosis in MM cells, as evidenced by the increase in the population of annexin VFITC- positive cells and caspase-3/7 activity. We identified ursolic acid in MRBE through LC/mass spectrum (MS) and observed that it also decreased intracellular β-catenin, c-myc, and cyclin D1 levels. Furthermore, it suppressed the proliferation of RPMI-8226 cells by stimulating cell cycle arrest and apoptosis. These findings suggest that MRBE and its active ingredient, ursolic acid, exert antiproliferative activity by promoting the degradation of β-catenin and may have significant chemopreventive potential against MM.
Collapse
Affiliation(s)
- Geu Rim Song
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
- Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea
| | - Yoon Jung Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Soo Jin Park
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Subeen Shin
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
- Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea
| | - Giseong Lee
- College of General Education, Kookmin University, Seoul 02707, Republic of Korea
| | - Hui Ji Choi
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Do Yup Lee
- Department of Agricultural Biotechnology, Center for Food and Bioconvergence, Research Institute for Agricultural and Life Sciences, Seoul National University, Seoul 08826, Republic of Korea
| | - Gyu-Yong Song
- College of Pharmacy, Chungnam National University, Daejeon 34134, Republic of Korea
| | - Sangtaek Oh
- Department of Bio and Fermentation Convergence Technology, Kookmin University, Seoul 02707, Republic of Korea
- Department of Interdisciplinary Program for Bio-Health Convergence, Kookmin University, Seoul 02707, Republic of Korea
| |
Collapse
|
12
|
Wu Y, Li Q, Liu Y, Li Y, Chen Y, Wu X, Liu X. Targeting hypoxia for sensitization of tumors to apoptosis enhancement through supramolecular biohybrid bacteria. Int J Pharm 2021; 605:120817. [PMID: 34166726 DOI: 10.1016/j.ijpharm.2021.120817] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/16/2021] [Accepted: 06/17/2021] [Indexed: 02/06/2023]
Abstract
Bacteria-driven drug-delivery systems have drawn considerable interests for their highly selective hypoxia-targeting and efficacy in tumor inhibition. For the first time, a supramolecular biohybrid bacterium (SA@HU) is constructed by coating attenuated Salmonella typhimurium (S. typhimurium ΔppGpp/Lux) with nanoassemblies. In addition, the host-guest inclusion complexes based on hydroxypropyl-β-cyclodextrin (HPCD) and amantadine (AMA) was developed to encapsulate the natural antineoplastic product, ursolic acid (UA). It is found that the drug-carried coating layer has no significant impact on the antitumor activity or tumor-targeting capacity of bacteria. Significant restraint of tumor progression is achieved by SA@HU due to the synergy of cellular immune activation and apoptosis enhancement. Most importantly, intravenous delivery of UA by this biohybrid vector can cause tumor lysis, as the bacteria-attracting nutrients beneficial for preferential accumulation of bacteria in tumor. The mutual promotion of bacteria and UA may also contribute to a superior anticancer effect. Hence, the SA@HU-based biotic/abiotic supramolecular therapeutic system represents a novel strategy for combined chemo-bacterial therapy.
Collapse
Affiliation(s)
- Yundi Wu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China
| | - Qiuwan Li
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yang Liu
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yuxuan Li
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Yinhua Chen
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China
| | - Xilong Wu
- School of Biomedical Engineering, State Key Laboratory of Marine Resource Utilization in South China Sea, Hainan University, Haikou 570228, China.
| | - Xiande Liu
- School of Life and Pharmaceutical Sciences, Hainan University, Haikou 570228, China.
| |
Collapse
|
13
|
Targeting the crosstalk between canonical Wnt/β-catenin and inflammatory signaling cascades: A novel strategy for cancer prevention and therapy. Pharmacol Ther 2021; 227:107876. [PMID: 33930452 DOI: 10.1016/j.pharmthera.2021.107876] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Accepted: 04/05/2021] [Indexed: 02/06/2023]
Abstract
Emerging scientific evidence indicates that inflammation is a critical component of tumor promotion and progression. Most cancers originate from sites of chronic irritation, infections and inflammation, underscoring that the tumor microenvironment is largely orchestrated by inflammatory cells and pro-inflammatory molecules. These inflammatory components are intimately involved in neoplastic processes which foster proliferation, survival, invasion, and migration, making inflammation the primary target for cancer prevention and treatment. The influence of inflammation and the immune system on the progression and development of cancer has recently gained immense interest. The Wnt/β-catenin signaling pathway, an evolutionarily conserved signaling strategy, has a critical role in regulating tissue development. It has been implicated as a major player in cancer development and progression with its regulatory role on inflammatory cascades. Many naturally-occurring and small synthetic molecules endowed with inherent anti-inflammatory properties inhibit this aberrant signaling pathway, making them a promising class of compounds in the fight against inflammatory cancers. This article analyzes available scientific evidence and suggests a crosslink between Wnt/β-catenin signaling and inflammatory pathways in inflammatory cancers, especially breast, gastrointestinal, endometrial, and ovarian cancer. We also highlight emerging experimental findings that numerous anti-inflammatory synthetic and natural compounds target the crosslink between Wnt/β-catenin pathway and inflammatory cascades to achieve cancer prevention and intervention. Current challenges, limitations, and future directions of research are also discussed.
Collapse
|
14
|
Zhang Y, Wang X. Targeting the Wnt/β-catenin signaling pathway in cancer. J Hematol Oncol 2020; 13:165. [PMID: 33276800 PMCID: PMC7716495 DOI: 10.1186/s13045-020-00990-3] [Citation(s) in RCA: 773] [Impact Index Per Article: 154.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Accepted: 11/02/2020] [Indexed: 12/16/2022] Open
Abstract
The aberrant Wnt/β-catenin signaling pathway facilitates cancer stem cell renewal, cell proliferation and differentiation, thus exerting crucial roles in tumorigenesis and therapy response. Accumulated investigations highlight the therapeutic potential of agents targeting Wnt/β-catenin signaling in cancer. Wnt ligand/ receptor interface, β-catenin destruction complex and TCF/β-catenin transcription complex are key components of the cascade and have been targeted with interventions in preclinical and clinical evaluations. This scoping review aims at outlining the latest progress on the current approaches and perspectives of Wnt/β-catenin signaling pathway targeted therapy in various cancer types. Better understanding of the updates on the inhibitors, antagonists and activators of Wnt/β-catenin pathway rationalizes innovative strategies for personalized cancer treatment. Further investigations are warranted to confirm precise and secure targeted agents and achieve optimal use with clinical benefits in malignant diseases.
Collapse
Affiliation(s)
- Ya Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.,School of medicine, Shandong University, Jinan, 250021, Shandong, China.,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China.,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,School of medicine, Shandong University, Jinan, 250021, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, the First Affiliated Hospital of Soochow University, Suzhou, 250021, China.
| |
Collapse
|
15
|
Smith DK, Hasanali SL, Wang J, Kallifatidis G, Morera DS, Jordan AR, Terris MK, Klaassen Z, Bollag R, Lokeshwar VB, Lokeshwar BL. Promotion of epithelial hyperplasia by interleukin-8-CXCR axis in human prostate. Prostate 2020; 80:938-949. [PMID: 32542667 PMCID: PMC8327464 DOI: 10.1002/pros.24026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 05/05/2020] [Indexed: 12/12/2022]
Abstract
BACKGROUND The clinical manifestation of benign prostatic hyperplasia (BPH) is causally linked to the inflammatory microenvironment and proliferation of epithelial and stromal cells in the prostate transitional zone. The CXC-chemokine interleukin-8 (IL-8) contributes to inflammation. We evaluated the expression of inflammatory cytokines in clinical specimens, primary cultures, and prostatic lineage cell lines. We investigated whether IL-8 via its receptor system (IL-8 axis) promotes BPH. METHODS The messenger RNA and protein expression of chemokines, including components of the IL-8 axis, were measured in normal prostate (NP; n = 7) and BPH (n = 21), urine (n = 24) specimens, primary cultures, prostatic lineage epithelial cell lines (NHPrE1, BHPrE1, BPH-1), and normal prostate cells (RWPE-1). The functional role of the IL-8 axis in prostate epithelial cell growth was evaluated by CRISPR/Cas9 gene editing. The effect of a combination with two natural compounds, oleanolic acid (OA) and ursolic acid (UA), was evaluated on the expression of the IL-8 axis and epithelial cell growth. RESULTS Among the 19 inflammatory chemokines and chemokine receptors we analyzed, levels of IL-8 and its receptors (CXCR1, CXCR2), as well as, of CXCR7, a receptor for CXCL12, were 5- to 25-fold elevated in BPH tissues when compared to NP tissues (P ≤ .001). Urinary IL-8 levels were threefold to sixfold elevated in BPH patients, but not in asymptomatic males and females with lower urinary tract symptoms (P ≤ .004). The expression of the IL-8 axis components was confined to the prostate luminal epithelial cells in both normal and BPH tissues. However, these components were elevated in BPH-1 and primary explant cultures as compared to RWPE-1, NHPrE1, and BHPrE1 cells. Knockout of CXCR7 reduced IL-8, and CXCR1 expression by 4- to 10-fold and caused greater than or equal to 50% growth inhibition in BPH-1 cells. Low-dose OA + UA combination synergistically inhibited the growth of BPH-1 and BPH primary cultures. In the combination, the drug reduction indices for UA and OA were 16.4 and 7852, respectively, demonstrating that the combination was effective in inhibiting BPH-1 growth at significantly reduced doses of UA or OA alone. CONCLUSION The IL-8 axis is a promotor of BPH pathogenesis. Low-dose OA + UA combination inhibits BPH cell growth by inducing autophagy and reducing IL-8 axis expression in BPH-epithelial cells.
Collapse
Affiliation(s)
- Diandra K. Smith
- Department of Medicine, Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia
| | - Sarrah L. Hasanali
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Jiaojiao Wang
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Georgios Kallifatidis
- Department of Medicine, Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia
- Department of Biological Sciences, College of Science and Mathematics, Augusta University, Augusta, Georgia
- Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia
| | - Daley S. Morera
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Andre R. Jordan
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Martha K. Terris
- Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia
- Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Zachary Klaassen
- Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Roni Bollag
- Department of Pathology, Bio-Repository Alliance of Georgia for Oncology (BRAG-Onc), Georgia Cancer Center, Augusta University, Augusta, Georgia
| | - Vinata B. Lokeshwar
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta University, Augusta, Georgia
- Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| | - Bal L. Lokeshwar
- Department of Medicine, Georgia Cancer Center, Medical College of Georgia, Augusta, Georgia
- Research Service, Charlie Norwood Veterans Administration Medical Center, Augusta, Georgia
- Division of Urology, Department of Surgery, Medical College of Georgia, Augusta University, Augusta, Georgia
| |
Collapse
|
16
|
Chen YM, Tang BX, Chen WY, Zhao MS. Ursolic acid inhibits the invasiveness of A498 cells via NLRP3 inflammasome activation. Oncol Lett 2020; 20:170. [PMID: 32934737 PMCID: PMC7471750 DOI: 10.3892/ol.2020.12027] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2020] [Accepted: 07/17/2020] [Indexed: 01/12/2023] Open
Abstract
Renal cell cancer is a common malignant tumor of the urinary system. Ursolic acid (UA) serves an important antitumor role in certain types of cancer, such as lung cancer, breast cancer and hepatocellular carcinoma; however, to the best of our knowledge, the effect of UA on renal cancer has not yet been investigated. In the present study, A498 cells were treated with different concentrations of UA for 12, 24 and 48 h, and then MCC950, an inhibitor of the NLR family pyrin domain-containing 3 (NLRP3) receptor, was added to block NLRP3 signaling. The proliferation of A498 cells was analyzed using an MTS assay and invasiveness was analyzed using a Transwell assay. The expression levels of NLRP3, cleaved caspase-1, IL-1β and MMP-2 were detected using western blotting. The present results demonstrated that the invasiveness of A498 cells was significantly decreased following UA treatment (P<0.05), while expression levels of NLRP3, cleaved caspase-1 and IL-1β were significantly increased, and MMP-2 expression was decreased following UA stimulation (P<0.05). This was reversed by MCC950 treatment (P<0.05), with the exception of NLRP3. In conclusion, the present results indicated that UA exposure decreased the proliferation and invasiveness of A498 cells. Additionally, UA exposure significantly decreased MMP-2 production and induced the activation of NLRP3 inflammasome, which was reversed by MCC950 treatment, indicating that NLRP3 activation may be involved in UA inhibition of A498 cell invasiveness.
Collapse
Affiliation(s)
- Yuan-Min Chen
- Department of Nephrology, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan 610000, P.R. China
| | - Bi-Xia Tang
- Department of Nephrology, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan 610000, P.R. China
| | - Wei-Yong Chen
- Department of Nephrology, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan 610000, P.R. China
| | - Ming-Sheng Zhao
- Department of Nephrology, The Seventh People's Hospital of Chengdu, Chengdu, Sichuan 610000, P.R. China
| |
Collapse
|
17
|
Khwaza V, Oyedeji OO, Aderibigbe BA. Ursolic Acid-Based Derivatives as Potential Anti-Cancer Agents: An Update. Int J Mol Sci 2020; 21:E5920. [PMID: 32824664 PMCID: PMC7460570 DOI: 10.3390/ijms21165920] [Citation(s) in RCA: 97] [Impact Index Per Article: 19.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Revised: 05/12/2020] [Accepted: 05/21/2020] [Indexed: 12/13/2022] Open
Abstract
Ursolic acid is a pharmacologically active pentacyclic triterpenoid derived from medicinal plants, fruit, and vegetables. The pharmacological activities of ursolic acid have been extensively studied over the past few years and various reports have revealed that ursolic acid has multiple biological activities, which include anti-inflammatory, antioxidant, anti-cancer, etc. In terms of cancer treatment, ursolic acid interacts with a number of molecular targets that play an essential role in many cell signaling pathways. It suppresses transformation, inhibits proliferation, and induces apoptosis of tumor cells. Although ursolic acid has many benefits, its therapeutic applications in clinical medicine are limited by its poor bioavailability and absorption. To overcome such disadvantages, researchers around the globe have designed and developed synthetic ursolic acid derivatives with enhanced therapeutic effects by structurally modifying the parent skeleton of ursolic acid. These structurally modified compounds display enhanced therapeutic effects when compared to ursolic acid. This present review summarizes various synthesized derivatives of ursolic acid with anti-cancer activity which were reported from 2015 to date.
Collapse
Affiliation(s)
| | | | - Blessing A. Aderibigbe
- Department of Chemistry, University of Fort Hare, Alice Campus, Alice 5700, Eastern Cape, South Africa; (V.K.); (O.O.O.)
| |
Collapse
|
18
|
Yang W, Chen X, Li Y, Guo S, Wang Z, Yu X. Advances in Pharmacological Activities of Terpenoids. Nat Prod Commun 2020. [DOI: 10.1177/1934578x20903555] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Terpenoids, the most abundant compounds in natural products, are a set of important secondary metabolites in plants with diverse structures. Terpenoids play key roles in plant growth and development, response to the environment, and physiological processes. As raw materials, terpenoids were also widely used in pharmaceuticals, food, and cosmetics industries. Terpenoids possess antitumor, anti-inflammatory, antibacterial, antiviral, antimalarial effects, promote transdermal absorption, prevent and treat cardiovascular diseases, and have hypoglycemic activities. In addition, previous studies have also found that terpenoids have many potential applications, such as insect resistance, immunoregulation, antioxidation, antiaging, and neuroprotection. Terpenoids have a complex structure with diverse effects and different mechanisms of action. Activities and mechanisms of terpenoids were reviewed in this paper. The development and application prospect of terpenoid compounds were also prospected, which provides a useful reference for new drug discovery and drug design based on terpenoids.
Collapse
Affiliation(s)
| | - Xu Chen
- School of Pharmacy, Linyi University, P. R. China
| | - Yanli Li
- School of Pharmacy, Linyi University, P. R. China
| | - Shaofen Guo
- School of Pharmacy, Linyi University, P. R. China
| | - Zhen Wang
- School of Pharmacy, Linyi University, P. R. China
| | - Xiuling Yu
- School of Pharmacy, Linyi University, P. R. China
| |
Collapse
|
19
|
Harikrishnan H, Jantan I, Alagan A, Haque MA. Modulation of cell signaling pathways by Phyllanthus amarus and its major constituents: potential role in the prevention and treatment of inflammation and cancer. Inflammopharmacology 2019; 28:1-18. [PMID: 31792765 DOI: 10.1007/s10787-019-00671-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022]
Abstract
The causal and functional connection between inflammation and cancer has become a subject of much research interest. Modulation of cell signaling pathways, such as those involving mitogen activated protein kinases (MAPKs), nuclear factor kappa β (NF-κB), phosphatidylinositol 3-kinase and protein kinase B (PI3K/Akt), and Wnt, and their outcomes play a fundamental role in inflammation and cancer. Activation of these cell signaling pathways can lead to various aspects of cancer-related inflammation. Hence, compounds able to modulate inflammation-related molecular targets are sought after in anticancer drug development programs. In recent years, plant extracts and their metabolites have been documented with potential in the prevention and treatment of cancer and inflammatory ailments. Plants possessing anticancer and anti-inflammatory properties due to their bioactive constituents have been reported to modulate the molecular and cellular pathways which are related to inflammation and cancer. In this review we focus on the flavonoids (astragalin, kaempferol, quercetin, rutin), lignans (phyllanthin, hypophyllanthin, and niranthin), tannins (corilagin, geraniin, ellagic acid, gallic acid), and triterpenes (lupeol, oleanolic acid, ursolic acid) of Phyllanthus amarus, which exert various anticancer and anti-inflammatory activities via perturbation of the NF-κB, MAPKs, PI3K/Akt, and Wnt signaling networks. Understanding the underlying mechanisms involved may help future research to develop drug candidates for prevention and new treatment for cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Hemavathy Harikrishnan
- Department of Pharmacology, School of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Ibrahim Jantan
- School of Pharmacy, Faculty of Health and Medical Sciences, Taylor's University, Lakeside Campus, 47500, Subang Jaya, Selangor, Malaysia. .,Institute of Systems Biology (INBIOSIS), Universiti Kebangsaan Malaysia, 43600 UKM, Bangi, Selangor, Malaysia.
| | - Akilandeshwari Alagan
- Crescent School of Pharmacy, B.S. Abdur Rahman Crescent Institute of Science and Technology, Chennai, 600 048, India
| | - Md Areeful Haque
- Department of Pharmacy, International Islamic University Chittagong, Chittagong, 4318, Bangladesh
| |
Collapse
|
20
|
Yang K, Chen Y, Zhou J, Ma L, Shan Y, Cheng X, Wang Y, Zhang Z, Ji X, Chen L, Dai H, Zhu B, Li C, Tao Z, Hu X, Yin W. Ursolic acid promotes apoptosis and mediates transcriptional suppression of CT45A2 gene expression in non-small-cell lung carcinoma harbouring EGFR T790M mutations. Br J Pharmacol 2019; 176:4609-4624. [PMID: 31322286 PMCID: PMC6965687 DOI: 10.1111/bph.14793] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/28/2019] [Accepted: 07/05/2019] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND AND PURPOSE In non-small-cell lung carcinoma (NSCLC) patients, the L858R/T790M mutation of the epithelial growth factor receptor (EGFR) is a major cause of acquired resistance to EGFR-TKIs treatment that limits their therapeutic efficacy. Identification of drugs that can preferentially kill the NSCLC harbouring L858R/T790M mutation is therefore critical. Here, we have evaluated the effects of ursolic acid, an active component isolated from herbal sources, on erlotinib-resistant H1975 cells that harbour the L858R/T790M mutation. EXPERIMENTAL APPROACH Gene expression omnibus (GEO) profiles analyses was applied to detect differentially expressed genes in NSCLC cells harbouring EGFR mutation. AnnexinV-FITC/PI, TUNEL staining, MTT, wound healing, RT-PCR, qRT-PCR, western blots, immunostaining, dual-luciferase reporters and ChIP-PCR were utilized to investigate the effects of ursolic acid in vitro and in vivo. KEY RESULTS The cancer/testis antigen family 45 member A2 (CT45A2) was highly expressed in H1975 cells. Ectopic expression of CT45A2 in H1975 cells increased cell proliferation and motility in vitro. Silencing the CT45A2 expression strongly attenuated H1975 cells motility and growth. The anti-cancer effect of ursolic acid was critically dependent on CT45A2 expression in H1975 cells. Ursolic acid suppressed CT45A2 gene transcription mediated by transcriptional factor TCF4 and β-catenin signalling. CONCLUSIONS AND IMPLICATIONS CT45A2 is a novel oncogene for NSCLC with an EGFR T790 mutation. Ursolic acid induced apoptosis and inhibited proliferation of H1975 cells by negatively regulating the β-catenin/TCF4/CT45A2 signalling pathway. Therefore, ursolic acid may be a potential candidate treatment for NSCLC harbouring the EGFR-L858R/T790M mutation.
Collapse
Affiliation(s)
- Kaiyong Yang
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Yan Chen
- Division of nutritionJiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer HospitalNanjingChina
| | - Jiaqian Zhou
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Lin Ma
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Yating Shan
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Xiaoying Cheng
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Yun Wang
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Zhaoxin Zhang
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Xiaojun Ji
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Lili Chen
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Hui Dai
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Biqing Zhu
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Chen Li
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| | - Zhonghua Tao
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xichun Hu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of Oncology, Shanghai Medical CollegeFudan UniversityShanghaiChina
| | - Wu Yin
- State Key Lab of Pharmaceutical Biotechnology, College of Life SciencesNanjing UniversityNanjingChina
| |
Collapse
|
21
|
Lin CW, Chin HK, Lee SL, Chiu CF, Chung JG, Lin ZY, Wu CY, Liu YC, Hsiao YT, Feng CH, Bai LY, Weng JR. Ursolic acid induces apoptosis and autophagy in oral cancer cells. ENVIRONMENTAL TOXICOLOGY 2019; 34:983-991. [PMID: 31062913 DOI: 10.1002/tox.22769] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2018] [Revised: 04/19/2019] [Accepted: 04/24/2019] [Indexed: 06/09/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the fifth common cause of cancer mortality in Taiwan with high incidence and recurrence and needs new therapeutic strategies. In this study, ursolic acid (UA), a triterpenoid, was examined the antitumor potency in OSCC cells. Our results showed that UA inhibited the proliferation of OSCC cells in a dose- and time-dependent manner in both Ca922 and SCC2095 oral cancer cells. UA induced caspase-dependent apoptosis accompanied with the modulation of various biological biomarkers including downregulating Akt/mTOR/NF-κB signaling, ERK, and p38. In addition, UA inhibited angiogenesis as evidenced by abrogation of migration/invasion and blocking MMP-2 secretion in Ca922 cells. Interestingly, UA induced autophagy in OSCC cells, as manifested by LC3B-II conversion and increased p62 expression and accumulation of autophagosomes. Inhibition by autophagy inhibitor enhanced UA-mediated apoptosis in Ca922 cells. The experiment provides a rationale for using triterpenoid in the treatment of OSCC.
Collapse
Affiliation(s)
- Cheng-Wen Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung, Taiwan
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
| | - Hsien-Kuo Chin
- Division of Cardiovascular Surgery, Department of Surgery, Kaohsiung Armed Forces General Hospital, Kaohsiung, Taiwan
| | - Shou-Lun Lee
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chang-Fang Chiu
- College of Medicine, China Medical University, Taichung, Taiwan
- Cancer Center, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Gung Chung
- Department of Biotechnology, Asia University, Wufeng, Taichung, Taiwan
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Zi-Yin Lin
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Yung Wu
- Cancer Center, China Medical University Hospital, Taichung, Taiwan
| | - Ying-Chen Liu
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Yung-Ting Hsiao
- Department of Biological Science and Technology, China Medical University, Taichung, Taiwan
| | - Chia-Hsien Feng
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Li-Yuan Bai
- College of Medicine, China Medical University, Taichung, Taiwan
- Division of Hematology and Oncology, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
| | - Jing-Ru Weng
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
- Graduate Institute of Pharmacognosy, College of Pharmacy, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
22
|
Knockdown of HOXB8 inhibits tumor growth and metastasis by the inactivation of Wnt/β-catenin signaling pathway in osteosarcoma. Eur J Pharmacol 2019; 854:22-27. [PMID: 30954562 DOI: 10.1016/j.ejphar.2019.04.004] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Revised: 03/29/2019] [Accepted: 04/02/2019] [Indexed: 01/08/2023]
Abstract
Homeobox B8 (HOXB8) is a member of HOX family and was reported to be dysregulated in human cancers. However, its expression pattern and function in human osteosarcoma (OS) remain unknown. The aim of the current study is to examine its expression and biological roles in human OS cells. Our results showed that HOXB8 was highly expressed in human OS tissues and cell lines. Knockdown of HOXB8 significantly suppressed the proliferation of OS cells in vitro and attenuated the tumor growth in a tumor xenograft model. In addition, knockdown of HOXB8 dramatically repressed the migration and invasion of OS cells. Furthermore, knockdown of HOXB8 efficiently prevented the activation of Wnt/β-catenin signaling pathway in OS cells. In conclusion, the findings of the present study demonstrated that knockdown of HOXB8 could suppress tumorigenesis and metastasis in OS through regulation of the Wnt/β-catenin signaling pathway. Thus, HOXB8 may represent a novel therapeutic target for the treatment of OS.
Collapse
|
23
|
Lee KC, Chen YL, Lin PY, Chuang WL. Ursolic Acid-Induced Apoptosis via Regulation of the PI3K/Akt and MAPK Signaling Pathways in Huh-7 Cells. Molecules 2018; 23:E2016. [PMID: 30104508 PMCID: PMC6222435 DOI: 10.3390/molecules23082016] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 08/02/2018] [Accepted: 08/10/2018] [Indexed: 01/11/2023] Open
Abstract
Ursolic acid (UA), is a kind of triterpene acid that exhibits wide biological properties. In this article, the effects of UA on apoptosis and the proliferation of human hepatoma Huh-7 cells were reported. The MTT results showed that cell viability of Huh-7 was reduced in a concentration and time-dependent effect. In addition, DAPI staining was used to detected condensation of chromatin in nucleus. Apoptotic cell population was examined using Annexin V/PI staining. The results showed that exposure to UA affected extrinsic and intrinsic pathways through, reduced expression of Bcl-2, Mcl-1, and TCTP; increased levels of the apoptotic proteins TNF-α, Fas, FADD, and Bax; and activation of cleaved caspase-3 and PARP. UA also inhibited the p-Akt and p38 MAPK signaling transduction pathways, and increased activity in the p-ERK signaling pathway. Taken together, UA inhibited the cell growth of Huh-7 cells and affected apoptosis, via regulated cellular signaling transduction.
Collapse
Affiliation(s)
- Kwong-Chiu Lee
- Department of Anesthesiology, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Yao-Li Chen
- Transplant Medicine & Surgery Research Center, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Department of Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan.
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Ping-Yi Lin
- Transplant Medicine & Surgery Research Center, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Wan-Ling Chuang
- Transplant Medicine & Surgery Research Center, Changhua Christian Hospital, Changhua 50006, Taiwan.
| |
Collapse
|
24
|
Knockdown of Sox2 Inhibits OS Cells Invasion and Migration via Modulating Wnt/β-Catenin Signaling Pathway. Pathol Oncol Res 2018; 24:907-913. [PMID: 29619662 DOI: 10.1007/s12253-018-0400-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2017] [Accepted: 12/15/2017] [Indexed: 12/14/2022]
Abstract
Osteosarcoma (OS) was a prevalent malignant bone tumor which threatens people's health worldwide. Wnt/β catenin signaling pathway had been proved significant in various cancers, indicating its possible function in OS as well. Sox2, a crucial member among SOX family could regulate cells biologically. How Sox2 modulated Wnt/β catenin signaling pathway in OS remained to be discussed. The study aimed to investigate the effects of Sox2 on the invasion and migration of OS cells and the related molecular mechanisms. Twenty-four human OS and adjacent tissue samples were involved in this study. Human OS cell lines MG63 and HOS were selected for further investigation. The liposome carrier si-Sox2 which could interfere with the expression of Sox2 gene was built to transfect MG63 and HOS cells). QRT-PCR assay and western blot were utilized to analyze the expression of mRNA and proteins of Sox2. Transwell assay and wound healing assay were conducted to test the invasion and migration level of cells. The expression of GSK3, β-catenin, cyclin D1 and c-myc proteins were detected by western blot assay after transfection with si-Sox2. Compared with normal tissues and cells, the expression of Sox2 in OS tissues and cells was significantly higher. The mRNA and protein levels of Sox2 significantly decreased after transfection with si-Sox2. The invasion and migration of OS cells were down-regulated significantly through the inhibition of Sox2 by inactivating Wnt/β-catenin signaling pathway related proteins. Knockdown of Sox2 could inhibit invasion and migration of OS cells via modulating Wnt/β-catenin signaling pathway.
Collapse
|
25
|
Li S, Dong Y, Wang K, Wang Z, Zhang X. Transcriptomic analyses reveal the underlying pro-malignant functions of PTHR1 for osteosarcoma via activation of Wnt and angiogenesis pathways. J Orthop Surg Res 2017; 12:168. [PMID: 29121993 PMCID: PMC5679487 DOI: 10.1186/s13018-017-0664-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 10/23/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Increasing evidence has indicated parathyroid hormone type 1 receptor (PTHR1) plays important roles for the development and progression of osteosarcoma (OS). However, its function mechanisms remain unclear. The goal of this study was to further illuminate the roles of PTHR1 in OS using microarray data. METHODS Microarray data were available from the Gene Expression Omnibus database under the accession number GSE46861, including six tumors from mice with PTHR1 knockdown (PTHR1.358) and six tumors from mice with control knockdown (Ren.1309). Differentially expressed genes (DEGs) between PTHR1.358 and Ren.1309 were identified using the LIMMA method, and then, protein-protein interaction (PPI) network was constructed using data from STRING database to screen crucial genes associated with PTHR1. KEGG pathway enrichment analysis was performed to investigate the underlying functions of DEGs using DAVID tool. RESULTS A total of 1163 genes were identified as DEGs, including 617 downregulated (Lef1, lymphoid enhancer-binding factor 1) and 546 upregulated genes (Dkk1, Dickkopf-related protein 1). KEGG enrichment analysis indicated upregulated DEGs were involved in Renin-angiotensin system (e.g., Agt, angiotensinogen) and Wnt signaling pathway (e.g., Dkk1), while downregulated DEGs participated in Basal cell carcinoma (e.g., Lef1). A PPI network (534 nodes and 2830 edges) was constructed, in which Agt gene was demonstrated to be the hub gene and its interactive genes (e.g., CCR3, CC chemokine receptor 3; and CCL9, chemokine CC chemokine ligand 9) were inflammation related. CONCLUSIONS Our present study preliminarily reveals the pro-malignant effects of PTHR1 in OS cells may be mediated by activating Wnt, angiogenesis, and inflammation pathways via changing the expressions of the crucial enriched genes (Dkk1, Lef1, Agt-CCR3, and Agt-CCL9).
Collapse
MESH Headings
- Animals
- Bone Neoplasms/genetics
- Bone Neoplasms/metabolism
- Gene Expression Profiling/methods
- Gene Regulatory Networks/genetics
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Neovascularization, Pathologic/genetics
- Neovascularization, Pathologic/metabolism
- Osteosarcoma/genetics
- Osteosarcoma/metabolism
- Receptor, Parathyroid Hormone, Type 1/biosynthesis
- Receptor, Parathyroid Hormone, Type 1/deficiency
- Receptor, Parathyroid Hormone, Type 1/genetics
- Wnt Signaling Pathway/physiology
Collapse
Affiliation(s)
- Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning, 110042, China
| | - Yujin Dong
- Department of Hand and Foot Surgery, Dalian Municipal Central Hospital Affiliated of Dalian Medical University, Dalian, Liaoning, 116033, China
| | - Ke Wang
- Molecular Pathology Testing Center, Foshan Chancheng Central Hospital, Foshan, Guangdong, 528031, China
| | - Zhe Wang
- Department of Orthopedics, Zhongshan Hospital Affiliated to Fudan University, Shanghai, 200032, China
| | - Xiaojing Zhang
- Department of Bone and Soft Tissue Tumor Surgery, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, 44, Xiaoheyan Road, Dadong District, Shenyang, Liaoning, 110042, China.
| |
Collapse
|
26
|
Baicalein inhibits progression of osteosarcoma cells through inactivation of the Wnt/β-catenin signaling pathway. Oncotarget 2017; 8:86098-86116. [PMID: 29156780 PMCID: PMC5689670 DOI: 10.18632/oncotarget.20987] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2017] [Accepted: 08/29/2017] [Indexed: 12/13/2022] Open
Abstract
Osteosarcoma is a very common type of malignant bone tumor in children and young adults and aberrant activation of Wnt/β-catenin signaling pathway has been discovered in osteosarcoma. The traditional Chinese medicine baicalein was proved to have anti-proliferative and anti-metastatic properties in osteosarcoma, but the mechanism remained poorly understood. In the present study, we assessed the effects of baicalein on osteosarcoma and detected the potential molecular mechanism. We found that baicalein significantly suppressed the proliferation of osteosarcoma cells in a concentration- and time-dependent manner. In additional, baicalein could induce apoptosis and cell cycle arrest and reduce cell motility. Moreover, the level of β-catenin and its target genes, including c-myc, cyclinD1, and survivin significantly decreased in baicalein-treated osteosarcoma cells, whereas exogenous expression of β-catenin could reverse the anti-proliferative and anti-metastatic effects of baicalein. Subsequently, we established a 143B xenograft tumor model and found that baicalein treatment significantly inhibited tumor growth accompanied with inhibiting Wnt/β-catenin pathway. Thus, these findings suggest that baicalein may be a potentially effective Chinese herbal medicine for therapeutics of osteosarcoma and Wnt/β-catenin signaling pathway may serve as an efficient molecular marker or predictive target for osteosarcoma.
Collapse
|
27
|
Jesus JA, Fragoso TN, Yamamoto ES, Laurenti MD, Silva MS, Ferreira AF, Lago JHG, Santos-Gomes G, Passero LFD. Therapeutic effect of ursolic acid in experimental visceral leishmaniasis. Int J Parasitol Drugs Drug Resist 2017; 7:1-11. [PMID: 27984757 PMCID: PMC5156607 DOI: 10.1016/j.ijpddr.2016.12.002] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2016] [Accepted: 12/01/2016] [Indexed: 12/14/2022]
Abstract
Leishmaniasis is an important neglected tropical disease, affecting more than 12 million people worldwide. The available treatments are not well tolerated and present diverse side effects in patients, justifying the search for new therapeutic compounds. In the present study, the therapeutic potential and toxicity of ursolic acid (UA), isolated from the leaves of Baccharis uncinella C. DC. (Asteraceae), were evaluated in experimental visceral leishmaniasis. To evaluate the therapeutic potential of UA, hamsters infected with L. (L.) infantum were treated daily during 15 days with 1.0 or 2.0 mg UA/kg body weight, or with 5.0 mg amphotericin B/kg body weight by intraperitoneal route. Fifteen days after the last dose, the parasitism of the spleen and liver was stimated and the main histopathological alterations were recorded. The proliferation of splenic mononuclear cells was evaluated and IFN-γ, IL-4, and IL-10 gene expressions were analyzed in spleen fragments. The toxicity of UA and amphotericin B were evaluated in healthy golden hamsters by histological analysis and biochemical parameters. Animals treated with UA had less parasites in the spleen and liver when compared with the infected control group, and they also showed preservation of white and red pulps, which correlate with a high rate of proliferation of splenic mononuclear cells, IFN-γ mRNA and iNOS production. Moreover, animals treated with UA did not present alterations in the levels of AST, ALT, creatinine and urea. Taken together, these findings indicate that UA is an interesting natural compound that should be considered for the development of prototype drugs against visceral leishmaniasis.
Collapse
Affiliation(s)
- Jéssica A Jesus
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, Av. Dr. Arnaldo, 455. Cerqueira César, São Paulo, 01246-903, SP, Brazil; Center of Natural Sciences and Humanities, Federal University of ABC, Santo Andre, São Paulo, 09210-180, Brazil
| | - Thais N Fragoso
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, Av. Dr. Arnaldo, 455. Cerqueira César, São Paulo, 01246-903, SP, Brazil
| | - Eduardo S Yamamoto
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, Av. Dr. Arnaldo, 455. Cerqueira César, São Paulo, 01246-903, SP, Brazil
| | - Márcia D Laurenti
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, Av. Dr. Arnaldo, 455. Cerqueira César, São Paulo, 01246-903, SP, Brazil
| | - Marcelo S Silva
- Global Health and Tropical Medicine, Instituto de Higiene e Medicina Tropical (IHMT), Universidade Nova de Lisboa, Rua da Junqueira 100, 1349-008 Lisboa, Portugal; Departamento de Análises Clínicas e Toxicológicas, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Rua General Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil
| | - Aurea F Ferreira
- Laboratory of Pathology of Infectious Diseases (LIM50), Department of Pathology, Medical School of São Paulo University, Av. Dr. Arnaldo, 455. Cerqueira César, São Paulo, 01246-903, SP, Brazil
| | - João Henrique G Lago
- Center of Natural Sciences and Humanities, Federal University of ABC, Santo Andre, São Paulo, 09210-180, Brazil
| | - Gabriela Santos-Gomes
- Departamento de Análises Clínicas e Toxicológicas, Centro de Ciências da Saúde, Universidade Federal do Rio Grande do Norte, Rua General Gustavo Cordeiro de Farias, 384, 59012-570 Natal, Brazil
| | - Luiz Felipe D Passero
- São Paulo State University (Unesp), Institute of Biosciences, São Vicente, Praça Infante Dom Henrique, s/n, 11330-900 São Vicente, SP, Brazil.
| |
Collapse
|
28
|
Zhou T, Liang Y, Jiang L, Yu T, Zeng C, Tao E. Mild hypothermia protects against oxygen glucose deprivation/reoxygenation-induced apoptosis via the Wnt/β-catenin signaling pathway in hippocampal neurons. Biochem Biophys Res Commun 2017; 486:1005-1013. [PMID: 28365156 DOI: 10.1016/j.bbrc.2017.03.153] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 01/31/2023]
Abstract
Mild hypothermia is thought to be one of the most effective therapies for cerebral ischemia/reperfusion injuries. Our previous research revealed that mild hypothermia inhibits the activation of caspase-3 and protects against oxygen glucose deprivation/reoxygenation (OGD/R)-induced injury in hippocampal neurons. However, the mechanisms behind the activation of caspase-3 remain unclear. The aims of this study were to determine whether the protective effects of mild hypothermia were exerted through the Wnt/β-catenin signaling pathway. We found that, under OGD/R conditions, the pathway was down regulated, but mild hypothermia induced the reactivation of the Wnt/β-catenin signaling pathway, which had been suppressed by OGD/R injury. Mild hypothermia also caused the down regulation of the expression of apoptosis promoting proteins (Bax cleaved caspase-3), up-regulated the expression of apoptosis inhibiting proteins (Bcl-2), and ameliorated OGD/R injury-induced apoptosis. The protective effects of mild hypothermia were blocked by DKK1 (an antagonist of the canonical Wnt signaling pathway). Taken together, these results indicate that the Wnt/β-catenin signaling pathway mediates the protective effects of mild hypothermia against OGD/R-induced apoptosis. Our study provides evidence that mild hypothermia reactivates the Wnt/β-catenin signaling pathway, which is suppressed by OGD/R injury, in hippocampal neurons and protects neurons from OGD/R-induced apoptosis via the reactivation of the Wnt/β-catenin signaling pathway, ultimately suggesting that mild hypothermia could have therapeutic effects on OGD/R-induced apoptosis.
Collapse
Affiliation(s)
- Tianen Zhou
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Yanran Liang
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Longyuan Jiang
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Tao Yu
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Chaotao Zeng
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China
| | - Enxiang Tao
- Department of Neurology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510120, China.
| |
Collapse
|
29
|
Li Y, Guo W, Liu S, Zhang B, Yu BB, Yang B, Kan SL, Feng SQ. Silencing Transmembrane Protein 45B (TNEM45B) Inhibits Proliferation, Invasion, and Tumorigenesis in Osteosarcoma Cells. Oncol Res 2016; 25:1021-1026. [PMID: 28244852 PMCID: PMC7841085 DOI: 10.3727/096504016x14821477992177] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Transmembrane protein 45B (TMEM45B) is a member of the TMEM family of proteins and has been reported to be expressed abnormally in different kinds of human tumors. However, the biological function of TMEM45B in osteosarcoma remains unclear. The objective of this study was to investigate the role of TMEM45B in regulating the biological behavior of osteosarcoma cells. Our results demonstrated that the expression of TMEM45B at both the protein and mRNA levels was dramatically upregulated in human osteosarcoma cell lines. Knockdown of TMEM45B significantly suppressed the proliferation, migration, and invasion of U2OS cells in vitro. Mechanistically, knockdown of TMEM45B sharply downregulated the expression level of β-catenin, cyclin D1, and c-Myc in U2OS cells. Finally, knockdown of TMEM45B attenuated tumor growth in transplanted U2OS-derived tumors in nude mice. Taken together, our results demonstrated that TMEM45B plays an important role in regulating the proliferation, migration, and invasion of osteosarcoma cells and that its effects on proliferation and invasion were mediated partially through the Wnt/β-catenin signaling pathway. These observations support our belief that TMEM45B may serve as an oncogene in the development and progression of osteosarcoma.
Collapse
|