1
|
Han J, Shen Y, Cao R, Wang W, Duan J, Duan J, Bao C. Active herbal ingredients and drug delivery design for tumor therapy: a review. Chin J Nat Med 2024; 22:1134-1162. [PMID: 39725513 DOI: 10.1016/s1875-5364(24)60741-7] [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: 04/13/2024] [Indexed: 12/28/2024]
Abstract
Active herbal ingredients are gaining recognition for their potent anti-tumor efficacy, attributable to various mechanisms including tumor cell inhibition, immune system activation, and tumor angiogenesis inhibition. Recent studies have revealed that numerous anti-tumor herbal ingredients, such as ginsenosides, ursolic acid, oleanolic acid, and Angelica sinensis polysaccharides, can be utilized to develop smart drug carriers like liposomes, micelles, and nanoparticles. These carriers can deliver active herbal ingredients and co-deliver anti-tumor drugs to enhance drug accumulation at tumor sites, thereby improving anti-tumor efficacy. This study provides a comprehensive analysis of the mechanisms by which these active herbal ingredients-derived carriers enhance therapeutic outcomes. Additionally, it highlights the structural properties of these active herbal ingredients, demonstrating how their unique features can be strategically employed to design smart drug carriers with improved anti-tumor efficacy. The insights presented aim to serve as a reference and guide future innovations in the design and application of smart drug carriers for cancer therapy that leverage active herbal ingredients.
Collapse
Affiliation(s)
- Jing Han
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Yanxi Shen
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Ruiying Cao
- School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Weiren Wang
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jinao Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jialun Duan
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| | - Chunjie Bao
- National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Jiangsu Province Key Laboratory of High Technology Research, Nanjing University of Chinese Medicine, Nanjing 210023, China; School of Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China.
| |
Collapse
|
2
|
Ghosh S, Das SK, Sinha K, Ghosh B, Sen K, Ghosh N, Sil PC. The Emerging Role of Natural Products in Cancer Treatment. Arch Toxicol 2024; 98:2353-2391. [PMID: 38795134 DOI: 10.1007/s00204-024-03786-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/08/2024] [Indexed: 05/27/2024]
Abstract
The exploration of natural products as potential agents for cancer treatment has garnered significant attention in recent years. In this comprehensive review, we delve into the diverse array of natural compounds, including alkaloids, carbohydrates, flavonoids, lignans, polyketides, saponins, tannins, and terpenoids, highlighting their emerging roles in cancer therapy. These compounds, derived from various botanical sources, exhibit a wide range of mechanisms of action, targeting critical pathways involved in cancer progression such as cell proliferation, apoptosis, angiogenesis, and metastasis. Through a meticulous examination of preclinical and clinical studies, we provide insights into the therapeutic potential of these natural products across different cancer types. Furthermore, we discuss the advantages and challenges associated with their use in cancer treatment, emphasizing the need for further research to optimize their efficacy, pharmacokinetics, and delivery methods. Overall, this review underscores the importance of natural products in advancing cancer therapeutics and paves the way for future investigations into their clinical applications.
Collapse
Affiliation(s)
- Sumit Ghosh
- Department of Zoology, Ramakrishna Mission Vidyamandira, Belur Math, Howrah, 711202, India
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India
| | - Sanjib Kumar Das
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India
| | - Krishnendu Sinha
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India.
| | - Biswatosh Ghosh
- Department of Zoology, Bidhannagar College, Kolkata, 700064, India
| | - Koushik Sen
- Department of Zoology, Jhargram Raj College, Jhargram, 721507, India
| | - Nabanita Ghosh
- Department of Zoology, Maulana Azad College, Kolkata, 700013, India
| | - Parames C Sil
- Division of Molecular Medicine, Bose Institute, Kolkata, 700054, India.
| |
Collapse
|
3
|
Younis SS, Salama AM, Elmehy DA, Heabah NA, Rabah HM, Elakshar SH, Awad RA, Gamea GA. Trichinella spiralis Larval Extract as a Biological Anti-Tumor Therapy in a Murine Model of Ehrlich Solid Carcinoma. Parasite Immunol 2024; 46:e13035. [PMID: 38712475 DOI: 10.1111/pim.13035] [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: 09/07/2023] [Revised: 03/27/2024] [Accepted: 04/04/2024] [Indexed: 05/08/2024]
Abstract
Trichinella spiralis (T. spiralis) is an immunomodulating parasite that can adversely affect tumor growth and extend host lifespan. The aim of this study was to elucidate the mechanisms by which T. spiralis larval antigens achieve this effect using Ehrlich solid carcinoma (ESC) murine model. Assessment was done by histopathological and immunohistochemical analysis of caspase-3, TNF-α, Ki-67 and CD31. Additionally, Bcl2 and Bcl2-associated protein X (Bax) relative gene expression was assessed by molecular analysis for studying the effect of T. spiralis crude larval extract (CLE) antigen on tumor necrosis, apoptosis, cell proliferation and angiogenesis. We found that both T. spiralis infection and CLE caused a decrease in the areas of necrosis in ESC. Moreover, they led to increased apoptosis through activation of caspase-3, up-regulation of pro-apoptotic gene, Bax and down-regulation of anti-apoptotic gene, Bcl2. Also, T. spiralis infection and CLE diminished ESC proliferation, as evidenced by decreasing Ki-67. T. spiralis infection and CLE were able to suppress the development of ESC by inhibiting tumor proliferation, inducing apoptosis and decreasing tumor necrosis, with subsequent decrease in tumor metastasis. T. spiralis CLE antigen may be considered as a promising complementary immunotherapeutic agent in the treatment of cancer.
Collapse
Affiliation(s)
- Salwa S Younis
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Amina M Salama
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Dalia A Elmehy
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Nehal A Heabah
- Department of Pathology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hanem M Rabah
- Department of Medical Biochemistry, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Sara H Elakshar
- Department of Clinical Oncology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Radwa A Awad
- Department of Clinical Oncology, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Ghada A Gamea
- Department of Medical Parasitology, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
4
|
Elgendy DI, Elmahy RA, Amer AIM, Ibrahim HA, Eltantawy AF, Mansour FR, Salama AM. Efficacy of artemether against toxocariasis in mice: parasitological and immunopathological changes in brain, liver, and lung. Pathog Glob Health 2024; 118:47-64. [PMID: 37978995 PMCID: PMC10769145 DOI: 10.1080/20477724.2023.2285182] [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] [Indexed: 11/19/2023] Open
Abstract
Toxocariasis is a zoonosis that represents a serious threat to public health particularly in tropical and subtropical areas. Currently, albendazole, the most effective drug for treating visceral toxocariasis, shows moderate efficacy against the larvae in tissues and has some adverse effects. Artemether is an antiparasitic drug mainly used in the treatment of malaria and showed effectiveness against numerous helminthic infections. Besides, it possesses potent anti-inflammatory, antiapoptotic, antifibrotic, and neuroprotective properties. Thus, the study's aim was to investigate artemether's effects in comparison with albendazole on the therapeutic outcome of experimental toxocariasis. For this aim, 140 laboratory-bred mice were divided into four main groups: uninfected control, treatment control, albendazole-treated, and artemether-treated groups. The treatment regimens were started at the 15th dpi (early treatment), and at the 35th dpi (late treatment). The effectiveness of treatment was determined by brain larval count, histopathological, immunohistochemical, and biochemical examination. Artemether showed more effectiveness than albendazole in reducing brain larval counts, markers of brain injury including NF-κB, GFAP, and caspase-3, the diameter and number of hepatic granulomas, hepatic oxidative stress, hepatic IL-6, and TG2 mRNA, and pulmonary inflammation and fibrosis. The efficacy of artemether was the same when administered early or late in the infection. Finally, our findings illustrated that artemether might be a promising therapy for T. canis infection and it could be a good substitution for albendazole in toxocariasis treatment.
Collapse
Affiliation(s)
- Dina I. Elgendy
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Rasha A. Elmahy
- Zoology Department, Faculty of Science, Tanta University, Tanta, Egypt
| | | | - Hoda A. Ibrahim
- Medical Biochemistry Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Asmaa Fawzy Eltantawy
- Medical Pharmacology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Fotouh Rashed Mansour
- Pharmaceutical Analytical Chemistry Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Amina M. Salama
- Medical Parasitology Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| |
Collapse
|
5
|
Othman AM, Abdel-Rahman N, Denewer M, Eissa LA. Sinapic acid and 3,3′-diindolylmethane potentiate cyclophosphamide antitumor activity through induction of apoptosis and inhibition of metastasis. Int Immunopharmacol 2023; 118:110074. [PMID: 36989898 DOI: 10.1016/j.intimp.2023.110074] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Revised: 03/08/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023]
Abstract
AIM New therapeutic strategies are required to enhance the anticancer efficacy of chemotherapeutic drugs and to reduce their cytotoxicity. The purpose of this study was to assess the anti-tumor, antimetastatic and anti-apoptotic activities of sinapic acid (SA) and 3,3'-diindolylmethane (DIM) in solid Ehrlich carcinoma (SEC) induced in mice and combining SA or DIM compounds with cyclophosphamide (CYP). METHODS For induction of solid tumor, the right hind limbs of mice were inoculated subcutaneously with Ehrlich carcinoma cells. After 5 days of tumor inoculation, mice were treated with SA (56 mg/kg), DIM (40 mg/kg), CYP (10 mg/kg), and their combinations (SA/CYP) and (SA/DIM) for 21 days. The mRNA levels of Elabela, Serpina3, caspase-3, MMP-2 and MMP-9 were assessed by qPCR. Tumor and liver tissues were stained with hematoxylin and eosin for histological examination. Serum was investigated for ALT and AST activities. MAIN FINDINGS Treatment of SEC mice with SA and DIM significantly reduced solid tumor weight by 45.6% and 33.2%, respectively. They also reduced tumor size and increased life span of SEC mice. SA and DIM diminished area of metastatic nodules of tumor cells in the liver by 54.1% and 47.4%, respectively. They also reduced serum aminotransferases activities. Both SA and DIM were found to upregulate caspase 3 and downregulate MMP-2 and MMP-9. Furthermore, SA and DIM reduced gene expression of Elabela by (44.8% and 35.1%) and Serpina3 by (30.7% and 23.5%), respectively. SA and DIM were also shown to potentiate the anti-tumor activity CYP. SA and DIM showed promising antitumor effects and enhanced CYP antitumor activity mostly through upregulation of apoptotic caspase 3 and suppressing metastatic enzymes MMP-2 and MMP-9. Additionally, SA and DIM exhibited a hepatoprotective effect. Our results suggest that these natural compounds may be used to improve the efficacy and reduce the adverse effects of chemotherapeutic drugs in the treatment of solid malignancies.
Collapse
|
6
|
Kornel A, Nadile M, Retsidou MI, Sakellakis M, Gioti K, Beloukas A, Sze NSK, Klentrou P, Tsiani E. Ursolic Acid against Prostate and Urogenital Cancers: A Review of In Vitro and In Vivo Studies. Int J Mol Sci 2023; 24:ijms24087414. [PMID: 37108576 PMCID: PMC10138876 DOI: 10.3390/ijms24087414] [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: 02/15/2023] [Revised: 04/07/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
Prostate cancer is the second most diagnosed form of cancer in men worldwide and accounted for roughly 1.3 million cases and 359,000 deaths globally in 2018, despite all the available treatment strategies including surgery, radiotherapy, and chemotherapy. Finding novel approaches to prevent and treat prostate and other urogenital cancers effectively is of major importance. Chemicals derived from plants, such as docetaxel and paclitaxel, have been used in cancer treatment, and in recent years, research interest has focused on finding other plant-derived chemicals that can be used in the fight against cancer. Ursolic acid, found in high concentrations in cranberries, is a pentacyclic triterpenoid compound demonstrated to have anti-inflammatory, antioxidant, and anticancer properties. In the present review, we summarize the research studies examining the effects of ursolic acid and its derivatives against prostate and other urogenital cancers. Collectively, the existing data indicate that ursolic acid inhibits human prostate, renal, bladder, and testicular cancer cell proliferation and induces apoptosis. A limited number of studies have shown significant reduction in tumor volume in animals xenografted with human prostate cancer cells and treated with ursolic acid. More animal studies and human clinical studies are required to examine the potential of ursolic acid to inhibit prostate and other urogenital cancers in vivo.
Collapse
Affiliation(s)
- Amanda Kornel
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Matteo Nadile
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Maria Ilektra Retsidou
- Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Minas Sakellakis
- Department of Medical Oncology, Metropolitan Hospital, 18547 Athens, Greece
| | - Katerina Gioti
- Department of Biomedical Sciences, School of Health Sciences, University of West Attica, 12243 Athens, Greece
| | - Apostolos Beloukas
- Department of Biomedical Sciences, School of Health Sciences, University of West Attica, 12243 Athens, Greece
- National AIDS Reference Centre of Southern Greece, School of Public Health, University of West Attica, 11521 Athens, Greece
| | - Newman Siu Kwan Sze
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Panagiota Klentrou
- Department of Kinesiology, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| | - Evangelia Tsiani
- Department of Health Sciences, Faculty of Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
- Centre for Bone and Muscle Health, Applied Health Sciences, Brock University, St. Catharines, ON L2S 3A1, Canada
| |
Collapse
|
7
|
Ismail CA, Eissa MM, Gaafar MR, Younis LK, El Skhawy N. Toxoplasma gondii-derived antigen modifies tumor microenvironment of Ehrlich solid carcinoma murine model and enhances immunotherapeutic activity of cyclophosphamide. Med Oncol 2023; 40:136. [PMID: 37014499 PMCID: PMC10073061 DOI: 10.1007/s12032-023-01994-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Accepted: 03/08/2023] [Indexed: 04/05/2023]
Abstract
Pathogen-based cancer vaccine is a promising immunotherapeutic weapon to stimulate cancer immunosuppressive state. Toxoplasma gondii is a potent immunostimulant, and low-dose infection was linked to cancer resistance. Our goal was to evaluate the therapeutic antineoplastic activity of autoclaved Toxoplasma vaccine (ATV) against Ehrlich solid carcinoma (ESC) in mice in reference to and in combination with low-dose cyclophosphamide (CP), a cancer immunomodulator. Mice inoculation with ESC was followed by applying different treatment modalities including ATV, CP, and CP/ATV. We evaluated the impact of the different treatments on liver enzymes and pathology, tumor weight, volume, and histopathological changes. Using immunohistochemistry, we evaluated CD8+ T cell, FOXP3+ Treg, CD8+/Treg outside and inside ESC, and angiogenesis. Results showed significant tumor weights and volumes reduction with all treatments with 13.3% inhibition of tumor development upon combined CP/ATV use. Significant necrosis and fibrosis were noted in ESC by all treatments with improved hepatic functions versus non-treated control. Although ATV was almost equivalent to CP in tumor gross and histopathology, it promoted an immunostimulatory activity with significant Treg cells depletion outside ESC and CD8+ T cells infiltration inside ESC with higher CD8+ T/Treg ratio inside ESC superior to CP. Combined with CP, ATV exhibited significant synergistic immunotherapeutic and antiangiogenic action compared to either treatment alone with significant Kupffer cells hyperplasia and hypertrophy. Exclusively, therapeutic antineoplastic and antiangiogenic activity of ATV against ESC was verified that boosted CP immunomodulatory action which highlights a novel biological cancer immunotherapeutic vaccine candidate.
Collapse
Affiliation(s)
- Cherine A Ismail
- Department of Clinical Pharmacology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Maha M Eissa
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Maha R Gaafar
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Layla K Younis
- Department of Pathology, Faculty of Medicine, Alexandria University, Alexandria, Egypt
| | - Nahla El Skhawy
- Department of Medical Parasitology, Faculty of Medicine, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
8
|
Sandhu SS, Rouz SK, Kumar S, Swamy N, Deshmukh L, Hussain A, Haque S, Tuli HS. Ursolic acid: a pentacyclic triterpenoid that exhibits anticancer therapeutic potential by modulating multiple oncogenic targets. Biotechnol Genet Eng Rev 2023:1-31. [PMID: 36600517 DOI: 10.1080/02648725.2022.2162257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Accepted: 12/20/2022] [Indexed: 01/06/2023]
Abstract
The world is currently facing a global challenge against neoplastic diseases. Chemotherapy, hormonal therapy, surgery, and radiation therapy are some approaches used to treat cancer. However, these treatments are frequently causing side effects in patients, such as multidrug resistance, fever, weakness, and allergy, among others side effects. As a result, current research has focused on phytochemical compounds isolated from plants to treat deadly cancers. Plants are excellent resources of bioactive molecules, and many natural molecules have exceptional anticancer properties. They produce diverse anticancer derivatives such as alkaloids, terpenoids, flavonoids, pigments, and tannins, which have powerful anticancer activities against various cancer cell lines and animal models. Because of their safety, eco-friendly, and cost-effective nature, research communities have recently focused on various phytochemical bioactive molecules. Ursolic acid (UA) and its derivative compounds have anti-inflammatory, anticancer, apoptosis induction, anti-carcinogenic, and anti-breast cancer proliferation properties. Ursolic acid (UA) can improve the clinical management of human cancer because it inhibits cancer cell viability and proliferation, preventing tumour angiogenesis and metastatic activity. Therefore, the present article focuses on numerous bioactivities of Ursolic acid (UA), which can inhibit cancer cell production, mechanism of action, and modulation of anticancer properties via regulating various cellular processes.
Collapse
Affiliation(s)
| | - Sharareh Khorami Rouz
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Suneel Kumar
- Bio-Design Innovation Centre, Rani Durgavati University, Jabalpur, India
| | - Nitin Swamy
- Fungal Biotechnology and Invertebrate Pathology Laboratory Department of Biological Sciences, Rani Durgavati University, Jabalpur, India
| | - Loknath Deshmukh
- School of Life and Allied Science, ITM University, Raipur, India
| | - Arif Hussain
- School of Life Sciences, Manipal Academy of Higher Education, Dubai, United Arab Emirates
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan, Saudi Arabia
- Arabia and Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana, India
| |
Collapse
|
9
|
Chu C, Song K, Zhang Y, Yang M, Fan B, Huang H, Chen G. Biotransformation of ursolic acid by Circinella muscae and their anti-neuroinflammatory activities of metabolites. Nat Prod Res 2022; 36:2777-2782. [PMID: 33977841 DOI: 10.1080/14786419.2021.1925893] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 10/21/2022]
Abstract
In this study, the biotransformation of ursolic acid by Circinella muscae CGMCC 3.2695 was investigated. Scaled-up biotransformation reactions yielded ten metabolites. Their structures were established based on extensive NMR and HR-ESI-MS data analyses, and four of them are new compounds. C. muscae could selectively catalyze hydroxylation, lactonisation, carbonylation and carboxyl reduction reactions. Furthermore, all the identified metabolites were evaluated for their anti-neuroinflammatory activities in LPS-induced BV-2 cells. Most metabolites displayed pronounced inhibitory effect on nitric oxide (NO) production. The results suggested that biotransformed derivatives of ursolic acid might be served as potential neuroinflammatory inhibitors.
Collapse
Affiliation(s)
- Chengjiao Chu
- School of Pharmacy, Nantong University, Nantong, China
| | - Kainan Song
- School of Pharmacy, Nantong University, Nantong, China
| | | | - Min Yang
- School of Pharmacy, Nantong University, Nantong, China
| | - Boyi Fan
- School of Pharmacy, Nantong University, Nantong, China
| | - Huilian Huang
- Key Laboratory of Modern Preparation of TCM, Jiangxi University of Traditional Chinese Medicine, ministry of education, Nanchang, China
| | | |
Collapse
|
10
|
El-Salam MA, Samy G, Bastos J, Metwaly H. Novel antitumor activity of the combined treatment of galloylquinic acids from Copaifera lucens and doxorubicin in solid Ehrlich carcinoma-bearing mice via the modulation of the Notch signaling pathway. Life Sci 2022; 299:120497. [PMID: 35339508 DOI: 10.1016/j.lfs.2022.120497] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 03/11/2022] [Accepted: 03/19/2022] [Indexed: 02/07/2023]
Abstract
AIMS This study aims to investigate the potential synergistic effect of the combined treatment of galloylquinic acids compounds from Copaifera lucens with doxorubicin via the modulation of the Notch pathway in Ehrlich carcinoma-bearing mice model. MAIN METHODS The solid tumor model was induced in mice by s.c. injection of Ehrlich cancerous cells in the right hind limb. Sixty mice were allocated into five different groups which included treated groups with galloylquinic acids compounds, doxorubicin and their combination. Normal and tumor control groups were also used. Different biological samples were collected to measure the levels of Notch1, Hes1, Jagged1, TNF-α, IL-6, and VEGF. Histopathological and immunohistochemical examinations of tumor tissues using specific anti-NF-kβ and anti-cyclin D1 antibodies were also performed. KEY FINDINGS Our results showed that the combined treatment of galloylquinic acids compounds with doxorubicin significantly inhibited Notch1, Hes1, Jagged1, TNF-α, IL-6, VEGF, NF-kβ, and cyclin D1 activities. SIGNIFICANCE Galloylquinic acids compounds exhibited promising synergistic chemotherapeutic and oncostatic effects and promoted the chemosensitivity of doxorubicin, mainly by inhibiting the Notch signaling pathway and its downstream effectors. These compounds may be considered in cancer therapy exhibiting improved efficacy and reduced side effects of chemotherapeutic agents.
Collapse
Affiliation(s)
- Mohamed Abd El-Salam
- Department of Pharmacognosy, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Egypt; Institute for Research in Biomedicine (IRB Barcelona), Barcelona Institute of Science and Technology (BIST), 08028 Barcelona, Spain; Department of Medicine, Harvard Medical School, Boston, 02115, MA, USA; Department of Medicine, VA Boston Healthcare System, Boston, MA 02132, USA.
| | - Ghada Samy
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, 11152, Egypt
| | - Jairo Bastos
- Department of Pharmaceutical Sciences, School of Pharmaceutical Sciences of Ribeirão Preto, University of São Paulo, 14040-900 Ribeirão Preto, São Paulo, Brazil
| | - Heba Metwaly
- Department of Pharmaceutical Biochemistry, Faculty of Pharmacy, Alexandria University, 21500 Alexandria, Egypt.
| |
Collapse
|
11
|
Rawat L, Nayak V. Piperlongumine induces ROS mediated apoptosis by transcriptional regulation of SMAD4/P21/P53 genes and synergizes with doxorubicin in osteosarcoma cells. Chem Biol Interact 2022; 354:109832. [PMID: 35085581 DOI: 10.1016/j.cbi.2022.109832] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 01/09/2022] [Accepted: 01/21/2022] [Indexed: 01/21/2023]
Abstract
Piperlongumine is a herbal drug, with well-known anti-microbial and anti-neoplastic properties. The anti-carcinogenic potential of piperlongumine has been extensively explored for breast, colorectal, lungs, pancreatic, prostate, and oral carcinoma. However, a few numbers of studies are available on its bio-activity in osteosarcoma. Therefore, the present study aimed at exploring the therapeutic potential and possible mechanisms of action of piperlongumine in three human osteosarcoma cell lines in-vitro. The cytotoxicity of piperlongumine was determined by MTT assay, which shows dose and time-dependent inhibition of MG-63, 143B and KHOS/NP cells. Piperlongumine arrest the cells in G2/M phase of cell cycle and increases reactive oxygen species production, which possibly leads to lethal oxidative stress and apoptosis. Piperlongumine treatment significantly upregulated the expression of genes BAX, P21, P53, and SMAD4; while the BCL-2, SURVIVIN, TNFA, and NFKB genes expression was found down-regulated. Furthermore, piperlongumine exposure inhibited the migration of osteosarcoma cells as the expression of migration marker genes CDH2, CTNNB1, FN1, and TWIST were found to be down-regulated. The drug combination studies show the synergistic effect of piperlongumine with the conventional chemotherapeutic drug doxorubicin in osteosarcoma cells. Taken together, the above results suggest that PL displays anticancer properties against osteosarcoma and can be used as a therapeutic agent for osteosarcoma treatment in clinical settings.
Collapse
Affiliation(s)
- Laxminarayan Rawat
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, K.K. Birla Goa Campus, NH-17B, Zuarinagar, Goa, 403726, India.
| | - Vijayashree Nayak
- Department of Biological Sciences, Birla Institute of Technology and Science, Pilani, K.K. Birla Goa Campus, NH-17B, Zuarinagar, Goa, 403726, India.
| |
Collapse
|
12
|
Therapeutic Potential of Certain Terpenoids as Anticancer Agents: A Scoping Review. Cancers (Basel) 2022; 14:cancers14051100. [PMID: 35267408 PMCID: PMC8909202 DOI: 10.3390/cancers14051100] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 01/19/2022] [Accepted: 02/05/2022] [Indexed: 02/01/2023] Open
Abstract
Cancer is a life-threatening disease and is considered to be among the leading causes of death worldwide. Chemoresistance, severe toxicity, relapse and metastasis are the major obstacles in cancer therapy. Therefore, introducing new therapeutic agents for cancer remains a priority to increase the range of effective treatments. Terpenoids, a large group of secondary metabolites, are derived from plant sources and are composed of several isoprene units. The high diversity of terpenoids has drawn attention to their potential anticancer and pharmacological activities. Some terpenoids exhibit an anticancer effect by triggering various stages of cancer progression, for example, suppressing the early stage of tumorigenesis via induction of cell cycle arrest, inhibiting cancer cell differentiation and activating apoptosis. At the late stage of cancer development, certain terpenoids are able to inhibit angiogenesis and metastasis via modulation of different intracellular signaling pathways. Significant progress in the identification of the mechanism of action and signaling pathways through which terpenoids exert their anticancer effects has been highlighted. Hence, in this review, the anticancer activities of twenty-five terpenoids are discussed in detail. In addition, this review provides insights on the current clinical trials and future directions towards the development of certain terpenoids as potential anticancer agents.
Collapse
|
13
|
Antiangiogenic Phytochemicals Constituent of Diet as Promising Candidates for Chemoprevention of Cancer. Antioxidants (Basel) 2022; 11:antiox11020302. [PMID: 35204185 PMCID: PMC8868078 DOI: 10.3390/antiox11020302] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/28/2022] [Accepted: 01/29/2022] [Indexed: 12/04/2022] Open
Abstract
Despite the extensive knowledge on cancer nature acquired over the last years, the high incidence of this disease evidences a need for new approaches that complement the clinical intervention of tumors. Interestingly, many types of cancer are closely related to dietary habits associated with the Western lifestyle, such as low fruit and vegetable intake. Recent advances around the old-conceived term of chemoprevention highlight the important role of phytochemicals as good candidates for the prevention or treatment of cancer. The potential to inhibit angiogenesis exhibited by many natural compounds constituent of plant foods makes them especially interesting for their use as chemopreventive agents. Here, we review the antitumoral potential, with a focus on the antiangiogenic effects, of phenolic and polyphenolic compounds, such as quercetin or myricetin; terpenoids, such as ursolic acid or kahweol; and anthraquinones from Aloe vera, in different in vitro and in vivo assays, and the available clinical data. Although clinical trials have failed to assess the preventive role of many of these compounds, encouraging preclinical data support the efficacy of phytochemicals constituent of diet in the prevention and treatment of cancer, but a deeper understanding of their mechanisms of action and better designed clinical trials are urgently needed.
Collapse
|
14
|
The ponatinib/gossypol novel combination provides enhanced anticancer activity against murine solid Ehrlich carcinoma via triggering apoptosis and inhibiting proliferation/angiogenesis. Toxicol Appl Pharmacol 2021; 432:115767. [PMID: 34699866 DOI: 10.1016/j.taap.2021.115767] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 10/17/2021] [Accepted: 10/18/2021] [Indexed: 11/20/2022]
Abstract
The search for new antitumor agents or combinations that are more effective and, hopefully, provide fewer health hazards is ongoing. Therefore, this study investigated the efficacy of a novel combination of ponatinib, a multi-targeted tyrosine kinase inhibitor, and the natural phytochemical gossypol against murine solid Ehrlich carcinoma. Six groups of ten mice each received vehicle (I), ponatinib in doses of 10 and 15 mg/kg (II, III) respectively, gossypol in a dose of 4 mg/kg (IV), and ponatinib (10 or 15 mg/kg) in combination with gossypol (4 mg/kg; V, VI). All treatments started on the 12th post-Ehrlich ascites carcinoma (EAC) implantation day and were administered intraperitoneally in daily doses for 3 weeks. Treatment of EAC-bearing mice with ponatinib/gossypol combination improved anticancer efficacy over either drug alone, as demonstrated by greater decreases in tumor weight and volume, and ponatinib (10 mg/kg)/gossypol combination was more efficient than ponatinib (15 mg/kg). Mechanistically, the ponatinib/gossypol combination significantly increased apoptotic markers p53, Bax, and caspase-9 while decreasing anti-apoptotic marker Bcl-2. Furthermore, it greatly decreased proliferative and angiogenic markers, FGFR4 and VEGF, respectively. Histopathology revealed a significant decline in neoplastic cells, the majority of which have necrotic changes and numerous apoptotic bodies, as well as a decrease in mitotic figures and tumor giant cells, indicating the capacity to suppress cancer proliferation/persistence. Overall, gossypol could be used as an adjuvant medication for ponatinib in cancer treatment, possibly leading to successful dose reductions and fewer side effects; however, further research is needed before a clinical application could be feasible.
Collapse
|
15
|
Li R, Song X, Guo Y, Song P, Duan D, Chen ZS. Natural Products: A Promising Therapeutics for Targeting Tumor Angiogenesis. Front Oncol 2021; 11:772915. [PMID: 34746014 PMCID: PMC8570131 DOI: 10.3389/fonc.2021.772915] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Accepted: 10/04/2021] [Indexed: 12/22/2022] Open
Abstract
Tumor-associated angiogenesis is a key target for anti-cancer therapy. The imbalance between pro-angiogenic and anti-angiogenic signals elicited by tumor cells or tumor microenvironment always results in activating "angiogenic switch". Tumor angiogenesis functions in multi-aspects of tumor biology, including endothelial cell apoptosis, tumor metastasis, and cancer stem cell proliferation. Numerous studies have indicated the important roles of inexpensive and less toxic natural products in targeting tumor angiogenesis-associated cytokines and apoptotic signaling pathways. Our current knowledge of tumor angiogenesis is based mainly on experiments performed on cells and animals, so we summarized the well-established models for angiogenesis both in vitro and in vivo. In this review, we classified and summarized the anti-angiogenic natural agents (Polyphenols, Polysaccharides, Alkaloids, Terpenoids, Saponins) in targeting various tumor types according to their chemical structures at present, and discussed the mechanistic principles of these natural products on regulating angiogenesis-associated cytokines and apoptotic signaling pathways. This review is to help understanding the recent progress of natural product research for drug development on anti-tumor angiogenesis.
Collapse
Affiliation(s)
- Ruyi Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xin Song
- School of Life Science and Engineering, Lanzhou University of Technology, Lanzhou, China
| | - Yanan Guo
- Research Center of Traditional Chinese Medicine in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, China.,Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine in Gansu Province, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Peng Song
- Research Center of Traditional Chinese Medicine in Gansu Province, Gansu University of Chinese Medicine, Lanzhou, China.,Key Laboratory of Prevention and Treatment for Chronic Diseases by Traditional Chinese Medicine in Gansu Province, Affiliated Hospital of Gansu University of Chinese Medicine, Lanzhou, China
| | - Dongzhu Duan
- Shaanxi Key Laboratory of Phytochemistry and College of Chemistry & Chemical Engineering, Baoji University of Arts and Sciences, Baoji, China
| | - Zhe-Sheng Chen
- College of Pharmacy and Health Sciences, St. John's University, Queens, NY, United States
| |
Collapse
|
16
|
Malík M, Velechovský J, Tlustoš P. Natural pentacyclic triterpenoid acids potentially useful as biocompatible nanocarriers. Fitoterapia 2021; 151:104845. [PMID: 33684460 DOI: 10.1016/j.fitote.2021.104845] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 01/29/2021] [Accepted: 01/30/2021] [Indexed: 12/14/2022]
Abstract
The importance of natural raw materials has grown recently because of their ready availability, renewable nature, biocompatibility and controllable degradability. One such group of plant-derived substances includes the triterpenoid acids, terpenic compounds consisting of six isoprene units, a carboxyl group and other functional groups producing various isomers. Most can be easily extracted from different parts of the plant and modified successfully. By themselves or as aglycones (genins) of triterpene saponins, they have potentially useful pharmaceutical activity. This review focuses on the supramolecular properties of triterpenoid acids with regard to their subsequent use as biocompatible nanocarriers. The review also considers the current list of pentacyclic triterpene acids for which molecular self-assembly has been confirmed without the need for structural modification.
Collapse
Affiliation(s)
- Matěj Malík
- Department of Agroenvironmental Chemistry and Plant Nutrition, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Praha 6 - Suchdol, Czech Republic.
| | - Jiří Velechovský
- Department of Agroenvironmental Chemistry and Plant Nutrition, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Praha 6 - Suchdol, Czech Republic.
| | - Pavel Tlustoš
- Department of Agroenvironmental Chemistry and Plant Nutrition, Faculty of Agrobiology, Food and Natural Resources, Czech University of Life Sciences Prague, Kamýcká 129, 165 00 Praha 6 - Suchdol, Czech Republic.
| |
Collapse
|
17
|
Abildgaard C, Rizza S, Christiansen H, Schmidt S, Dahl C, Abdul-Al A, Christensen A, Filomeni G, Guldberg P. Screening of metabolic modulators identifies new strategies to target metabolic reprogramming in melanoma. Sci Rep 2021; 11:4390. [PMID: 33623106 PMCID: PMC7902673 DOI: 10.1038/s41598-021-83796-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 01/22/2021] [Indexed: 12/13/2022] Open
Abstract
The prognosis of metastatic melanoma remains poor due to de novo or acquired resistance to immune and targeted therapies. Previous studies have shown that melanoma cells have perturbed metabolism and that cellular metabolic pathways represent potential therapeutic targets. To support the discovery of new drug candidates for melanoma, we examined 180 metabolic modulators, including phytochemicals and anti-diabetic compounds, for their growth-inhibitory activities against melanoma cells, alone and in combination with the BRAF inhibitor vemurafenib. Two positive hits from this screen, 4-methylumbelliferone (4-MU) and ursolic acid (UA), were subjected to validation and further characterization. Metabolic analysis showed that 4-MU affected cellular metabolism through inhibition of glycolysis and enhanced the effect of vemurafenib to reduce the growth of melanoma cells. In contrast, UA reduced mitochondrial respiration, accompanied by an increase in the glycolytic rate. This metabolic switch potentiated the growth-inhibitory effect of the pyruvate dehydrogenase kinase inhibitor dichloroacetate. Both drug combinations led to increased production of reactive oxygen species, suggesting the involvement of oxidative stress in the cellular response. These results support the potential use of metabolic modulators for combination therapies in cancer and may encourage preclinical validation and clinical testing of such treatment strategies in patients with metastatic melanoma.
Collapse
Affiliation(s)
- Cecilie Abildgaard
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
- Department of Clinical Genetics, Lillebaelt Hospital - University Hospital of Southern Denmark, Vejle, Denmark
| | - Salvatore Rizza
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Helle Christiansen
- Lundbeckfonden Center of Excellence NanoCAN, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Steffen Schmidt
- Lundbeckfonden Center of Excellence NanoCAN, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Molecular Oncology, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
- Roche Innovation Center Copenhagen, Hørsholm, Denmark
| | - Christina Dahl
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Ahmad Abdul-Al
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Annette Christensen
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark
| | - Giuseppe Filomeni
- Redox Biology Group, Danish Cancer Society Research Center, Copenhagen, Denmark
- Department of Biology, Tor Vergata University of Rome, Rome, Italy
- Center for Healthy Aging, Copenhagen University, Copenhagen, Denmark
| | - Per Guldberg
- Molecular Diagnostics Group, Danish Cancer Society Research Center, Strandboulevarden 49, 2100, Copenhagen, Denmark.
- Department of Cancer and Inflammation Research, Institute for Molecular Medicine, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
18
|
Barua A, Choudhury P, Mandal S, Panda CK, Saha P. Anti-Metastatic Potential of a Novel Xanthone Sourced by Swertia chirata Against In Vivo and In Vitro Breast Adenocarcinoma Frameworks. Asian Pac J Cancer Prev 2020; 21:2865-2875. [PMID: 33112542 PMCID: PMC7798162 DOI: 10.31557/apjcp.2020.21.10.2865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The Anticancer property of Swertia chirata has been well established. It forms a rich source of compounds to which its anticancer property can be attributed, among the compounds found in S. chirata xanthones form an important group. Among the most abundant xanthones found in S. chirata, 1,5,8-trihydroxy-3-methoxy xanthone (TMX) was found to be most effective. As metastasis is the underlying cause of most cancer-related deaths, in this study, we evaluated the anti-metastatic potential of TMX against adenocarcinoma both in vivo and in vitro. MATERIALS AND METHODS In vivo anti-metastatic potential was proved by histological evidence of different organs, giemsa staining of bone marrow, subcutaneous re-injection of the aberrant bone marrow cells into the right flank of the mice to observe the formation of tumors and analyzing the markers related to metastasis by immunohistochemistry (IHC) and western blot. In vitro validation of anti-metastatic potential was carried out against human breast adenocarcinoma cell line MCF-7 by primarily analyzing the migratory property of cells through scratch wound healing assay and the ability of cells to form colonies. The re-validation part was performed by western blot of markers related to metastasis and real-time analysis of EMT related markers. RESULTS In vivo, TMX treatment restricted metastasis of EAC induced solid tumor to liver, lung, bone marrow, and validation of this finding was achieved by down regulation of metastatic and EMT markers. In vitro, TMX treatment restricted migratory and colony forming ability of MCF-7 cells by down regulating metastatic and EMT markers. CONCLUSION It was proved from our study that TMX treatment successfully reduced the metastatic potential of EAC induced solid tumor, with in vitro validation TMX on the MCF-7 cell line.
Collapse
Affiliation(s)
- Atish Barua
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, West Bengal, India
| | - Pritha Choudhury
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, West Bengal, India
| | - Suvra Mandal
- National Research Institute of Ayurvedic Drug Development, 4 Minerva Road, CN Block, Sector V, Bidhannagar, Kolkata, West Bengal, India
| | - Chinmay Kumar Panda
- Department of Oncogenne regulation, Chittaranjan National Cancer Institute, West Bengal, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, West Bengal, India
| |
Collapse
|
19
|
Ruthenium(II)/(III) DMSO-Based Complexes of 2-Aminophenyl Benzimidazole with In Vitro and In Vivo Anticancer Activity. Molecules 2020; 25:molecules25184284. [PMID: 32962014 PMCID: PMC7570852 DOI: 10.3390/molecules25184284] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 09/11/2020] [Indexed: 12/11/2022] Open
Abstract
New anticancer ruthenium(II/III) complexes [RuCl2(DMSO)2(Hapbim)] (1) and [RuCl3(DMSO) (Hapbim)] (2) (Hapbim = 2-aminophenyl benzimidazole) have been synthesized and characterized, and their chemotherapeutic potential evaluated. The interaction of the compounds with DNA was studied by both UV-Visible and fluorescence spectroscopies, revealing intercalation of both the Hapbim ligand and the Ru complexes. The in vitro cytotoxicity of the compounds was tested on human breast cancer (MCF7), human colorectal cancer (Caco2), and normal human liver cell lines (THLE-2), with compound (2) the most potent against cancer cells. The cytotoxic effect of (2) is shown to correlate with the ability of the Ru(III) complex to induce apoptosis and to cause cell-cycle arrest in the G2/M phase. Notably, both compounds were inactive in the noncancerous cell line. The anticancer effect of (2) has also been studied in an EAC (Ehrlich Ascites Carcinoma) mouse model. Significantly, the activity of the complex was more pronounced in vivo, with removal of the cancer burden at doses that resulted in only low levels of hepatotoxicity and nephrotoxicity. An apoptosis mechanism was determined by the observation of increased Bax and caspase 3 and decreased Bcl2 expression. Furthermore, (2) decreased oxidative stress and increased the levels of antioxidant enzymes, especially SOD, suggesting the enhancement of normal cell repair. Overall, compound (2) shows great potential as a chemotherapeutic candidate, with promising activity and low levels of side effects.
Collapse
|
20
|
Feng Y, Wei Z, Zhang J. Determination of Ursolic Acid in Extracts From Ligustri lucidum Fruit Using an Electrochemical Method. Front Chem 2020; 8:444. [PMID: 32537451 PMCID: PMC7267720 DOI: 10.3389/fchem.2020.00444] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/28/2020] [Indexed: 12/30/2022] Open
Abstract
In this work, we reported a facile wet chemical method for depositing Pt nanoparticles on the surface of boron nitride nanosheets (BNNS-Pt NPs). The deposited nanocomposite was applied for glassy carbon electrode surface modification. The modified electrode was then used for detecting ursolic acid (UA). The results indicate that the BNNS-Pt NPs exhibited excellent electrocatalytic activity toward UA oxidation compared with that of the bare glassy carbon electrode (GCE) and Pt NPs/GCE. The UA oxidation currents is linearly related its concentration from 1 to 1,200 pM. The limit of detection can be calculated to be 0.5 pM. In addition, the UA sensor was also successfully used for the determination of UA in Ligustri lucidum fruit samples.
Collapse
Affiliation(s)
- Yingpu Feng
- Centre of Cerebrovascular, Zhengzhou University People's Hospital, Zhengzhou, China.,Centre of Cerebrovascular, Henan Provincial People's Hospital, Zhengzhou, China
| | - Zheng Wei
- Department of Traditional Chinese Medicine, Henan Academy Institute of Traditional Chinese Medicine, Zhengzhou, China
| | - Junping Zhang
- Department of Traditional Chinese Medicine, Henan Academy Institute of Traditional Chinese Medicine, Zhengzhou, China
| |
Collapse
|
21
|
Redox cycling of copper by coumarin-di(2-picolyl)amine hybrid molecule leads to ROS-mediated modulation of redox scavengers, DNA damage and cell death in diethylnitrosamine induced hepatocellular carcinoma. Bioorg Chem 2020; 99:103818. [PMID: 32276135 DOI: 10.1016/j.bioorg.2020.103818] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/29/2020] [Accepted: 04/02/2020] [Indexed: 12/24/2022]
Abstract
Targeted therapy is a new strategy for cancer treatment that targets chemical entities specific to cancer cells than normal ones. One of the features associated with malignancy is the elevated copper which plays an integral role in angiogenesis. Work is in progress in our lab to identify new copper chelators to target elevated copper under targeted therapy for the killing of cancer cells. Recently, a coumarin-based copper chelator, di(2-picolyl)amine-3(bromoacetyl)coumarin hybrid molecule (ligand-L) has been synthesized by us, and also studied its copper-dependent macromolecular damage response in copper overloaded lymphocytes. The present study investigates the anticancer activity of ligand-L and its mode of action in rat model of diethylnitrosamine (DEN) induced hepatocellular carcinoma. It has been found that liver tissue has a marked increase in copper levels in DEN induced hepatocellular carcinoma. Ex vivo results showed that ligand-L inhibited cell viability, induced reactive oxygen species (ROS) generation, DNA damage, loss of mitochondrial membrane potential and caspase-3 activation in isolated hepatocellular carcinoma cells (HCC). All these effects induced by ligand-L were abrogated by neocuproine and N-acetylcysteine (ROS scavenger). Further, ligand-L treatment of animals bearing hepatocellular carcinoma results in an increment in the cellular redox scavengers, lipid peroxidation and DNA breakage in malignant hepatocytes. In vivo studies using ligand-L also showed that ligand-L possesses anticancer properties as evidenced by improvement in liver marker enzymes and liver surface morphology, and reduced alpha-fetoprotein in the treated group compared to untreated cancer-induced group. Overall, this study suggests that copper-ligand-L interaction leads to ROS generation which caused DNA damage and apoptosis in malignant cells. This study provides enough support to establish ligand-L as a clinically relevant lead molecule for the treatment of different malignancies.
Collapse
|
22
|
Luo H, Vong CT, Chen H, Gao Y, Lyu P, Qiu L, Zhao M, Liu Q, Cheng Z, Zou J, Yao P, Gao C, Wei J, Ung COL, Wang S, Zhong Z, Wang Y. Naturally occurring anti-cancer compounds: shining from Chinese herbal medicine. Chin Med 2019; 14:48. [PMID: 31719837 PMCID: PMC6836491 DOI: 10.1186/s13020-019-0270-9] [Citation(s) in RCA: 310] [Impact Index Per Article: 51.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2019] [Accepted: 10/23/2019] [Indexed: 12/24/2022] Open
Abstract
Numerous natural products originated from Chinese herbal medicine exhibit anti-cancer activities, including anti-proliferative, pro-apoptotic, anti-metastatic, anti-angiogenic effects, as well as regulate autophagy, reverse multidrug resistance, balance immunity, and enhance chemotherapy in vitro and in vivo. To provide new insights into the critical path ahead, we systemically reviewed the most recent advances (reported since 2011) on the key compounds with anti-cancer effects derived from Chinese herbal medicine (curcumin, epigallocatechin gallate, berberine, artemisinin, ginsenoside Rg3, ursolic acid, silibinin, emodin, triptolide, cucurbitacin B, tanshinone I, oridonin, shikonin, gambogic acid, artesunate, wogonin, β-elemene, and cepharanthine) in scientific databases (PubMed, Web of Science, Medline, Scopus, and Clinical Trials). With a broader perspective, we focused on their recently discovered and/or investigated pharmacological effects, novel mechanism of action, relevant clinical studies, and their innovative applications in combined therapy and immunomodulation. In addition, the present review has extended to describe other promising compounds including dihydroartemisinin, ginsenoside Rh2, compound K, cucurbitacins D, E, I, tanshinone IIA and cryptotanshinone in view of their potentials in cancer therapy. Up to now, the evidence about the immunomodulatory effects and clinical trials of natural anti-cancer compounds from Chinese herbal medicine is very limited, and further research is needed to monitor their immunoregulatory effects and explore their mechanisms of action as modulators of immune checkpoints.
Collapse
Affiliation(s)
- Hua Luo
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Chi Teng Vong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Hanbin Chen
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yan Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peng Lyu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Ling Qiu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Mingming Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Qiao Liu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zehua Cheng
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jian Zou
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Peifen Yao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Caifang Gao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Jinchao Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Carolina Oi Lam Ung
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Shengpeng Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Zhangfeng Zhong
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| | - Yitao Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macao, China
| |
Collapse
|
23
|
Frolova TS, Lipeeva AV, Baev DS, Baiborodin SI, Orishchenko КE, Kochetov AV, Sinitsyna OI. Fluorescent labeling of ursolic acid with FITC for investigation of its cytotoxic activity using confocal microscopy. Bioorg Chem 2019; 87:876-887. [PMID: 30538052 DOI: 10.1016/j.bioorg.2018.11.052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Revised: 11/26/2018] [Accepted: 11/29/2018] [Indexed: 11/17/2022]
Abstract
Fluorescent labeling is a widely-used approach in the study of intracellular processes. This method is becoming increasingly popular for studying small bioactive molecules of natural origin; it allows us to estimate the vital intracellular changes which occur under their influence. We propose a new approach for visualization of the intracellular distribution of triterpene acids, based on fluorescent labeling by fluoresceine isothiocyanate. As a model compound we took the most widely-used and best-studied acid in the ursane series - ursolic acid, as this enabled us to compare the results obtained during our research with the available data, in order to evaluate the validity of the proposed method. Experimental tracing of the dynamics of penetration and distribution of the labeled ursolic acid has shown that when the acid enters the cell, it initially localizes on the inner membranes where the predicted target Akt1/protein kinase B - a protein that inhibits apoptosis - is located.
Collapse
Affiliation(s)
- Tatiana S Frolova
- Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia; Novosibirsk Institute of Organic Chemistry of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia; Novosibirsk State University, 2, Pirogov Street, 630090 Novosibirsk, Russia; Federal Research Center of Fundamental and Translational Medicine of Siberian Branch of the Russian Academy of Sciences, 2, Timakov Street, 630117 Novosibirsk, Russia.
| | - Alla V Lipeeva
- Novosibirsk Institute of Organic Chemistry of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia
| | - Dmitry S Baev
- Novosibirsk Institute of Organic Chemistry of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia; Novosibirsk State University, 2, Pirogov Street, 630090 Novosibirsk, Russia
| | - Sergey I Baiborodin
- Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia
| | - Кonstantin E Orishchenko
- Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia
| | - Alexey V Kochetov
- Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia; Novosibirsk State University, 2, Pirogov Street, 630090 Novosibirsk, Russia
| | - Olga I Sinitsyna
- Federal Research Center Institute of Cytology and Genetics of Siberian Branch of the Russian Academy of Sciences, 10, Lavrentyev Ave., 630090 Novosibirsk, Russia; Novosibirsk State University, 2, Pirogov Street, 630090 Novosibirsk, Russia
| |
Collapse
|
24
|
Feng XM, Su XL. Anticancer effect of ursolic acid via mitochondria-dependent pathways. Oncol Lett 2019; 17:4761-4767. [PMID: 31186681 DOI: 10.3892/ol.2019.10171] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 02/01/2019] [Indexed: 01/03/2023] Open
Abstract
Ursolic acid is a plant-derived pentacyclic triterpenoid found in various medicinal herbs and fruits. It has generated clinical interest due to its anti-inflammatory, antioxidative, antiapoptotic and anticarcinogenic effects. An increasing amount of evidence supports the anticancer effect of ursolic acid in various cancer cells. One of the hallmarks of malignant transformation is metabolic reprogramming that sustains macromolecule synthesis, bioenergetic demand and tumor cell survival. Mitochondria are important regulators of tumorigenes is as well as a major site of the metabolic reactions that facilitate this reprogramming and adaption to cellular and environmental changes. The current review explored the close association between the anticancer effect of ursolic acid and the activation of mitochondrial-dependent signaling pathways.
Collapse
Affiliation(s)
- Xue-Min Feng
- Clinical Medical Research Center of The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| | - Xiu-Lan Su
- Clinical Medical Research Center of The Affiliated Hospital, Inner Mongolia Medical University, Hohhot, Inner Mongolia 010050, P.R. China
| |
Collapse
|
25
|
Barua A, Choudhury P, Maity JK, Mandal SB, Mandal S, Saha P. Chemotherapeutic potential of novel non-toxic nucleoside analogues on EAC ascitic tumour cells. Free Radic Res 2019; 53:57-67. [DOI: 10.1080/10715762.2018.1551999] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Affiliation(s)
- Atish Barua
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, Kolkata, India
| | - Pritha Choudhury
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, Kolkata, India
| | - Joy Krishna Maity
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, Kolkata, India
| | - Sukhendu Bikash Mandal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, Kolkata, India
| | - Suvra Mandal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, Kolkata, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, Kolkata, India
| |
Collapse
|
26
|
Ji X, Tang Q, Pang P, Wu J, Kirk TB, Xu J, Ma D, Xue W. Redox-responsive chemosensitive polyspermine delivers ursolic acid targeting to human breast tumor cells: The depletion of intracellular GSH contents arouses chemosensitizing effects. Colloids Surf B Biointerfaces 2018; 170:293-302. [PMID: 29936382 DOI: 10.1016/j.colsurfb.2018.06.029] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Revised: 05/16/2018] [Accepted: 06/17/2018] [Indexed: 01/08/2023]
Abstract
Antitumor efficacy of ursolic acid (UA) is seriously limited due to its low hydrophilicity and needy bioavailability. To overcome these obstacles, chemosensitive polyspermine (CPSP) conjugated with UA and folic acid (FA) as a novel targeted prodrug was designed and successfully synthesized in this investigation. This prodrug not only showed high aqueous solubility, GSH-triggered degradation and good biocompatibility, but also exhibited better inhibition effect on the tumor cells proliferation in comparison with free UA. FA-CPSP-UA could down-regulate the generation of GSH and manifest excellent ability in enhancing antitumor efficacy. In addition, FA-CPSP-UA could inhibit the expression of MMP-9, which led to restricting MCF-7 cells migration. Taken together, the results indicated that FA-CPSP-UA, as a carrier, can efficiently deliver UA to folate receptor positive cancer cells and improve tumor therapy of UA by Chemosensitive effect.
Collapse
Affiliation(s)
- Xin Ji
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Qiao Tang
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China
| | - Peng Pang
- College of Traditional Chinese Medicine, Jinan University, Guangzhou 510632, China
| | - Jianping Wu
- 3D Imaging and Bioengineering Laboratory, Department of Mechanical Engineering, Curtin University, Australia
| | - Thomas Brett Kirk
- 3D Imaging and Bioengineering Laboratory, Department of Mechanical Engineering, Curtin University, Australia
| | - Jiake Xu
- The School of Pathology and Laboratory Medicine, University of Western Australia, Perth, Australia
| | - Dong Ma
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| | - Wei Xue
- Key Laboratory of Biomaterials of Guangdong Higher Education Institutes, Department of Biomedical Engineering, Jinan University, Guangzhou 510632, China.
| |
Collapse
|
27
|
Fan L, Zhang B, Xu A, Shen Z, Guo Y, Zhao R, Yao H, Shao JW. Carrier-Free, Pure Nanodrug Formed by the Self-Assembly of an Anticancer Drug for Cancer Immune Therapy. Mol Pharm 2018; 15:2466-2478. [PMID: 29727577 DOI: 10.1021/acs.molpharmaceut.8b00444] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Ursolic acid (UA) is a food-plant-derived natural product which has good anticancer activities and low toxicity. However, the poor water solubility of UA limits its application in clinic. To address this issue, we developed a carrier-free nanodrug by self-assembly of UA. Here, we showed that UA nanoparticles (NPs) have a near-spherical shape with a diameter of ∼150 nm. UA NPs exhibited higher antiproliferative activity; significantly caused apoptosis; decreased the expression of COX-2/VEGFR2/VEGFA; and increased the immunostimulatory activity of TNF-α, IL-6, and IFN-β and decreased the activity of STAT-3 in A549 cells in vitro. Furthermore, UA NPs could inhibit tumor growth and have the ability of liver protection in vivo. More importantly, UA NPs could significantly improve the activation of CD4+ T-cells, which indicated that UA NPs have the potential for immunotherapy. Overall, a carrier-free UA nanodrug may be a promising drug to further enhance their anticancer efficacy and immune function.
Collapse
Affiliation(s)
- Lulu Fan
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| | - Bingchen Zhang
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| | - Aixiao Xu
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| | - Zhichun Shen
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| | - Yan Guo
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| | - Ruirui Zhao
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| | - Huilu Yao
- School of Physical Science and Technology , Guangxi University , Guangxi 530004 , China
| | - Jing-Wei Shao
- Cancer Metastasis Alert and Prevention Center, Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, and Fujian Provincial Key Laboratory of Cancer Metastasis Chemoprevention and Chemotherapy, College of Chemistry , Fuzhou University , Fuzhou 350108 , China
| |
Collapse
|
28
|
Iqbal J, Abbasi BA, Batool R, Mahmood T, Ali B, Khalil AT, Kanwal S, Shah SA, Ahmad R. Potential phytocompounds for developing breast cancer therapeutics: Nature’s healing touch. Eur J Pharmacol 2018. [DOI: 10.1016/j.ejphar.2018.03.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
29
|
Zhang CX, Ma WJ, Liu DL, Jia XJ, Zhao YM. Biotransformation of ursolic acid by Alternaria longipes AS3.2875. Nat Prod Res 2018; 32:536-543. [PMID: 28553725 DOI: 10.1080/14786419.2017.1327860] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2017] [Accepted: 04/27/2017] [Indexed: 01/27/2023]
Abstract
Microbial transformation of ursolic acid (1) was carried out by Alternaria longipes AS 3.2875. Six transformed products (2-7) from 1 were isolated and their structures were identified as 3-carbonyl ursolic acid 28-O-β-D-glucopyranosyl ester (2), ursolic acid 3-O-β-D-glucopyranoside (3), ursolic acid 28-O-β-D-glucopyranosyl ester (4), 2α, 3β-dihydroxy ursolic acid 28-O-β-D-glucopyranosyl ester (5), 3β, 21β dihydroxy ursolic acid 28-O-β-D-glucopyranosyl ester (6), and 3-O-(β-D-glucopyranosyl)- ursolic acid 28-O-(β-D-glucopyranosyl) ester (7) based on the analysis of 1D NMR, 2DNMR and MS data. The product 2 was a new compound among them and showed stronger antibacterial activity against S. aureu, MRSA and MRCA than substrate. In this study, we modified structure of ursolic acid through biotransformation to enhance its activities and preliminarily discussed the transformation way of the products.
Collapse
Affiliation(s)
- Chen-Xi Zhang
- a Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard , Logistics University of Chinese People's Armed Police Forces , Tianjin , P.R. China
- b College of Traditional Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , P.R. China
| | - Wei-Jun Ma
- a Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard , Logistics University of Chinese People's Armed Police Forces , Tianjin , P.R. China
| | - Dai-Lin Liu
- a Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard , Logistics University of Chinese People's Armed Police Forces , Tianjin , P.R. China
| | - Xiu-Juan Jia
- b College of Traditional Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , P.R. China
| | - Yan-Min Zhao
- a Tianjin Key Laboratory for Prevention and Control of Occupational and Environmental Hazard , Logistics University of Chinese People's Armed Police Forces , Tianjin , P.R. China
- b College of Traditional Chinese Medicine , Tianjin University of Traditional Chinese Medicine , Tianjin , P.R. China
| |
Collapse
|
30
|
Frolova TS, Lipeeva AV, Baev DS, Tsepilov YA, Sinitsyna OI. Apoptosis as the basic mechanism of cytotoxic action of ursolic and pomolic acids in glioma cells. Mol Biol 2017. [DOI: 10.1134/s0026893317050090] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
|
31
|
Yuan Z, Wu W, Zhang Z, Sun Z, Cheng R, Pan G, Wang X, Cui W. In situ adjuvant therapy using a responsive doxorubicin-loaded fibrous scaffold after tumor resection. Colloids Surf B Biointerfaces 2017; 158:363-369. [DOI: 10.1016/j.colsurfb.2017.06.052] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2017] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 01/01/2023]
|
32
|
Bergamin LS, Figueiró F, Dietrich F, Manica FDM, Filippi-Chiela EC, Mendes FB, Jandrey EHF, Lopes DV, Oliveira FH, Nascimento IC, Ulrich H, Battastini AMO. Interference of ursolic acid treatment with glioma growth: An in vitro and in vivo study. Eur J Pharmacol 2017; 811:268-275. [PMID: 28663034 DOI: 10.1016/j.ejphar.2017.06.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 06/21/2017] [Accepted: 06/26/2017] [Indexed: 01/01/2023]
Abstract
Glioblastoma multiforme is the most devastating tumor in the brain. Ursolic acid (UA) is found in a variety of plants, and exhibits several pharmacological activities. In this study, we investigated the effects of UA in vitro, clarifying the mechanisms that mediate its toxicity and the long-lasting actions of UA in C6 glioma cells. We also evaluated the antitumor activity of UA in an in vivo orthotopic glioma model. Cell numbers were assessed using the Trypan blue exclusion test, and the cell cycle was characterized by flow cytometry using propidium iodide staining. Apoptosis was analyzed using an Annexin V kit and by examining caspase-3. Akt immunocontent was verified by Western blot and the long-lasting actions of UA were measured by cumulative population doubling (CPD). In vivo experiments were performed in rats to measure the effects on tumor size, malignant features and toxicological parameters. In vitro results showed that UA decreased glioma cell numbers, increased the sub-G1 fraction and induced apoptotic death, accompanied by increased active caspase-3 protein levels. Akt phosphorylation/activation in cells was also diminished by UA. With regard to CPD, cell proliferation was almost completely restored upon single UA treatments, but when the UA was added again, the majority of cells died, demonstrating the importance of re-treatment cycles with chemotherapeutic agents for abolishing tumor growth. In vivo, ursolic acid slightly reduced glioma tumor size but did not decrease malignant features. Ursolic acid may be a potential candidate as an adjuvant for glioblastoma therapy.
Collapse
Affiliation(s)
- Letícia Scussel Bergamin
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Fabrício Figueiró
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Fabrícia Dietrich
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Fabiana de Mattos Manica
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Eduardo C Filippi-Chiela
- Programa de Pós-Graduação em Gastroenterologia e Hepatologia, Faculdade de Medicina, UFRGS, Porto Alegre, RS, Brazil
| | - Franciane Brackman Mendes
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | | | | | - Francine H Oliveira
- Serviço de Patologia, Hospital de Clínicas de Porto Alegre, UFRGS, Porto Alegre, RS, Brazil
| | - Isis C Nascimento
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Henning Ulrich
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil; Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
33
|
Wang H, Sim MK, Loke WK, Chinnathambi A, Alharbi SA, Tang FR, Sethi G. Potential Protective Effects of Ursolic Acid against Gamma Irradiation-Induced Damage Are Mediated through the Modulation of Diverse Inflammatory Mediators. Front Pharmacol 2017; 8:352. [PMID: 28670276 PMCID: PMC5472704 DOI: 10.3389/fphar.2017.00352] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 05/23/2017] [Indexed: 01/08/2023] Open
Abstract
This study was aimed to evaluate the possible protective effects of ursolic acid (UA) against gamma radiation induced damage both in vitro as well as in vivo. It was observed that the exposure to gamma radiation dose- and time-dependently caused a significant decrease in the cell viability, while the treatment of UA attenuated this cytotoxicity. The production of free radicals including reactive oxygen species (ROS) and NO increased significantly post-irradiation and further induced lipid peroxidation and oxidative DNA damage in cells. These deleterious effects could also be effectively blocked by UA treatment. In addition, UA also reversed gamma irradiation induced inflammatory responses, as indicated by the decreased production of TNF-α, IL-6, and IL-1β. NF-κB signaling pathway has been reported to be a key mediator involved in gamma radiation-induced cellular damage. Our results further demonstrated that gamma radiation dose- and time-dependently enhanced NF-κB DNA binding activity, which was significantly attenuated upon UA treatment. The post-irradiation increase in the expression of both phospho-p65, and phospho-IκBα was also blocked by UA. Moreover, the treatment of UA was found to significantly prolong overall survival in mice exposed to whole body gamma irradiation, and reduce the excessive inflammatory responses. Given its radioprotective efficacy as described here, UA as an antioxidant and NF-κB pathway blocker, may function as an important pharmacological agent in protecting against gamma irradiation-induced injury.
Collapse
Affiliation(s)
- Hong Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
- Singapore Nuclear Research and Safety Initiative, National University of SingaporeSingapore, Singapore
| | - Meng-Kwoon Sim
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
| | - Weng Keong Loke
- Agent Diagnostic and Therapeutic Laboratory, Defence and Environmental Research Institute, DSO National LaboratoriesSingapore, Singapore
| | - Arunachalam Chinnathambi
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh, Saudi Arabia
| | - Sulaiman Ali Alharbi
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh, Saudi Arabia
| | - Feng Ru Tang
- Singapore Nuclear Research and Safety Initiative, National University of SingaporeSingapore, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of SingaporeSingapore, Singapore
- Department of Botany and Microbiology, College of Science, King Saud UniversityRiyadh, Saudi Arabia
- School of Biomedical Sciences, Curtin Health Innovation Research Institute, Curtin University, PerthWA, Australia
| |
Collapse
|
34
|
Cheng W, Dahmani FZ, Zhang J, Xiong H, Wu Y, Yin L, Zhou J, Yao J. Anti-angiogenic activity and antitumor efficacy of amphiphilic twin drug from ursolic acid and low molecular weight heparin. NANOTECHNOLOGY 2017; 28:075102. [PMID: 28091396 DOI: 10.1088/1361-6528/aa53c6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Heparin, a potential blood anti-coagulant, is also known for its binding ability to several angiogenic factors through electrostatic interactions due to its polyanionic character. However, the clinical application of heparin for cancer treatment is limited by several drawbacks, such as unsatisfactory therapeutic effects and severe anticoagulant activity that could induce hemorrhaging. Herein, low molecular weight heparin (LMWH) was conjugated to ursolic acid (UA), which is also an angiogenesis inhibitor, by binding the amine group of aminoethyl-UA (UA-NH2) with the carboxylic groups of LMWH. The resulting LMWH-UA conjugate as an amphiphilic twin drug showed reduced anticoagulant activity and could also self-assemble into nanomicelles with a mean particle size ranging from 200-250 nm. An in vitro endothelial tubular formation assay and an in vivo Matrigel plug assay were performed to verify the anti-angiogenic potential of LMWH-UA. Meanwhile, the in vivo antitumor effect of LMWH-UA was also evaluated using a B16F10 mouse melanoma model. LMWH-UA nanomicelles were shown to inhibit angiogenesis both in vitro and in vivo. In addition, the i.v. administration of LMWH-UA to the B16F10 tumor-bearing mice resulted in a significant inhibition of tumor growth as compared to the free drug solutions. These findings demonstrate the therapeutic potential of LMWH-UA as a new therapeutic remedy for cancer therapy.
Collapse
Affiliation(s)
- Wenming Cheng
- State Key Laboratory of Natural Medicines, Department of Pharmaceutics, China Pharmaceutical University, 24 Tongjiaxiang, Nanjing 210009, People's Republic of China
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Bidirectional regulation of angiogenesis by phytoestrogens through estrogen receptor-mediated signaling networks. Chin J Nat Med 2017; 14:241-254. [PMID: 27114311 DOI: 10.1016/s1875-5364(16)30024-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2015] [Indexed: 01/21/2023]
Abstract
Sex hormone estrogen is one of the most active intrinsic angiogenesis regulators; its therapeutic use has been limited due to its carcinogenic potential. Plant-derived phytoestrogens are attractive alternatives, but reports on their angiogenic activities often lack in-depth analysis and sometimes are controversial. Herein, we report a data-mining study with the existing literature, using IPA system to classify and characterize phytoestrogens based on their angiogenic properties and pharmacological consequences. We found that pro-angiogenic phytoestrogens functioned predominantly as cardiovascular protectors whereas anti-angiogenic phytoestrogens played a role in cancer prevention and therapy. This bidirectional regulation were shown to be target-selective and, for the most part, estrogen-receptor-dependent. The transactivation properties of ERα and ERβ by phytoestrogens were examined in the context of angiogenesis-related gene transcription. ERα and ERβ were shown to signal in opposite ways when complexed with the phytoestrogen for bidirectional regulation of angiogenesis. With ERα, phytoestrogen activated or inhibited transcription of some angiogenesis-related genes, resulting in the promotion of angiogenesis, whereas, with ERβ, phytoestrogen regulated transcription of angiogenesis-related genes, resulting in inhibition of angiogenesis. Therefore, the selectivity of phytoestrogen to ERα and ERβ may be critical in the balance of pro- or anti-angiogenesis process.
Collapse
|
36
|
Li T, Chen X, Liu Y, Fan L, Lin L, Xu Y, Chen S, Shao J. pH-Sensitive mesoporous silica nanoparticles anticancer prodrugs for sustained release of ursolic acid and the enhanced anti-cancer efficacy for hepatocellular carcinoma cancer. Eur J Pharm Sci 2017; 96:456-463. [PMID: 27771513 DOI: 10.1016/j.ejps.2016.10.019] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2016] [Revised: 10/18/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
Abstract
Ursolic acid (UA) as a nature product exhibits good anti-cancer activity, low toxicity, and good liver protection features. However, the low-solubility and poor bioavailability restrict its further clinical application. To overcome this problem, a pH-sensitive prodrug delivery system (UA@MSN-UA) that incorporated acid-sensitive linkage between drug and silica-based mesoporous nanosphere (MSN) was successfully designed and synthesized. The physicochemical properties of the UA@MSN-UA nanoparticles were investigated for shape, particle size, zeta potential, nitrogen adsorption-desorption and infrared (IR) spectroscopy. The nanoparticles were further evaluated for in vitro cytotoxicity, including proliferation inhibition, cell cycle distribution and apoptotic effects against human hepatocellular carcinoma HepG2 cells. The TEM image showed that the size of synthesized MSN nanoparticle was a near-spherical shape with ~100nm diameter. In vitro cytotoxicity testing demonstrated that UA@MSN-UA nanoparticles prodrug exhibited higher proliferation inhibition, cell cycle arrest at the G2/M phase and significantly caused the early and late apoptosis in HepG2 cells, which would be contributed to high loading capacity, high cellular uptake and sustained release of UA. Overall, the UA-modified MSN prodrug delivery system can be a promising drug carrier for improving the bioavailability of UA, and further enhance its anti-cancer efficacy.
Collapse
Affiliation(s)
- Tao Li
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Fujian Provincial Key Laboratory of Cancr Metastasis Chemoprevention, Fuzhou University, Fuzhou, China
| | - Xiufen Chen
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Fujian Provincial Key Laboratory of Cancr Metastasis Chemoprevention, Fuzhou University, Fuzhou, China
| | - Yajun Liu
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Fujian Provincial Key Laboratory of Cancr Metastasis Chemoprevention, Fuzhou University, Fuzhou, China
| | - Lulu Fan
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Fujian Provincial Key Laboratory of Cancr Metastasis Chemoprevention, Fuzhou University, Fuzhou, China
| | - Liqing Lin
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Yu Xu
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Sijia Chen
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China
| | - Jingwei Shao
- Cancer Metastasis Alert and Prevention Center, and Pharmaceutical Photocatalysis of State Key Laboratory of Photocatalysis on Energy and Environment, College of Chemistry, Fuzhou University, Fuzhou 350108, China; Fujian Provincial Key Laboratory of Cancr Metastasis Chemoprevention, Fuzhou University, Fuzhou, China.
| |
Collapse
|
37
|
Elmeligie S, Ahmed EM, Abuel-Maaty SM, Zaitone SAB, Mikhail DS. Design and Synthesis of Pyridazine Containing Compounds with Promising Anticancer Activity. Chem Pharm Bull (Tokyo) 2017; 65:236-247. [DOI: 10.1248/cpb.c16-00532] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Salwa Elmeligie
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University
| | - Eman Mohamed Ahmed
- Pharmaceutical Organic Chemistry Department, Faculty of Pharmacy, Cairo University
| | | | | | | |
Collapse
|
38
|
Valdés K, Morales J, Rodríguez L, Günther G. Potential use of nanocarriers with pentacyclic triterpenes in cancer treatments. Nanomedicine (Lond) 2016; 11:3139-3156. [PMID: 27809705 DOI: 10.2217/nnm-2016-0251] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Ursolic, oleanolic and betulinic acids are representative pentacyclic triterpenoids found in various plants and fruits. Despite having marked antitumor potentials, the very poor water solubility of these triterpenes hinders treatment development. Nanotechnology can enhance solubility, stability, bioavailability and phytochemical delivery, improving the therapeutic efficiency of triterpenes. This review focuses on the formulation, characterization and in vitro/in vivo evaluation of several delivery nanosystems used to enhance the physicochemical properties of ursolic, oleanolic and betulinic acids.
Collapse
Affiliation(s)
- Karina Valdés
- Departamento de Ciencias y Tecnología Farmacéutica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Javier Morales
- Departamento de Ciencias y Tecnología Farmacéutica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| | - Lennin Rodríguez
- Facultad de Farmacia y Bioquímica, Universidad Nacional de Trujillo, Trujillo, Perú
| | - Germán Günther
- Departamento de Química Orgánica y Fisicoquímica, Facultad de Ciencias Químicas y Farmacéuticas, Universidad de Chile, Santiago, Chile
| |
Collapse
|
39
|
Wu CC, Huang YF, Hsieh CP, Chueh PJ, Chen YL. Combined Use of Zoledronic Acid Augments Ursolic Acid-Induced Apoptosis in Human Osteosarcoma Cells through Enhanced Oxidative Stress and Autophagy. Molecules 2016; 21:E1640. [PMID: 27916903 PMCID: PMC6274426 DOI: 10.3390/molecules21121640] [Citation(s) in RCA: 18] [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: 10/25/2016] [Revised: 11/16/2016] [Accepted: 11/25/2016] [Indexed: 01/10/2023] Open
Abstract
Ursolic acid (UA), a naturally occurring pentacyclic triterpene acid found in many medicinal herbs and edible plants, triggers apoptosis in several tumor cell lines but not in human bone cancer cells. Most recently, we have demonstrated that UA exposure reduces the viability of human osteosarcoma MG-63 cells through enhanced oxidative stress and apoptosis. Interestingly, an inhibitor of osteoclast-mediated bone resorption, zoledronic acid (ZOL), also a third-generation nitrogen-containing bisphosphonate, is effective in the treatment of bone metastases in patients with various solid tumors. In this present study, we found that UA combined with ZOL to significantly suppress cell viability, colony formation, and induce apoptosis in two lines of human osteosarcoma cells. The pre-treatment of the antioxidant had reversed the oxidative stress and cell viability inhibition in the combined treatment, indicating that oxidative stress is important in the combined anti-tumor effects. Moreover, we demonstrated that ZOL combined with UA significantly induced autophagy and co-administration of autophagy inhibitor reduces the growth inhibitory effect of combined treatment. Collectively, these data shed light on the pathways involved in the combined effects of ZOL and UA that might serve as a potential therapy against osteosarcoma.
Collapse
Affiliation(s)
- Chia-Chieh Wu
- Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung 40227, Taiwan.
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
| | - Yi-Fu Huang
- Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Chen-Pu Hsieh
- Orthopedics & Sports Medicine Laboratory, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Department of Orthopedic Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan.
| | - Pin-Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing University, 145 Xingda Rd., South Dist., Taichung 40227, Taiwan.
- Department of Biotechnology, Asia University, Taichung 41354, Taiwan.
- Graduate Institute of Basic Medicine, China Medical University, Taichung 40402, Taiwan.
- Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan.
| | - Yao-Li Chen
- School of Medicine, Kaohsiung Medical University, Kaohsiung 80708, Taiwan.
- Transplant Medicine & Surgery Research Centre, Changhua Christian Hospital, Changhua 50006, Taiwan.
- Department of Surgery, Changhua Christian Hospital, 135 Nansiao St., Changhua 50006, Taiwan.
| |
Collapse
|
40
|
Bhattacharjee A, Basu A, Biswas J, Sen T, Bhattacharya S. Chemoprotective and chemosensitizing properties of selenium nanoparticle (Nano-Se) during adjuvant therapy with cyclophosphamide in tumor-bearing mice. Mol Cell Biochem 2016; 424:13-33. [PMID: 27696310 DOI: 10.1007/s11010-016-2839-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2016] [Accepted: 09/24/2016] [Indexed: 12/26/2022]
Abstract
Cyclophosphamide (CP) is one of the widely used anticancer agents; however, it has serious deleterious effects on normal host cells due to its nonspecific action. The essential trace element Selenium (Se) is suggested to have chemopreventive and chemotherapeutic efficacy and currently used in pharmaceutical formulations. Previous report had shown Nano-Se could protect CP-induced hepatotoxicity and genotoxicity in normal Swiss albino mice; however, its role in cancer management is still not clear. The aim of present study is to investigate the chemoprotective efficacy of Nano-Se against CP-induced toxicity as well as its chemoenhancing capability when used along with CP in Swiss albino mice against Ehrlich's ascites carcinoma (EAC) cells. CP was administered (25 mg/kg b.w., i.p.) and Nano-Se was given (2 mg Se/kg b.w., p.o.) in concomitant and pretreatment schedule. Increase levels of serum hepatic marker, hepatic lipid peroxidation, DNA damage, and chromosomal aberration in CP-treated mice were significantly (P < 0.05) reversed by Nano-Se. The lowered status of various antioxidant enzymes in tumor-bearing mice after CP treatment was also effectively increased by Nano-Se. Administration of Nano-Se along with CP caused a significant reduction in tumor volume, packed cell volume, viable tumor cell count, and increased the survivability of the tumor-bearing hosts. The results suggest that Nano-Se exhibits significant antitumor and antioxidant effects in EAC-bearing mice. The potential for Nano-Se to ameliorate the CP-evoked toxicity as well as to improve the chemotherapeutic effect could have beneficial implications for patients undergoing chemotherapy with CP.
Collapse
Affiliation(s)
- Arin Bhattacharjee
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Abhishek Basu
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Jaydip Biswas
- Department of Translational Research, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India
| | - Tuhinadri Sen
- Division of Pharmacology, Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mullick Road, Kolkata, West Bengal, 700032, India
| | - Sudin Bhattacharya
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute, 37, S. P. Mukherjee Road, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
41
|
Jiang Q, Hao R, Wang W, Gao H, Wang C. SIRT1/Atg5/autophagy are involved in the antiatherosclerosis effects of ursolic acid. Mol Cell Biochem 2016; 420:171-84. [PMID: 27514536 DOI: 10.1007/s11010-016-2787-x] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 08/05/2016] [Indexed: 12/22/2022]
Abstract
The purpose of this study was to investigate the antiatherosclerosis effects of ursolic acid (UA) in high-fat diet-fed quails (Coturnix coturnix) and potential mechanism. Quails were treated with high-fat diet (14 % pork oil, 1 % cholesterol w/w) with or without UA (50, 150, or 300 mg/kg/day) for 10 weeks. Serum lipid profile was assessed at 0, 4.5, and 10 weeks. After 10 weeks, serum antioxidant status and morphology of aorta were assessed. Additionally, human umbilical vein endothelial cells (HUVECs) were exposed to 100 μg/ml oxidized low-density lipoprotein (ox-LDL) for 24 h, with or without pretreatment with UA (5, 10 or 20 μM) for 16 h, autophagy inhibitor 3-MA 5 mM for 2 h, or SIRT1 inhibitor EX-527 10 μM for 2 h. Cell viability and oxidative stress status were assessed and autophagy status was determined. Acetylation of lysine residue on Atg5 was assessed with immunoprecipitation. In results, high-fat diet negatively affected serum lipid profile and antioxidant status in quails and induced significant histological changes. Cotreatment with UA remarkably alleviated such changes. In HUVECs, ox-LDL treatment induced significant cytotoxicity along with oxidative stress, while UA cotreatment alleviated such changes significantly. UA treatment induced autophagy, enhanced SIRT1 expression, and decreased acetylation of lysine residue on Atg5. Cotreatment with 3-MA or EX-527 effectively abolished UA's protective effects. In summary, UA exerted antiatherosclerosis effects in quails and protected HUVECs from ox-LDL induced cytotoxicity, and the mechanism is associated with increased SIRT1 expression, decreased Atg5 acetylation on lysine residue, and increased autophagy.
Collapse
Affiliation(s)
- Qixiao Jiang
- Department of Pharmacology, Qingdao University Medical College, Boya Building Room 422, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Ranran Hao
- Department of Pharmacology, Qingdao University Medical College, Boya Building Room 422, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Wencheng Wang
- The Institute of Human Nutrition Medical College, 38 Dengzhou Road, Qingdao, 266071, Shandong, China
| | - Hui Gao
- Department of Pharmacology, Qingdao University Medical College, Boya Building Room 422, 308 Ningxia Road, Qingdao, 266071, Shandong, China
| | - Chunbo Wang
- Department of Pharmacology, Qingdao University Medical College, Boya Building Room 422, 308 Ningxia Road, Qingdao, 266071, Shandong, China.
| |
Collapse
|
42
|
Jin H, Pi J, Yang F, Wu C, Cheng X, Bai H, Huang D, Jiang J, Cai J, Chen ZW. Ursolic acid-loaded chitosan nanoparticles induce potent anti-angiogenesis in tumor. Appl Microbiol Biotechnol 2016; 100:6643-6652. [PMID: 26883344 DOI: 10.1007/s00253-016-7360-8] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 01/22/2016] [Accepted: 01/27/2016] [Indexed: 01/08/2023]
Abstract
Angiogenesis provides necessary nutrients and oxygen for tumor growth and metastasis; thus, every stage of angiogenesis process is the potential target for cancer therapies. Ursolic acid (UA) is reported to decrease tumor burden through anti-angiogenesis pathway, but its poor water solubility greatly limits its efficiency and clinical application. Here, a simple method for preparing UA-loaded chitosan nanoparticles (CH-UA-NPs) with anti-angiogenesis and anti-tumor activity was demonstrated. In vitro, CH-UA-NPs could significantly inhibit the proliferation, migration, and tube formation of human umbilical vascular endothelial cells (HUVECs). After uptake by HUVECs, CH-UA-NPs were mainly localized in lysosomes and mitochondria, but not nuclei. CH-UA-NPs induced the destruction of lysosome membrane integrity, collapse of mitochondrial membrane potential, and reorganization of cell cytoskeleton. All these changes led to the apoptosis or necrosis in HUVECs. In vivo, CH-UA-NPs could inhibit the angiogenesis in chicken chorioallantoic membrane (CAM) model and H22 xenograft model. Notably, comparing with free UA, such synthesized CH-UA-NPs could save about tenfold of UA doses, implying that this could significantly decrease the side effects induced by high doses of UA in biological organism. Our data showed that CH-UA-NPs and this nanoparticle-based drug delivery system could be as a potential drug candidate for anti-angiogenesis treatment.
Collapse
Affiliation(s)
- Hua Jin
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 000853, China
- Department of Microbiology and Immunology, University of Illinois, Chicago, 60612, USA
| | - Jiang Pi
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 000853, China
| | - Fen Yang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 000853, China
| | - Chaomin Wu
- Qingpu District, Qingpu Branch of Zhongshan Hospital, Fudan University, Shanghai, 201700, China
| | - Xueli Cheng
- Material Science Lab, SAE Technologies Development (Dongguan) Co. Ltd, Dongguan City, 523087, China
| | - Haihua Bai
- Material Science Lab, SAE Technologies Development (Dongguan) Co. Ltd, Dongguan City, 523087, China
| | - Dan Huang
- Department of Microbiology and Immunology, University of Illinois, Chicago, 60612, USA
| | - Jinhuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 000853, China
| | - Jiye Cai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 000853, China.
| | - Zheng W Chen
- Department of Microbiology and Immunology, University of Illinois, Chicago, 60612, USA.
| |
Collapse
|
43
|
Jin H, Pi J, Yang F, Jiang J, Wang X, Bai H, Shao M, Huang L, Zhu H, Yang P, Li L, Li T, Cai J, Chen ZW. Folate-Chitosan Nanoparticles Loaded with Ursolic Acid Confer Anti-Breast Cancer Activities in vitro and in vivo. Sci Rep 2016; 6:30782. [PMID: 27469490 PMCID: PMC4965748 DOI: 10.1038/srep30782] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2015] [Accepted: 07/08/2016] [Indexed: 01/26/2023] Open
Abstract
Ursolic acid (UA) has proved to have broad-spectrum anti-tumor effects, but its poor water solubility and incompetent targeting property largely limit its clinical application and efficiency. Here, we synthesized a nanoparticle-based drug carrier composed of chitosan, UA and folate (FA-CS-UA-NPs) and demonstrated that FA-CS-UA-NPs could effectively diminish off-target effects and increase local drug concentrations of UA. Using MCF-7 cells as in vitro model for anti-cancer mechanistic studies, we found that FA-CS-UA-NPs could be easily internalized by cancer cells through a folate receptor-mediated endocytic pathway. FA-CS-UA-NPs entered into lysosome, destructed the permeability of lysosomal membrane, and then got released from lysosomes. Subsequently, FA-CS-UA-NPs localized into mitochondria but not nuclei. The prolonged retention of FA-CS-UA-NPs in mitochondria induced overproduction of ROS and destruction of mitochondrial membrane potential, and resulted in the irreversible apoptosis in cancer cells. In vivo experiments showed that FA-CS-UA-NPs could significantly reduce breast cancer burden in MCF-7 xenograft mouse model. These results suggested that FA-CS-UA-NPs could further be explored as an anti-cancer drug candidate and that our approach might provide a platform to develop novel anti-cancer drug delivery system.
Collapse
Affiliation(s)
- Hua Jin
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
- Department of Microbiology and Immunology, University of Illinois, Chicago 60612, USA
| | - Jiang Pi
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Fen Yang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Jinhuan Jiang
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Xiaoping Wang
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Haihua Bai
- Department of Chemistry, materials science and engineering, Jinan University, Guangzhou 510632, China
| | - Mingtao Shao
- The First Affiliated Hospital of Jinan University, Guangzhou 510632, China
| | - Lei Huang
- Treatment and Research Center of Infectious Diseases, the 302 Hospital of PLA, Beijing, 100039, China
| | - Haiyan Zhu
- Department of Chemistry, materials science and engineering, Jinan University, Guangzhou 510632, China
| | - Peihui Yang
- Department of Chemistry, materials science and engineering, Jinan University, Guangzhou 510632, China
| | - Lihua Li
- Department of Chemistry, materials science and engineering, Jinan University, Guangzhou 510632, China
| | - Ting Li
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
| | - Jiye Cai
- State Key Laboratory of Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau, 999078, China
- Department of Chemistry, materials science and engineering, Jinan University, Guangzhou 510632, China
| | - Zheng W. Chen
- Department of Microbiology and Immunology, University of Illinois, Chicago 60612, USA
| |
Collapse
|
44
|
Jiang Q, Han Y, Gao H, Tian R, Li P, Wang C. Ursolic acid induced anti-proliferation effects in rat primary vascular smooth muscle cells is associated with inhibition of microRNA-21 and subsequent PTEN/PI3K. Eur J Pharmacol 2016; 781:69-75. [PMID: 27085898 DOI: 10.1016/j.ejphar.2016.04.001] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Revised: 03/20/2016] [Accepted: 04/04/2016] [Indexed: 01/08/2023]
Abstract
This study focused on the anti-proliferation effects of ursolic acid (UA) in rat primary vascular smooth muscle cells (VSMCs) and investigated underlying molecular mechanism of action. Rat primary VSMCs were pretreated with UA (10, 20 or 30μM) or amino guanidine (AG, 50μM) for 12h or with PI3K inhibitor LY294002 for 30min or with Akt inhibitor MK2206 for 24h, then 10% fetal bovine serum was used to induce proliferation. CCK-8 was used to assess cell proliferation. To explore the mechanism, cells were treated with UA (10, 20 or 30μM), LY294002 or MK2206, or transient transfected to inhibit miRNA-21 (miRNA-21) or to overexpress PTEN, then quantitative real-time PCR was used to assess the mRNA levels of miRNA-21 and phosphatase and tensin homolog (PTEN) for cells treated with UA or miRNA-21 inhibitor; western blotting was used to measure the protein levels of PTEN and PI3K. UA exerted significant anti-proliferation effects in rat primary VSMCs. Furthermore, UA inhibited the expression of miRNA-21 and subsequently enhanced the expression of PTEN. PTEN was found to inhibit the expression of PI3K. In conclusion, UA exerts anti-proliferation effects in rat primary VSMCs, which is associated with the inhibition of miRNA-21 expression and modulation of PTEN/PI3K signaling pathway.
Collapse
Affiliation(s)
- Qixiao Jiang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong, China
| | - Yantao Han
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong, China
| | - Hui Gao
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong, China
| | - Rong Tian
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong, China
| | - Ping Li
- The Affiliated Hospital of Qingdao University, 16 Jiangsu Road, Qingdao, Shandong, China
| | - Chunbo Wang
- Qingdao University Medical College, 308 Ningxia Road, Qingdao, Shandong, China.
| |
Collapse
|
45
|
Liu B, Piao X, Guo L, Liu S, Chai F, Gao L. Ursolic acid protects against ulcerative colitis via anti-inflammatory and antioxidant effects in mice. Mol Med Rep 2016; 13:4779-85. [PMID: 27082984 DOI: 10.3892/mmr.2016.5094] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Accepted: 02/16/2016] [Indexed: 11/05/2022] Open
Abstract
Ursolic acid (UA) has been reported to have a protective effect in colitis. However, the underlying mechanisms remain to be elucidated. In the present study, experimental ulcerative colitis was induced in male BALB/c mice by the administration of 5% dextran sulfate sodium (DSS) for 7 days, followed by treatment with UA for another 7 days. Hematoxylin & eosin staining was performed to evaluate colon tissue damage, and enzyme assays were used to measure malondialdehyde (MDA) content and superoxide dismutase (SOD) activity in colon homogenate. In addition, serum levels of interleukin (IL)‑1β and tumor necrosis factor (TNF)‑α were measured using an ELISA, and the level of nuclear factor (NF)‑κB p65 in the colonic tissues was assessed by western blotting. The 7‑day DSS administration induced marked colon damage, increased the serum levels of IL‑1β and TNF‑α, increased MDA content and decreased SOD activity in the colon homogenate. These changes were significantly improved by treatment with UA. UA also reduced the DSS‑stimulated high nuclear level of NF‑κB p65 in the colon tissues. These results demonstrate a protective role of UA in ulcerative colitis, and suggest that anti-inflammatory and antioxidant activities are involved in the underlying mechanisms.
Collapse
Affiliation(s)
- Baohai Liu
- Department of Gastroenterology, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Xuehua Piao
- Department of Traditional Chinese Medicine, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Lianyi Guo
- Department of Gastroenterology, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Shanshan Liu
- Department of Clinical Laboratory, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Fang Chai
- Department of General Surgery, The First Affiliated Hospital, Liaoning Medical University, Jinzhou, Liaoning 121001, P.R. China
| | - Leming Gao
- The 2nd Clinic, Stomatology Hospital, Peking University, Beijing 100101, P.R. China
| |
Collapse
|
46
|
Kashyap D, Tuli HS, Sharma AK. Ursolic acid (UA): A metabolite with promising therapeutic potential. Life Sci 2016; 146:201-13. [PMID: 26775565 DOI: 10.1016/j.lfs.2016.01.017] [Citation(s) in RCA: 198] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2015] [Revised: 01/11/2016] [Accepted: 01/12/2016] [Indexed: 12/12/2022]
Abstract
Plants are known to produce a variety of bioactive metabolites which are being used to cure various life threatening and chronic diseases. The molecular mechanism of action of such bioactive molecules, may open up new avenues for the scientific community to develop or improve novel therapeutic approaches to tackle dreadful diseases such as cancer and cardiovascular and neurodegenerative disorders. Ursolic acid (UA) is one among the categories of such plant-based therapeutic metabolites having multiple intracellular and extracellular targets that play role in apoptosis, metastasis, angiogenesis and inflammatory processes. Moreover, the synthetic derivatives of UA have also been seen to be involved in a range of pharmacological applications, which are associated with prevention of diseases. Evidences suggest that UA could be used as a potential candidate to develop a comprehensive competent strategy towards the treatment and prevention of health disorders. The review article herein describes the possible therapeutic effects of UA along with putative mechanism of action.
Collapse
Affiliation(s)
- Dharambir Kashyap
- Department of Medical Microbiology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, Punjab 160012, India
| | - Hardeep Singh Tuli
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Ambala, Haryana 133207, India.
| | - Anil K Sharma
- Department of Biotechnology, Maharishi Markandeshwar University, Mullana, Ambala, Haryana 133207, India
| |
Collapse
|
47
|
Anti-cancer and cardioprotective effects of indol-3-carbinol in doxorubicin-treated mice. J Infect Chemother 2016; 22:36-43. [PMID: 26603425 DOI: 10.1016/j.jiac.2015.10.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 09/28/2015] [Accepted: 10/06/2015] [Indexed: 12/17/2022]
|
48
|
Ko EY, Moon A. Natural Products for Chemoprevention of Breast Cancer. J Cancer Prev 2015; 20:223-31. [PMID: 26734584 PMCID: PMC4699749 DOI: 10.15430/jcp.2015.20.4.223] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 11/26/2015] [Accepted: 11/30/2015] [Indexed: 11/20/2022] Open
Abstract
Breast cancer is the primary cause of cancer death in women. Although current therapies have shown some promise against breast cancer, there is still no effective cure for the majority of patients in the advanced stages of breast cancer. Development of effective agents to slow, reduce, or reverse the incidence of breast cancer in high-risk women is necessary. Chemoprevention of breast cancer by natural products is advantageous, as these compounds have few side effects and low toxicity compared to synthetic compounds. In the present review, we summarize natural products which exert chemopreventive activities against breast cancer, such as curcumin, sauchinone, lycopene, denbinobin, genipin, capsaicin, and ursolic acid. This review examines the current knowledge about natural compounds and their mechanisms that underlie breast cancer chemopreventive activity both in vitro and in vivo. The present review may provide information on the use of these compounds for the prevention of breast cancer.
Collapse
Affiliation(s)
- Eun-Yi Ko
- College of Pharmacy, Duksung Women’s University, Seoul,
Korea
| | - Aree Moon
- College of Pharmacy, Duksung Women’s University, Seoul,
Korea
| |
Collapse
|
49
|
Shao F, Lv M, Zheng Y, Jiang J, Wang Y, Lv L, Wang J. The anti-tumour activity of rLj-RGD4, an RGD toxin protein from Lampetra japonica, on human laryngeal squamous carcinoma Hep-2 cells in nude mice. Biochimie 2015; 119:183-91. [DOI: 10.1016/j.biochi.2015.11.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Accepted: 11/03/2015] [Indexed: 01/15/2023]
|
50
|
Ishii M, Nakahara T, Ikeuchi S, Nishimura M. β-Amyrin induces angiogenesis in vascular endothelial cells through the Akt/endothelial nitric oxide synthase signaling pathway. Biochem Biophys Res Commun 2015; 467:676-82. [DOI: 10.1016/j.bbrc.2015.10.085] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Accepted: 10/17/2015] [Indexed: 11/16/2022]
|