1
|
Xu S, Wu J, Yang Q, Fang H, Xu T, He B, Chen N, Xing S. Isodon rubescens research literature based on Web of Science database for visual analysis: A review. Medicine (Baltimore) 2025; 104:e41945. [PMID: 40324265 PMCID: PMC12055185 DOI: 10.1097/md.0000000000041945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 05/07/2025] Open
Abstract
Isodon rubescens has been used as an herbal medicine in China for a long time. The significant value of development and utilization is affirmative. Bibliometrics is used as an approach to sort out, analyze, and visualize relevant literature in a particular field. So, it can intuitively express the research trends, hot directions, significant achievements, core journals, and outstanding authors in the field. But there is no bibliometrics analysis of I rubescens. The relevant dataset was retrieved and exported from the Web of Science database, and the results were obtained and visualized using the R Programming Language, CiteSpace, and VOSviewer, with the creation of time zone maps also using Scimago Graphica and Gephi. There were 506 valid data retrieved and 465 analyzed data selected. The country with the most significant number of publications is China, the institution with the largest annual publication volume is China Pharmaceutical University, the publication with the most relevant literature is the International Journal of Oncology, and the author with the most publications is Zhou. The keyword with the greatest intensity is "matastasis," which is also an emerging keyword. The role of I rubescens has been continuously diversifying. It has been proven to play a role in treating major diseases such as multiple cancers, leukemia, liver and kidney function impairment, and cardiovascular and cerebrovascular diseases. This study will highlight the main research direction in this field, namely the use of I rubescens for the treatment of cancer.
Collapse
Affiliation(s)
- Shaowei Xu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Jing Wu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Qingshan Yang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- Institute of Traditional Chinese Medicine Resources Protection and Development, Anhui Academy of Chinese Medicine, Hefei, China
| | - Huqiang Fang
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Teng Xu
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Bing He
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Na Chen
- Joint Research Center for Chinese Herbal Medicine of Anhui, Institute of Health and Medicine, Hefei Comprehensive National Science Center, Bozhou, China
| | - Shihai Xing
- College of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
- MOE-Anhui Joint Collaborative Innovation Center for Quality Improvement of Anhui Genuine Chinese Medicinal Materials, Hefei, China
- Anhui Province Key Laboratory of Research & Development of Chinese Medicine, Anhui University of Chinese Medicine, Hefei, China
| |
Collapse
|
2
|
Su Y, Liu L, Lin C, Deng D, Li Y, Huang M, Wang Y, Ling K, Wang H, Chen Q, Huang G. Enhancing cancer therapy: advanced nanovehicle delivery systems for oridonin. Front Pharmacol 2024; 15:1476739. [PMID: 39691396 PMCID: PMC11649421 DOI: 10.3389/fphar.2024.1476739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 11/18/2024] [Indexed: 12/19/2024] Open
Abstract
Oridonin (ORI), an ent-kaurane diterpenoid derived from Rabdosia rubescens (Hemsl.) H.Hara, serves as the primary bioactive component of this plant. It demonstrates a broad spectrum of therapeutic activities, including moderate to potent anticancer properties, alongside anti-inflammatory, antibacterial, antifibrotic, immunomodulatory, and neuromodulatory effects, thus influencing diverse biological processes. However, its clinical potential is significantly constrained by poor aqueous solubility and limited bioavailability. In alignment with the approach of developing drug candidates from natural compounds, various strategies, such as structural modification and nanocarrier systems, have been employed to address these challenges. This review provides an overview of ORI-based nano-delivery systems, emphasizing their potential to improve the clinical applicability of oridonin in oncology. Although some progress has been made in advancing ORI nano-delivery research, it remains insufficient for clinical implementation, necessitating further investigation.
Collapse
Affiliation(s)
- Yilin Su
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Lisha Liu
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Chongyang Lin
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Dashi Deng
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yunfei Li
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Mou Huang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Yu Wang
- Institute of Pain, The Affiliated Hospital of Southwest Jiaotong University, The Chengdu Third People’s Hospital, Chengdu, China
| | - Kangqiu Ling
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Haobing Wang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Qiyu Chen
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| | - Guixiao Huang
- Institute of Urology, The Third Affiliated Hospital of Shenzhen University, Shenzhen University, Shenzhen, China
| |
Collapse
|
3
|
Ali MA, Khan N, Ali A, Akram H, Zafar N, Imran K, Khan T, Khan K, Armaghan M, Palma‐Morales M, Rodríguez‐Pérez C, Caunii A, Butnariu M, Habtemariam S, Sharifi‐Rad J. Oridonin from Rabdosia rubescens: An emerging potential in cancer therapy - A comprehensive review. Food Sci Nutr 2024; 12:3046-3067. [PMID: 38726411 PMCID: PMC11077219 DOI: 10.1002/fsn3.3986] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 01/11/2024] [Accepted: 01/15/2024] [Indexed: 05/12/2024] Open
Abstract
Cancer incidences are rising each year. In 2020, approximately 20 million new cancer cases and 10 million cancer-related deaths were recorded. The World Health Organization (WHO) predicts that by 2024 the incidence of cancer will increase to 30.2 million individuals annually. Considering the invasive characteristics of its diagnostic procedures and therapeutic methods side effects, scientists are searching for different solutions, including using plant-derived bioactive compounds, that could reduce the probability of cancer occurrence and make its treatment more comfortable. In this regard, oridonin (ORI), an ent-kaurane diterpenoid, naturally found in the leaves of Rabdosia rubescens species, has been found to have antitumor, antiangiogenesis, antiasthmatic, antiinflammatory, and apoptosis induction properties. Extensive research has been performed on ORI to find various mechanisms involved in its anticancer activities. This review article provides an overview of ORI's effectiveness on murine and human cancer populations from 1976 to 2022 and provides insight into the future application of ORI in different cancer therapies.
Collapse
Affiliation(s)
| | - Noohela Khan
- Department of Nutrition SciencesRashid Latif Medical CollegeLahorePakistan
| | - Ahmad Ali
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Hira Akram
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Noushaba Zafar
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Kinza Imran
- Department of Food Science and Human NutritionUVASLahorePakistan
| | - Tooba Khan
- Department of Healthcare Biotechnology, Atta‐ur‐Rahman School of Applied BiosciencesNational University of Sciences and TechnologyIslamabadPakistan
| | | | - Muhammad Armaghan
- Department of Healthcare Biotechnology, Atta‐ur‐Rahman School of Applied BiosciencesNational University of Sciences and TechnologyIslamabadPakistan
| | - Marta Palma‐Morales
- Departamento de Nutrición y Bromatología, Facultad de FarmaciaUniversidad de GranadaGranadaSpain
- Instituto de Nutrición y Tecnología de los Alimentos ‘José Mataix’Universidad de GranadaGranadaSpain
| | - Celia Rodríguez‐Pérez
- Departamento de Nutrición y Bromatología, Facultad de FarmaciaUniversidad de GranadaGranadaSpain
- Instituto de Nutrición y Tecnología de los Alimentos ‘José Mataix’Universidad de GranadaGranadaSpain
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA)GranadaSpain
| | - Angela Caunii
- “Victor Babes” University of Medicine and PharmacyTimisoaraRomania
| | - Monica Butnariu
- University of Life Sciences "King Mihai I" from TimisoaraTimisoaraRomania
| | - Solomon Habtemariam
- Pharmacognosy Research & Herbal Analysis Services UKUniversity of GreenwichKentUK
| | | |
Collapse
|
4
|
Enhancement of cytotoxic and antioxidant activities of Digenea simplex chloroform extract using the nanosuspension technique. Bioprocess Biosyst Eng 2023; 46:279-296. [PMID: 36536224 PMCID: PMC9879839 DOI: 10.1007/s00449-022-02833-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Accepted: 12/05/2022] [Indexed: 12/24/2022]
Abstract
Digenea simplex (D. simplex), an Egyptian marine red macroalga, contains a diverse group of phytochemicals with unique bioactivities. At the same time, the synthesis of nanosuspension (NS) has received increasing interest to optimize the technological aspects of drugs. Thence, the main objective of this work was to use the chloroform extract (ChlE) of D. simplex to prepare its nanosuspension (ChlE-NS) formulation to increase its aqueous solubility, thereby improving its bioactivity. By using FTIR, GC/MS analysis, and phytochemical screening assays, the chemical profiling of ChlE was assessed. NS was prepared by the antisolvent precipitation technique using 1.5% w/v polyvinyl alcohol (PVA). A light microscope, FTIR, particle size distribution, polydispersity index (PDI), and zeta potential (ZP) measurements was used to characterize the prepared NS. Four cancer cell lines were used in the MTT experiment to investigate the anticancer potential of ChlE and ChlE-NS. An apoptotic mechanism was established using acridine orange/ethidium bromide (AO/EB) dual staining, DNA fragmentation, and increased caspase activity. ChlE and ChlE-NS were also evaluated as antioxidants using DPPH and ABTS free radical assays. The results showed that, when compared to ChlE, ChlE-NS had greater cytotoxic activity against the four cancer cell lines. However, results of antioxidant activity showed that ChlE-NS had an IC50 of 36.86 ± 0.09 and 63.5 ± 0.47%, while ChlE had values of 39.90 ± 0.08 and 86.5 ± 0.8% in DPPH and ABTS assays, respectively. Based on the results of this research, D. simplex ChlE-NS may be an effective strategy for enhancing ChlE's cytotoxic and antioxidant activities.
Collapse
|
5
|
Isoliquiritigenin Nanoemulsion Preparation by Combined Sonication and Phase-Inversion Composition Method: In Vitro Anticancer Activities. Bioengineering (Basel) 2022; 9:bioengineering9080382. [PMID: 36004907 PMCID: PMC9404772 DOI: 10.3390/bioengineering9080382] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/06/2022] [Accepted: 08/08/2022] [Indexed: 11/16/2022] Open
Abstract
Isoliquiritigenin (ILQ) has a number of biological activities such as antitumor and anti-inflammatory effects. However, biomedical applications of ILQ are impeded by its poor aqueous solubility. Therefore, in this research, we prepared a novel ILQ-loaded nanoemulsion, i.e., ILQ-NE, which consisted of Labrafil® M 1944 CS (oil), Cremophor® EL (surfactant), ILQ, and phosphate-buffered saline, by employing a combined sonication (high-energy) and phase-inversion composition (low-energy) method (denoted as the SPIC method). The ILQ-NE increased the ILQ solubility ~1000 times more than its intrinsic solubility. It contained spherical droplets with a mean diameter of 44.10 ± 0.28 nm and a narrow size distribution. The ILQ loading capacity was 4%. The droplet size of ILQ-NE remained unchanged during storage at 4 °C for 56 days. Nanoemulsion encapsulation effectively prevented ILQ from degradation under ultraviolet light irradiation, and enhanced the ILQ in vitro release rate. In addition, ILQ-NE showed higher cellular uptake and superior cytotoxicity to 4T1 cancer cells compared with free ILQ formulations. In conclusion, ILQ-NE may facilitate the biomedical application of ILQ, and the SPIC method presents an attractive avenue for bridging the merits and eliminating the shortcomings of traditional high-energy methods and low-energy methods.
Collapse
|
6
|
John R, Dalal B, Shankarkumar A, Devarajan PV. Innovative Betulin Nanosuspension exhibits enhanced anticancer activity in a Triple Negative Breast Cancer Cell line and Zebrafish angiogenesis model. Int J Pharm 2021; 600:120511. [PMID: 33766639 DOI: 10.1016/j.ijpharm.2021.120511] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2021] [Revised: 02/26/2021] [Accepted: 03/17/2021] [Indexed: 01/11/2023]
Abstract
We present a nanosuspension of betulin, a BCS class II anticancer drug, particularly effective against resistant breast cancer. As anticancer efficacy of betulin is hampered by poor aqueous solubility, a nanosuspension with surface area was considered to enhance efficacy. An innovative approach wherein the betulin nanosuspension is generated instantaneously in situ, by adding a betulin preconcentrate (BeTPC) comprising drug and excipients, to aqueous medium, is successfully demonstrated. The optimal BeTPC when added to isotonic dextrose solution instantaneously generated an in situ nanosuspension (BeTNS-15) with high precipitation efficiency (92.7 ± 1.21%), average particle size (383.74 ± 7.24 nm) and good stability as per ICH guidelines. TEM revealed elongated particles while DSC and XRD indicated partial amorphization. Significantly higher cytotoxicity of BeTNS-15 (IC50 38.44 µg/ml) compared to betulin (BetS) (IC50 69.54 µg/ml) in the resistant triple negative human breast cancer cell line MDA-MB-231, was attributed to high intracellular uptake confirmed by HPLC and Imaging Flow cytometry (IFC). IFC confirmed superior anti-cancer efficacy of BeTNS-15 mediated by mitochondrial membrane disruption and inhibition of the G0/G1 phase. BeTNS-15 also exhibited significantly greater anti-angiogenic efficacy (p < 0.05) in the zebrafish model confirming superior efficacy. Simplicity of the innovative in situ approach coupled with superior efficacy proposes BeTNS as an innovative and highly promising anticancer formulation.
Collapse
Affiliation(s)
- Rijo John
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Elite Status and Centre of Excellence (Maharashtra), N.P. Marg, Matunga East, Mumbai, Maharashtra 400019, India
| | - Bhavik Dalal
- Transfusion Transmitted Diseases Department, ICMR-National Institute of Immunohaematology, KEM Hospital Campus, Parel, Mumbai, Maharashtra 400012, India
| | - Aruna Shankarkumar
- Transfusion Transmitted Diseases Department, ICMR-National Institute of Immunohaematology, KEM Hospital Campus, Parel, Mumbai, Maharashtra 400012, India
| | - Padma V Devarajan
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Deemed University, Elite Status and Centre of Excellence (Maharashtra), N.P. Marg, Matunga East, Mumbai, Maharashtra 400019, India.
| |
Collapse
|
7
|
Taymouri S, Ahmadi Z, Mirian M, Tavakoli N. Simvastatin nanosuspensions prepared using a combination of pH-sensitive and timed-release approaches for potential treatment of colorectal cancer. Pharm Dev Technol 2021; 26:335-348. [PMID: 33430677 DOI: 10.1080/10837450.2021.1872086] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Abstract
A dual pH- and time-dependent polymeric coated capsule was developed to achieve the site specificity of simvastatin (SIM) release in the colon. To improve the SIM solubility, soluplus-based nanosuspension of the drug were prepared by applying the anti-solvent crystallization technique; this was then followed by lyophilization. Particle size, polydispersity index, and saturation solubility were evaluated. The optimized nanosuspension was combined with SLS and freeze-dried before filling into hard gelatin capsules. Drug release characteristics of the coated capsules were studied in HCl 0.1 N, the phosphate buffers 6.8 and 7.4, and the simulated colonic fluid (pH 6.8). The in-vitro cytotoxic effects of SIM nanoparticles against HT29 cells were then evaluated using the MTT assay. The prepared nanoparticles were spherical with a mean size of 261.66 nm, the zeta potential of -18.20 and the dissolution efficiency of 59.71%. X-ray diffraction and differential scanning calorimetry studies showed that the nanosizing technique transformed the crystalline drug into the more soluble amorphous form. The coated capsules had no release in the gastric media, providing the specific delivery of SIM in the colon. The cytotoxic effect of the SIM nanoparticles was significantly increased, as compared to the free SIM. The findings, therefore, showed that the coated capsules using the two polymers of ethyl cellulose and Eudragit S100 could be suitable for the colon target delivery of SIM.
Collapse
Affiliation(s)
- Somayeh Taymouri
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Zoha Ahmadi
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Mina Mirian
- Department of Biotechnology, School of Pharmacy and Pharmaceutical Sciences, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Naser Tavakoli
- Department of Pharmaceutics, School of Pharmacy and Novel Drug Delivery Systems Research Centre, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
8
|
Abdallah ME, El-Readi MZ, Althubiti MA, Almaimani RA, Ismail AM, Idris S, Refaat B, Almalki WH, Babakr AT, Mukhtar MH, Abdalla AN, Idris OF. Tamoxifen and the PI3K Inhibitor: LY294002 Synergistically Induce Apoptosis and Cell Cycle Arrest in Breast Cancer MCF-7 Cells. Molecules 2020; 25:E3355. [PMID: 32722075 PMCID: PMC7436112 DOI: 10.3390/molecules25153355] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Revised: 07/18/2020] [Accepted: 07/21/2020] [Indexed: 02/07/2023] Open
Abstract
Breast cancer is considered as one of the most aggressive types of cancer. Acquired therapeutic resistance is the major cause of chemotherapy failure in breast cancer patients. To overcome this resistance and to improve the efficacy of treatment, drug combination is employed as a promising approach for this purpose. The synergistic cytotoxic, apoptosis inducing, and cell cycle effects of the combination of LY294002 (LY), a phosphatidylinositide-3-kinase (PI3K) inhibitor, with the traditional cytotoxic anti-estrogen drug tamoxifen (TAM) in breast cancer cells (MCF-7) were investigated. LY and TAM exhibited potent cytotoxic effect on MCF-7 cells with IC50 values 0.87 µM and 1.02 µM. The combination of non-toxic concentration of LY and TAM showed highly significant synergistic interaction as observed from isobologram (IC50: 0.17 µM, combination index: 0.18, colony formation: 9.01%) compared to untreated control. The percentage of early/late apoptosis significantly increased after treatment of MCF-7 cells with LY and TAM combination: 40.3%/28.3% (p < 0.001), compared to LY single treatment (19.8%/11.4%) and TAM single treatment (32.4%/5.9%). In addition, LY and TAM combination induced the apoptotic genes Caspase-3, Caspase-7, and p53, as well as p21 as cell cycle promotor, and significantly downregulated the anti-apoptotic genes Bcl-2 and survivin. The cell cycle assay revealed that the combination induced apoptosis by increasing the pre-G1: 28.3% compared to 1.6% of control. pAKT and Cyclin D1 protein expressions were significantly more downregulated by the combination treatment compared to the single drug treatment. The results suggested that the synergistic cytotoxic effect of LY and TAM is achieved by the induction of apoptosis and cell cycle arrest through cyclin D1, pAKT, caspases, and Bcl-2 signaling pathways.
Collapse
Affiliation(s)
- Mohamed E. Abdallah
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Mahmoud Zaki El-Readi
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
- Department of Biochemistry, Faculty of Pharmacy, Al-Azhar University, Assiut 71524, Egypt
| | - Mohammad Ahmad Althubiti
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Riyad Adnan Almaimani
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Amar Mohamed Ismail
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Al-Neelain University, Khartoum 11121, Sudan; (A.M.I.); (O.F.I.)
| | - Shakir Idris
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 7607, Saudi Arabia; (S.I.); (B.R.)
| | - Bassem Refaat
- Department of Laboratory Medicine, Faculty of Applied Medical Sciences, Umm Al-Qura University, Makkah 7607, Saudi Arabia; (S.I.); (B.R.)
| | - Waleed Hassan Almalki
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
| | - Abdullatif Taha Babakr
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Mohammed H. Mukhtar
- Department of Biochemistry, Faculty of Medicine, Umm Al-Qura University, Makkah 21955, Saudi Arabia; (M.E.A.); (M.A.A.); (R.A.A.); (A.T.B.); (M.H.M.)
| | - Ashraf N. Abdalla
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Umm Al-Qura University, Makkah 21955, Saudi Arabia;
- Department of Pharmacology and Toxicology, Medicinal and Aromatic Plants Research Institute, National Center for Research, Khartoum 2404, Sudan
| | - Omer Fadul Idris
- Department of Biochemistry and Molecular Biology, Faculty of Science and Technology, Al-Neelain University, Khartoum 11121, Sudan; (A.M.I.); (O.F.I.)
| |
Collapse
|
9
|
Md S, Alhakamy NA, Akhter S, Awan ZAY, Aldawsari HM, Alharbi WS, Haque A, Choudhury H, Sivakumar PM. Development of Polymer and Surfactant Based Naringenin Nanosuspension for Improvement of Stability, Antioxidant, and Antitumour Activity. J CHEM-NY 2020; 2020:1-10. [DOI: 10.1155/2020/3489393] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
Nanosuspensions are widely reported to enhance the solubility of poorly soluble drugs. In addition to enhancement in solubility, improvement of stability and therapeutic efficacy would be an added advantage. In the present study, premilling and subsequent high-pressure homogenization were carried out to produce naringenin nanosuspension. Hydroxypropyl methylcellulose and sodium dodecyl sulfate were evaluated for their performance as stabilizers under various homogenization cycles. The prepared nanosuspensions were studied for average particle size and size distribution, zeta potential, solubility, drug release, antioxidant activity, and in vitro antitumor activity. It was observed that both hydroxypropyl methylcellulose-stabilized nanosuspension and sodium dodecyl sulfate-stabilized nanosuspension produced an enhancement in physical stability, antioxidant potential, and in vitro cytotoxicity compared with naringenin. Furthermore, hydroxypropyl methylcellulose-stabilized nanosuspension was found to be better than sodium dodecyl sulfate-stabilized nanosuspension in terms of particle size and size distribution, storage stability, and drug release. This study showed that nanosuspension formulations could be a potential strategy for improving dissolution and antitumor activity of naringenin.
Collapse
Affiliation(s)
- Shadab Md
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Nabil A. Alhakamy
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sohail Akhter
- Department of New Product Development, Combination Product & Devices, Global R&D, Teva Pharmaceuticals, Runcorn, UK
| | - Zuhier A. Y. Awan
- Department of Medicine and Biochemistry, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Hibah M. Aldawsari
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
- Center of Excellence for Drug Research & Pharmaceutical Industries, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Waleed S. Alharbi
- Department of Pharmaceutics, Faculty of Pharmacy, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Anzarul Haque
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-kharj, Saudi Arabia
| | - Hira Choudhury
- Department of Pharmaceutical Technology, School of Pharmacy, International Medical University, Jalan Jalil Perkasa, Bukit Jalil, Kuala Lumpur 57000, Malaysia
| | | |
Collapse
|
10
|
Zhang Y, Wang S, Dai M, Nai J, Zhu L, Sheng H. Solubility and Bioavailability Enhancement of Oridonin: A Review. Molecules 2020; 25:E332. [PMID: 31947574 PMCID: PMC7024198 DOI: 10.3390/molecules25020332] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2019] [Revised: 01/07/2020] [Accepted: 01/09/2020] [Indexed: 12/12/2022] Open
Abstract
Oridonin (ORI), an ent-kaurene tetracyclic diterpenoid compound, is isolated from Chinese herb Rabdosia rubescens with various biological and pharmacological activities including anti-tumor, anti-microbial and anti-inflammatory effects. However, the clinical application of ORI is limited due to its low solubility and poor bioavailability. In order to overcome these shortcomings, many strategies have been explored such as structural modification, new dosage form, etc. This review provides a detailed discussion on the research progress to increase the solubility and bioavailability of ORI.
Collapse
Affiliation(s)
| | | | | | | | - Liqiao Zhu
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Z.); (S.W.); (M.D.); (J.N.)
| | - Huagang Sheng
- College of pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China; (Y.Z.); (S.W.); (M.D.); (J.N.)
| |
Collapse
|
11
|
Moustafa AMY, Bakare SB. Synthesis of Some Hybrid 7-Hydroxy Quinolinone Derivatives As Anti Breast Cancer Drugs. RESEARCH ON CHEMICAL INTERMEDIATES 2019. [DOI: 10.1007/s11164-019-03827-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
|
12
|
Targeting the NRF-2/RHOA/ROCK signaling pathway with a novel aziridonin, YD0514, to suppress breast cancer progression and lung metastasis. Cancer Lett 2018; 424:97-108. [DOI: 10.1016/j.canlet.2018.03.029] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2018] [Revised: 03/06/2018] [Accepted: 03/21/2018] [Indexed: 01/08/2023]
|
13
|
Gatti G, Lucini V, Dugnani S, Calastretti A, Spadoni G, Bedini A, Rivara S, Mor M, Canti G, Scaglione F, Bevilacqua A. Antiproliferative and pro-apoptotic activity of melatonin analogues on melanoma and breast cancer cells. Oncotarget 2017; 8:68338-68353. [PMID: 28978121 PMCID: PMC5620261 DOI: 10.18632/oncotarget.20124] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 07/29/2017] [Indexed: 12/15/2022] Open
Abstract
Melatonin plays different physiological functions ranging from the regulation of circadian rhythms to tumor inhibition, owing to its antioxidant, immunomodulatory and anti-aging properties. Due to its pleiotropic functions, melatonin has been shown to elicit cytoprotective processes in normal cells and trigger pro-apoptotic signals in cancer cells. The therapeutic potential of melatonin analogues prompted us to investigate the in vitro and in vivo antitumor activity of new melatonin derivatives and explore the underlying molecular mechanisms. The experiments revealed that the new melatonin analogues inhibited the growth of melanoma and breast cancer cells in a dose- and time-dependent manner. In addition, our results indicated that melatonin derivative UCM 1037 could induce apoptosis in melanoma and breast cancer cells, as well as cell necrosis, in MCF-7. Together, apoptosis and necrosis could be two possible mechanisms to explain the cytotoxic effect of the melatonin analogue against cancer cells. The suppression of tumor growth by the melatonin analogues was further demonstrated in vivo in a xenograft mice model. A decrease in the activation of MAPK pathway was observed in all cancer cells following UCM 1037 treatment. Overall, this study describes a promising antitumor compound showing antiproliferative and cytotoxic activity in melanoma and breast cancer cells.
Collapse
Affiliation(s)
- Giuliana Gatti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Valeria Lucini
- Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - Silvana Dugnani
- Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - Angela Calastretti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Gilberto Spadoni
- Department of Biomolecular Sciences, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Annalida Bedini
- Department of Biomolecular Sciences, Università degli Studi di Urbino "Carlo Bo", Urbino, Italy
| | - Silvia Rivara
- Department of Food and Drug, Università degli Studi di Parma, Parma, Italy
| | - Marco Mor
- Department of Food and Drug, Università degli Studi di Parma, Parma, Italy
| | - Gianfranco Canti
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| | - Francesco Scaglione
- Department of Oncology and Hemato-oncology, Università degli Studi di Milano, Milan, Italy
| | - Annamaria Bevilacqua
- Department of Medical Biotechnology and Translational Medicine, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
14
|
Targeting 3-phosphoinositide-dependent protein kinase 1 associated with drug-resistant renal cell carcinoma using new oridonin analogs. Cell Death Dis 2017; 8:e2701. [PMID: 28333136 PMCID: PMC5386527 DOI: 10.1038/cddis.2017.121] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/15/2017] [Accepted: 02/16/2017] [Indexed: 01/06/2023]
Abstract
The current agents used for renal cell carcinoma (RCC) only exhibit the moderate response rate among patients. Development of drug resistance eventually fuels the need of either more potent drugs or new drugs to target the resistant pathways. Oridonin is a diterpenoid isolated from the Chinese medicinal herb Rabdosia rubescens and has been shown to have antitumor activities in many cancers. We previously developed new synthetic methodologies to modify structurally diversified diterpenoids and designed a series of nitrogen-enriched oridonin analogs. In this study, we screened a variety of oridonin analogs based on their cytotoxicity using MTT assay and identify the most potent candidate, namely, CYD-6-17. CYD-6-17 exhibited a high potency to inhibit the in vitro growth of several drug-resistant RCC cells as well as endothelial cells stimulated by tumor cells at nanomolar range. Delivery of CYD-6-17 significantly inhibited RCC tumor growth using xenograft model. Mechanistically, it targeted the 3-phosphoinositide-dependent protein kinase 1 gene that appeared to be a potent regulator of AKT and was associated with patient survival after targeted therapies. This offers a new rational therapeutic regimen of CYD-6-17 to drug-resistant RCC based on its novel mechanism of action.
Collapse
|
15
|
Wang S, He M, Li L, Liang Z, Zou Z, Tao A. Cell-in-Cell Death Is Not Restricted by Caspase-3 Deficiency in MCF-7 Cells. J Breast Cancer 2016; 19:231-241. [PMID: 27721872 PMCID: PMC5053307 DOI: 10.4048/jbc.2016.19.3.231] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 07/20/2016] [Indexed: 12/30/2022] Open
Abstract
Purpose Cell-in-cell structures are created by one living cell entering another homotypic or heterotypic living cell, which usually leads to the death of the internalized cell, specifically through caspase-dependent cell death (emperitosis) or lysosome-dependent cell death (entosis). Although entosis has attracted great attention, its occurrence is controversial, because one cell line used in its study (MCF-7) is deficient in caspase-3. Methods We investigated this issue using MCF-7 and A431 cell lines, which often display cell-in-cell invasion, and have different levels of caspase-3 expression. Cell-in-cell death morphology, microstructures, and signaling pathways were compared in the two cell lines. Results Our results confirmed that MCF-7 cells are caspase-3 deficient with a partial deletion in the CASP-3 gene. These cells underwent cell death that lacked typical apoptotic properties after staurosporine treatment, whereas caspase-3-sufficient A431 cells displayed typical apoptosis. The presence of caspase-3 was related neither to the lysosome-dependent nor to the caspase-dependent cell-in-cell death pathway. However, the existence of caspase-3 was associated with a switch from lysosome-dependent cell-in-cell death to the apoptotic cell-in-cell death pathway during entosis. Moreover, cellular hypoxia, mitochondrial swelling, release of cytochrome C, and autophagy were observed in internalized cells during entosis. Conclusion The occurrence of caspase-independent entosis is not a cell-specific process. In addition, entosis actually represents a cellular self-repair system, functioning through autophagy, to degrade damaged mitochondria resulting from cellular hypoxia in cell-in-cell structures. However, sustained autophagy-associated signal activation, without reduction in cellular hypoxia, eventually leads to lysosome-dependent intracellular cell death.
Collapse
Affiliation(s)
- Shan Wang
- The State Key Clinical Specialty in Allergy, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.; The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Meifang He
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Linmei Li
- The State Key Clinical Specialty in Allergy, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.; The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Zhihua Liang
- The State Key Clinical Specialty in Allergy, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.; The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Zehong Zou
- The State Key Clinical Specialty in Allergy, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.; The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| | - Ailin Tao
- The State Key Clinical Specialty in Allergy, the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.; Guangdong Provincial Key Laboratory of Allergy & Clinical Immunology, Guangzhou Medical University, Guangzhou, China.; The State Key Laboratory of Respiratory Disease, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
16
|
Abstract
Oridonin has attracted considerable attention in the last decade because of its anti-cancer pharmacological properties. This ent-kaurane diterpenoid, isolated from the Chinese herb Rabdosia rubescens and some related species, has
demonstrated great potential in the treatment profile of many diseases by exerting anti-tumor, anti-inflammatory, pro-apoptotic, and neurological effects. Unfortunately, the mechanisms via which oridonin exerts these effects remain poorly understood. This review provides an overview of the multifunctional effects of oridonin as well as the reasons for its potential for investigations in the treatment of many diseases other than cancer.
Collapse
Affiliation(s)
- Brice Ayissi Owona
- Division of Immunopathology of the Nervous System, Institute of Pathology and Neuropathology, University of Tübingen, Calwer Street 3, Tübingen, Germany,
| | | |
Collapse
|
17
|
Wu QX, Yuan SX, Ren CM, Yu Y, Sun WJ, He BC, Wu K. Oridonin upregulates PTEN through activating p38 MAPK and inhibits proliferation in human colon cancer cells. Oncol Rep 2016; 35:3341-8. [PMID: 27108927 DOI: 10.3892/or.2016.4735] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2015] [Accepted: 01/12/2016] [Indexed: 11/06/2022] Open
Abstract
Oridonin (ORI) has been reported as an antiproliferation and apoptosis-inducing natural product in various cancer cells. However, the exact molecular mechanism underlying these effects remains unclear. In the present study, we demonstrated the antiproliferation effect of ORI in HCT116 cells, and analyzed the possible molecular mechanism which mediates this effect. We found that ORI inhibits proliferation, induces cell cycle arrest and apoptosis in HCT116 cells, thus also tumor growth. Mechanically, we found that ORI has no substantial effect on mRNA expression of phosphatase and tensin homologue (PTEN), but increases the total protein level of PTEN and markedly reduces the phosphorylation of PTEN; Exogenous expression of PTEN potentiates the anticancer effect of ORI, while knockdown of PTEN attenuates it. ORI also increases the phosphorylation of p38 MAPK, and p38 MAPK-specific inhibitor reduces the antiproliferation effect ORI in HCT116 cells. Moreover, inhibition of p38 MAPK increases the phosphorylation of PTEN, and reverses ORI-induced decrease of PTEN phosphorylation. Our findings suggested that ORI may be a potential anticancer drug for colon cancer, this effect may be mediated by enhancing the function of PTEN through reducing its phosphorylation, which may be resulted from the ORI-induced activation of p38 MAPK.
Collapse
Affiliation(s)
- Qiu-Xiang Wu
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Shuang-Xue Yuan
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Chun-Mei Ren
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Yu Yu
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Wen-Juan Sun
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Bai-Cheng He
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| | - Ke Wu
- Chongqing Municipal Key Laboratory of Higher Education Institutions for Biochemistry and Molecular Pharmacology, School of Pharmacy, Chongqing Medical University, Yuzhong, Chongqing 400016, P.R. China
| |
Collapse
|
18
|
Wang Y, Song J, Chow SF, Chow AHL, Zheng Y. Particle size tailoring of ursolic acid nanosuspensions for improved anticancer activity by controlled antisolvent precipitation. Int J Pharm 2015; 494:479-89. [PMID: 26302857 DOI: 10.1016/j.ijpharm.2015.08.052] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Revised: 08/10/2015] [Accepted: 08/18/2015] [Indexed: 02/06/2023]
Abstract
The present study was aimed at tailoring the particle size of ursolic acid (UA) nanosuspension for improved anticancer activity. UA nanosuspensions were prepared by antisolvent precipitation using a four-stream multi-inlet vortex mixer (MIVM) under defined conditions of varying solvent composition, drug feeding concentration or stream flow rate. The resulting products were characterized for particle size and polydispersity. Two of the UA nanosuspensions with mean particle sizes of 100 and 300 nm were further assessed for their in-vitro activity against MCF-7 breast cancer cells using fluorescence microscopy with 4',6-diamidino-2-phenylindole (DAPI) staining, as well as flow cytometry with propidium (PI) staining and with double staining by fluorescein isothiocyanate. It was revealed that the solvent composition, drug feeding concentration and stream flow rate were critical parameters for particle size control of the UA nanosuspensions generated with the MIVM. Specifically, decreasing the UA feeding concentration or increasing the stream flow rate or ethanol content resulted in a reduction of particle size. Excellent reproducibility for nanosuspension production was demonstrated for the 100 and 300 nm UA preparations with a deviation of not more than 5% in particle size from the mean value of three independent batches. Fluorescence microscopy and flow cytometry revealed that these two different sized UA nanosuspensions, particularly the 300 nm sample, exhibited a higher anti-proliferation activity against the MCF-7 cells and afforded a larger population of these cells in both early and late apoptotic phases. In conclusion, MIVM is a robust and pragmatic tool for tailoring the particle size of the UA nanosuspension. Particle size appears to be a critical determinant of the anticancer activity of the UA nanoparticles.
Collapse
Affiliation(s)
- Yancai Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; School of Chemistry and Pharmaceutical Engineering, Qilu University of Technology, Jinan 250353, China
| | - Ju Song
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China; Beijing Aohe Pharmaceutical Research Institute Co. Ltd., Beijing 101113, China
| | - Shing Fung Chow
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong
| | - Albert H L Chow
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong.
| | - Ying Zheng
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao, China.
| |
Collapse
|
19
|
Singh J, Garg T, Rath G, Goyal AK. Advances in nanotechnology-based carrier systems for targeted delivery of bioactive drug molecules with special emphasis on immunotherapy in drug resistant tuberculosis – a critical review. Drug Deliv 2015; 23:1676-98. [DOI: 10.3109/10717544.2015.1074765] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
20
|
Oridonin triggers apoptosis in colorectal carcinoma cells and suppression of microRNA-32 expression augments oridonin-mediated apoptotic effects. Biomed Pharmacother 2015; 72:125-34. [DOI: 10.1016/j.biopha.2015.04.016] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2014] [Revised: 04/07/2015] [Accepted: 04/15/2015] [Indexed: 12/12/2022] Open
|
21
|
Hao L, Luan J, Zhang D, Li C, Guo H, Qi L, Liu X, Li T, Zhang Q. Research on the in vitro anticancer activity and in vivo tissue distribution of Amoitone B nanocrystals. Colloids Surf B Biointerfaces 2014; 117:258-66. [PMID: 24657612 DOI: 10.1016/j.colsurfb.2014.02.042] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 02/13/2014] [Accepted: 02/16/2014] [Indexed: 11/18/2022]
Abstract
Amoitone B, a natural agonist to Nur77, is a promising anticancer drug. However, its application is seriously restricted due to the water-insolubility and short biological half-life. Amoitone B nanocrystals (AmB-NC) were formulated by microfluidization method to overcome the above obstacles. This study aims to evaluate the cytotoxicity and tissue distribution of AmB-NC. MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide] assay revealed the improved in vitro antitumor activity of AmB-NC against stomach, colon, liver and lung cancer cells compared with Amoitone B solution. Meanwhile, observation of morphological changes, cell cycle and apoptosis examination using flow cytometry exhibited that AmB-NC could induce G1 cycle arrest and markedly enhance the apoptosis of human gastric cancer BGC-823 cell line. Tissue distribution study demonstrated that AmB-NC had a higher distribution in liver and lung, which was helpful for relevant cancer treatment. In conclusion, AmB-NC could be a potential delivery system for treatment of human cancer.
Collapse
Affiliation(s)
- Leilei Hao
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Jingjing Luan
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Dianrui Zhang
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China.
| | - Caiyun Li
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Hejian Guo
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Lisi Qi
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Xinquan Liu
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Tingting Li
- Department of Pharmaceutics, College of Pharmacy, Shandong University, 44 Wenhua Xilu, Jinan 250012, PR China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, 38 Xueyuan Road, Beijing 100083, PR China
| |
Collapse
|
22
|
Talekar M, Ganta S, Amiji M, Jamieson S, Kendall J, Denny WA, Garg S. Development of PIK-75 nanosuspension formulation with enhanced delivery efficiency and cytotoxicity for targeted anti-cancer therapy. Int J Pharm 2013; 450:278-89. [PMID: 23632263 DOI: 10.1016/j.ijpharm.2013.04.057] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Revised: 04/15/2013] [Accepted: 04/16/2013] [Indexed: 02/07/2023]
Abstract
PIK-75 is a phosphatidylinositol 3-kinase (PI3K) inhibitor that shows selectivity toward p110-α over the other PI3K class Ia isoforms p110-β and p110-δ, but it lacks solubility, stability and other kinase selectivity. The purpose of this study was to develop folate-targeted PIK-75 nanosuspension for tumor targeted delivery and to improve therapeutic efficacy in human ovarian cancer model. High pressure homogenization was used to prepare the non-targeted and targeted PIK-75 nanosuspensions which were characterized for size, zeta potential, entrapment efficiency, morphology, saturation solubility and dissolution velocity. In vitro analysis of drug uptake, cell viability and cell survival was conducted in SKOV-3 cells. Drug pharmacokinetics and pAkt expression were determined in SKOV-3 tumor bearing mice. PIK-75 nanosuspensions showed an improvement in dissolution velocity and an 11-fold increase in saturation solubility over pre-milled PIK-75. In vitro studies in SKOV-3 cells indicated a 2-fold improvement in drug uptake and 0.4-fold decrease in IC50 value of PIK-75 following treatment with targeted nanosuspension compared to non-targeted nanosuspension. The improvement in cytotoxicity was attributed to an increase in caspase 3/7 and hROS activity. In vivo studies indicated a 5-10-fold increased PIK-75 accumulation in the tumor with both the nanosuspension formulations compared to PIK-75 suspension. The targeted nanosuspension showed an enhanced downregulation of pAkt compared to non-targeted formulation system. These results illustrate the opportunity to formulate PIK-75 as a targeted nanosuspension to enhance uptake and cytotoxicity of the drug in tumor.
Collapse
Affiliation(s)
- Meghna Talekar
- School of Pharmacy, Faculty of Medical and Health Sciences, The University of Auckland, Private Bag 92019, Auckland, New Zealand
| | | | | | | | | | | | | |
Collapse
|
23
|
Sun KW, Ma YY, Guan TP, Xia YJ, Shao CM, Chen LG, Ren YJ, Yao HB, Yang Q, He XJ. Oridonin induces apoptosis in gastric cancer through Apaf-1, cytochrome c and caspase-3 signaling pathway. World J Gastroenterol 2012; 18:7166-74. [PMID: 23326121 PMCID: PMC3544018 DOI: 10.3748/wjg.v18.i48.7166] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2012] [Revised: 09/21/2012] [Accepted: 10/16/2012] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effect and mechanism of oridonin on the gastric cancer cell line HGC-27 in vitro.
METHODS: The inhibitory effect of oridonin on HGC-27 cells was detected using the 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay. After treatment with 10 μg/mL oridonin for 24 h and 48 h, the cells were stained with acridine orange/ethidium bromide. The morphologic changes were observed under an inverted fluorescence microscope. DNA fragmentation (a hallmark of apoptosis) and lactate dehydrogenase activity were examined using DNA ladder assay and lactate dehydrogenase-release assay. After treated with oridonin (0, 1.25, 2.5, 5 and 10 μg/mL), HGC-27 cells were collected for anexin V-phycoerythrin and 7-amino-actinomycin D double staining and tested by flow cytometric analysis, and oridonin- induced apoptosis in HGC-27 cells was detected. After treatment with oridonin for 24 h, the effects of oridonin on expression of Apaf-1, Bcl-2, Bax, caspase-3 and cytochrome c were also analyzed using reverse-transcript polymerase chain reaction (RT-PCR) and Western blotting.
RESULTS: Oridonin significantly inhibited the proliferation of HGC-27 cells in a dose- and time-dependent manner. The inhibition rates of HGC-27 treated with four different concentrations of oridonin for 24 h (1.25, 2.5, 5 and 10 μg/mL) were 1.78% ± 0.36%, 4.96% ± 1.59%, 10.35% ± 2.76% and 41.6% ± 4.29%, respectively, which showed a significant difference (P < 0.05). The inhibition rates of HGC-27 treated with oridonin at the four concentrations for 48 h were 14.77% ± 4.21%, 21.57% ± 3.75%, 30.31% ± 4.91% and 61.19% ± 5.81%, with a significant difference (P < 0.05). The inhibition rates of HGC-27 treated with oridonin for 72 h at the four concentrations were 25.77% ± 4.85%, 31.86% ± 3.86%, 48.30% ± 4.16% and 81.80% ± 6.72%, with a significant difference (P < 0.05). Cells treated with oridonin showed typical apoptotic features with acridine orange/ethidium bromide staining. After treatment with oridonin, the cells became round, shrank, and developed small buds around the nuclear membrane while forming apoptotic bodies. Lactate dehydrogenase (LDH) release assay showed that after treated with 1.25 μg/mL and 20 μg/mL oridonin for 24 h, LDH release of HGC-27 caused by apoptosis increased from 22.94% ± 3.8% to 52.68% ± 2.4% (P < 0.001). However, the change in the release of LDH caused by necrosis was insignificant, suggesting that the major cause of oridonin-induced HGC-27 cell death was apoptosis. Flow cytometric analysis also revealed that oridonin induced significant apoptosis compared with the controls (P < 0.05). And the apoptosis rates of HGC-27 induced by the four different concentrations of oridonin were 5.3% ± 1.02%, 12.8% ± 2.53%, 28.5% ± 4.23% and 49.6% ± 3.76%, which were in a dose-dependent manner (P < 0.05). After treatment for 24 h, DNA ladder showed that oridonin induced a significant increase in DNA fragmentation in a dose-dependent manner. RT-PCR revealed that mRNA expression levels were up-regulated compared with the controls in caspase-3 (0.917 ± 0.103 vs 0.357 ± 0.019, P < 0.05), cytochrome c (1.429 ± 0.111 vs 1.002 ± 0.014, P < 0.05), Apaf-1 (0.688 ± 0.101 vs 0.242 ± 0.037, P < 0.05) and Bax (0.856 ± 0.101 vs 0.278 ± 0.027, P < 0.05) (P < 0.05), whereas down-regulated in Bcl-2 (0.085 ± 0.012 vs 0.175 ± 0.030, P < 0.05). Western blotting analysis also confirmed this result.
CONCLUSION: Apoptosis of HGC-27 induced by oridonin may be associated with differential expression of Apaf-1, caspase-3 and cytochrome c, which are highly dependent upon the mitochondrial pathway.
Collapse
|
24
|
Abstract
Nanosuspension drug delivery has obtained great success in the preparation of insoluble drugs. The nanosuspension technology can confer a series of special characteristics to the drugs, such as the enhanced dissolution rate and saturation solubility. This mini review first described the differences between the nanocrystals and nanosuspensions. Next, the product techniques, the stable measures, the special features, and the routes of administration of the nanosuspensions were reviewed and compared. Finally, some existing shortcomings of the nanosuspensions were mentioned and the perspectives of the nanosuspensions were also made.
Collapse
Affiliation(s)
- Yue Liu
- Department of Pharmaceutics, College of Pharmacy, Shandong University, Jinan, PR China
| | | | | | | |
Collapse
|
25
|
Jilani K, Qadri SM, Zelenak C, Lang F. Stimulation of suicidal erythrocyte death by oridonin. Arch Biochem Biophys 2011; 511:14-20. [PMID: 21575590 DOI: 10.1016/j.abb.2011.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2011] [Revised: 04/26/2011] [Accepted: 05/01/2011] [Indexed: 01/04/2023]
Abstract
Oridonin triggers apoptosis of cancer cells and was suggested as anticancer agent. Oridonin is partially effective through mitochondrial depolarization and partially by modifying gene expression. Erythrocytes lack mitochondria and nuclei but may undergo eryptosis, a suicidal cell death characterized by cell shrinkage and cell membrane scrambling with phosphatidylserine exposure at the cell surface. Triggers of eryptosis include increase of cytosolic Ca(2+)-activity, ATP depletion and ceramide formation. The present study explored, whether oridonin triggers eryptosis. Cytosolic Ca(2+)-concentration was estimated from Fluo3-fluorescence, cell volume from forward scatter in FACS analysis, phosphatidylserine exposure from binding of fluorescent annexin V, hemolysis from hemoglobin release, ATP concentration utilizing a luciferin-luciferase assay and ceramide abundance utilizing fluorescent anti-ceramide antibodies. A 48 h exposure to oridonin (≥25μM) significantly increased cytosolic Ca(2+)-concentration, increased ceramide formation, decreased forward scatter and triggered annexin V-binding (the latter in >20% of the erythrocytes). Oridonin didn't decrease ATP concentration and hemolysed <5% of erythrocytes. The effects of oridonin on annexin V binding were partially reversed in the nominal absence of Ca(2+) and by the addition of amiloride (1mM). The present observations reveal a completely novel effect of oridonin, i.e. triggering of Ca(2+) entry and ceramide formation as well as suicidal death of erythrocytes.
Collapse
Affiliation(s)
- Kashif Jilani
- Department of Physiology, University of Tuebingen, Gmelinstraße 5, D-72076 Tuebingen, Germany
| | | | | | | |
Collapse
|