1
|
Shi F, Gong M, Adu-Frimpong M, Jiang X, Wang X, Hua Q, Li T, Li J, Yu J, Toreniyazov E, Cao X, Wang Q, Xu X. Isolation, Purification of Phenolic Glycoside 1 from Moringa oleifera Seeds and Formulation of Its Liposome Delivery System. AAPS PharmSciTech 2024; 25:196. [PMID: 39174848 DOI: 10.1208/s12249-024-02911-w] [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: 04/22/2024] [Accepted: 07/31/2024] [Indexed: 08/24/2024] Open
Abstract
In this study, N, N '-bis {4- [(α-L- rhamnosyloxy) benzyl]} thiourea (PG-1), a phenolic glycoside compound was purified from Moringa seed. The PG-1 has attracted extensive attention due to its anti-cancer, antioxidant, anti-inflammatory and hypoglycemic properties. However, some of its physicochemical properties such as oral bioavailability has not been studied. Herein, a highly purified PG-1 was extracted and incorporated in multiple layered liposomes (PG-1-L) to avoid its burst release and enhance oral bioavailability. After appropriate characterization, it was discovered that the obtained PG-1-L was stable, homogeneous and well dispersed with the average particle size being 89.26 ± 0.23 nm. Importantly, the in vitro release and in vivo oral bioavailability of PG-1-L were significantly improved compared with PG-1. In addition, MTT results showed that compared with the free PG-1, PG-1-L displayed obvious inhibitory effect on the HepG2 cells, while the inhibitory effect on healthy non-malignant 3T6 and LO-2 cells was not significant, indicating that PG-1-L had high safety. In conclusion, PG-1-L can be used as a promising delivery system and an ideal novel approach to improve the oral bioavailability and anticancer activity of PG-1.
Collapse
Affiliation(s)
- Feng Shi
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Mingjie Gong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), UK 0215-5321, Navrongo, Ghana
| | - Xia Jiang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Xiaowen Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Qinyang Hua
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Tingyuan Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiaying Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China
| | | | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
2
|
Alneghery LM, Al-Zharani M, Nasr FA, Eldin ZE, Al Hujran TA, Tawfeek HM, Fayed MH, Elbeltagi S. Fabrication and optimization of naringin-loaded MOF-5 encapsulated by liponiosomes as smart drug delivery, cytotoxicity, and apoptotic on breast cancer cells. Drug Dev Ind Pharm 2024:1-14. [PMID: 39101770 DOI: 10.1080/03639045.2024.2388786] [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: 06/01/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 08/06/2024]
Abstract
INTRODUCTION Cancers are regarded as hazardous due to their high worldwide death rate, with breast cancer (BC), which affects practically all cancer patients globally, playing a significant role in this statistic. The therapeutic approach for BC has not advanced using standard techniques, such as specialized naringin (NG) chemotherapy. Instead, a novel strategy has been utilized to enhance smart drug delivery (SDD) to tumors. SIGNIFICANCE Herein, we established NG-loaded zinc metal-organic framework-5 (NG-MOF-5) coated with liponiosomes (LNs) to manufacture NG-MOF-5@LNs nanoparticles (NPs) for antibacterial and cancer treatment. METHODS MOF-5, NG, and NG-MOF-5@LNs were evaluated with XRD, thermogravimetric analysis (TGA), FTIR, SEM, TEM, PDI, ZP, encapsulation efficiency (EE), loading efficiency (LE), and drug release (DR) kinetics. We examined the antibacterial activity involving minimum inhibitory concentration (MIC) and zone of inhibition by NG, MOF-5, and NG-MOF-5@LNs. The cell viability, necrosis, and total apoptosis (late and early) were evaluated for anti-cancer activity against MCF-7 BC cells. RESULTS TEM results demonstrated that NG-MOF-5@LNs formed monodispersed spherical-like particles with a size of 122.5 nm, PDI of 0.139, and ZP of +21 mV. The anti-microbial activity results indicated that NG-MOF-5@LNs exhibited potent antibacterial effects, as evidenced by inhibition zones and MIC values. The Higuchi model indicates an excellent fit (R2 = 0.9988). The MTT assay revealed anti-tumor activity against MCF-7 BC cells, with IC50 of 21 µg/mL for NG-MOF-5@LNs and demonstrating a total apoptosis effect of 68.2% on MCF-7 cells. CONCLUSION NG-MOF-5@LNs is anticipated to show as an effective antimicrobial and novel long-term-release antitumor agent and might be more suitable for MCF-7 cell therapy.
Collapse
Affiliation(s)
- Lina M Alneghery
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Mohammed Al-Zharani
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Fahd A Nasr
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Zienab E Eldin
- Center for Material Science, Zewail City of Science and Technology, 6th of October, Egypt
- Department of Material Science and Nanotechnology, Faculty of Postgraduate Studies for Advanced Sciences (PSAS), Beni-Suef University, Beni-Suef, Egypt
| | - Tayel A Al Hujran
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Hesham M Tawfeek
- Department of Industrial Pharmacy, Faculty of Pharmacy, Assiut University, Assiut, Egypt
| | - Mohamed H Fayed
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Albatin, Hafr Albatin, Saudi Arabia
- Department of Pharmaceutics, Faculty of Pharmacy, Fayoum University, Fayoum, Egypt
| | - Shehab Elbeltagi
- Department of Physics, Faculty of Science, New Valley University, Kharga, Egypt
| |
Collapse
|
3
|
Alshaikh F, Al-Samydai A, Issa R, Alshaer W, Alqaraleh M, Al-Halaseh LK, Alsanabrah A, Ghanim BY, Al Azzam KM, Qinna NA. Encapsulation of gingerol into nanoliposomes: Evaluation of in vitro anti-inflammatory and anti-cancer activity. Biomed Chromatogr 2024; 38:e5899. [PMID: 38797863 DOI: 10.1002/bmc.5899] [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: 03/20/2024] [Revised: 04/16/2024] [Accepted: 05/06/2024] [Indexed: 05/29/2024]
Abstract
Nanoliposomes (NLs) are ideal carriers for delivering complex molecules and phytochemical products, but ginger by-products, despite their therapeutic benefits, have poor bioavailability due to their low water solubility and stability. Crude ginger extracts (CGEs) and 6-gingerol were individually encapsulated within NLs for in vitro activity assessment. In vitro evaluation of anti-proliferative and anti-inflammatory properties of encapsulated 6-gingerol and CGE was performed on healthy human periodontal ligament (PDL) fibroblasts and MDA-MB-231 breast cancer cells. Encapsulation efficiency and loading capacity of 6-gingerol reached 25.23% and 2.5%, respectively. NLs were found stable for up to 30 days at 4°C with a gradual load loss of up to 20%. In vitro cytotoxic effect of encapsulated 6-gingerol exceeded 70% in the MDA-MB-231 cell line, in a comparable manner with non-encapsulated 6-gingerol and CGE. The effect of CGE with an IC50 of 3.11 ± 0.39, 7.14 ± 0.80, and 0.82 ± 0.55 μM and encapsulated 6-gingerol on inhibiting IL-8 was evident, indicating its potential anti-inflammatory activity. Encapsulating 6-gingerol within NLs enhanced its stability and facilitated its biological activity. All compounds, including vitamin C, were equivalent at concentrations below 2 mg/mL, with a slight difference in antioxidant activity. The concentrations capable of inhibiting 50% of 2,2-diphenyl-1-picrylhydrazyl (DPPH) substrate were comparable.
Collapse
Affiliation(s)
- Fatima Alshaikh
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Ali Al-Samydai
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Reem Issa
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Moath Alqaraleh
- Department of Medical Laboratory Sciences, Faculty of Science, Al-Balqa Applied University, Al-Salt, Jordan
| | - Lidia K Al-Halaseh
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Mutah University, Al-Karak, Jordan
| | - Alaa Alsanabrah
- Pharmacological and Diagnostic Research Centre, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Bayan Y Ghanim
- University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| | - Khaldun M Al Azzam
- Department of Chemistry, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Nidal A Qinna
- University of Petra Pharmaceutical Center (UPPC), Faculty of Pharmacy and Medical Sciences, University of Petra, Amman, Jordan
| |
Collapse
|
4
|
Cao X, Li Q, Li X, Liu Q, Liu K, Deng T, Weng X, Yu Q, Deng W, Yu J, Wang Q, Xiao G, Xu X. Enhancing Anticancer Efficacy of Formononetin Microspheres via Microfluidic Fabrication. AAPS PharmSciTech 2023; 24:241. [PMID: 38017231 DOI: 10.1208/s12249-023-02691-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/30/2023] [Indexed: 11/30/2023] Open
Abstract
Formononetin is a flavonoid compound with anti-tumor and anti-inflammatory properties. However, its low solubility limits its clinical use. We employed microfluidic technology to prepare formononetin-loaded PLGA-PEGDA microspheres (Degradable polymer PLGA, Crosslinking agent PEGDA), which can encapsulate and release drugs in a controlled manner. We optimized and characterized the microspheres, and evaluated their antitumor effects. The microspheres had uniform size, high drug loading efficiency, high encapsulation efficiency, and stable release for 35 days. They also inhibited the proliferation, migration, and apoptosis. The antitumor mechanism involved the induction of reactive oxygen species and modulation of Bcl-2 family proteins. These findings suggested that formononetin-loaded PLGA-PEGDA microspheres, created using microfluidic technology, could be a novel drug delivery system that can overcome the limitations of formononetin and enhance its antitumor activity.
Collapse
Affiliation(s)
- Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Qingwen Li
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Xiaoli Li
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Qi Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Kai Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Tianwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Xuedi Weng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qintong Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Wenwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China.
| | - Gao Xiao
- College of Environment and Safety Engineering, Fuzhou University, Fuzhou, 350108, Fujian, People's Republic of China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.
- Medicinal function development of new food resources, Jiangsu Provincial Research Center, Jiangsu, People's Republic of China.
| |
Collapse
|
5
|
Shi F, Yin W, Adu-Frimpong M, Li X, Xia X, Sun W, Ji H, Toreniyazov E, Qilong W, Cao X, Yu J, Xu X. In-vitro and in-vivo evaluation and anti-colitis activity of esculetin-loaded nanostructured lipid carrier decorated with DSPE-MPEG2000. J Microencapsul 2023; 40:442-455. [PMID: 37191893 DOI: 10.1080/02652048.2023.2215345] [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: 09/27/2022] [Accepted: 05/12/2023] [Indexed: 05/17/2023]
Abstract
OBJECTIVE Encapsulation of esculetin into DSPE-MPEG2000 carrier was performed to improve its water solubility and oral bioavailability, as well as enhance its anti-inflammatory effect on a mouse model of ulcerative colitis that was induced with dextran sulphate sodium (DSS). METHODS We determined the in-vitro and in-vivo high-performance liquid chromatographic (HPLC) analysis method of esculetin; Esculetin-loaded nanostructure lipid carrier (Esc-NLC) was prepared using a thin-film dispersion method, wherein a particle size analyser was used to measure the particle size (PS) and zeta potential (ZP) of the Esc-NLC, while a transmission electron microscope (TEM) was employed to observe its morphology. Also, HPLC was used to measure its drug loading (DL), encapsulation efficiency (EE) and the in-vitro release of the preparation, as well as investigate the pharmacokinetic parameters. In addition, its anti-colitis effect was evaluated via histopathological examination of HE-stained sections and detection of the concentrations of tumour necrosis factor-alpha (TNF-α), interleukin (IL)-1 beta (β), and IL-6 in serum with ELISA kits. RESULTS The PS of Esc-NLC was 102.29 ± 0.63 nm with relative standard deviation (RSD) of 1.08% (with poly-dispersity index-PDI of 0.197 ± 0.023), while the ZP was -15.67 ± 1.39 mV with RSD of 1.24%. Solubility of esculetin was improved coupled with prolonged release time. Its pharmacokinetic parameters were compared with that of free esculetin, wherein the maximum concentration of the drug in plasma was increased by 5.5 times. Of note, bioavailability of the drug was increased by 1.7 times, while the half-life was prolonged by 2.4 times. In the anti-colitis efficacy experiment, the mice in Esc and Esc-NLC groups exhibited significantly reduced levels of TNF-α, IL-1β, and IL-6 in their sera comparable to the DSS group. Colon histopathological examination revealed that mice with ulcerative colitis in both Esc and Esc-NLC groups displayed improved inflammation, amid the Esc-NLC groups having the best prophylactic treatment effect. CONCLUSION Esc-NLC could ameliorate DSS-induced ulcerative colitis by improving bioavailability, prolonging drug release time and regulating cytokine release. This observation confirmed the potential of Esc-NLC to reduce inflammation in ulcerative colitis, albeit the need for follow-up research to verify the application of this strategy to clinical treatment of ulcerative colitis.
Collapse
Affiliation(s)
- Feng Shi
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Wenxiong Yin
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School Chemical and Biochemical Sciences C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, GH, 0215-5321, UK
| | - Xiaoxiao Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Xiaoli Xia
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Weigang Sun
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
| | - Hao Ji
- Jiangsu Tian Sheng Pharmaceutical Co., Ltd, Zhenjiang, CN, P.R. China
| | - Elmurat Toreniyazov
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
- Tashkent State Agricultural University (Nukus branch), Nukus, UZ, P.R. China
| | - Wang Qilong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, P.R. China
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, P.R. China
| |
Collapse
|
6
|
Zagórska J, Kukula-Koch W, Czop M, Iłowiecka K, Koch W. Impact of Thermal Processing on the Composition of Curcuma longa Rhizome. Foods 2023; 12:3086. [PMID: 37628084 PMCID: PMC10453412 DOI: 10.3390/foods12163086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/03/2023] [Accepted: 08/15/2023] [Indexed: 08/27/2023] Open
Abstract
Curcuma longa L. (Zingiberaceae), known as turmeric, is a perennial tuberous plant from the genus Curcuma, which includes about 100 plant species. The chemical composition of the turmeric rhizome is very diverse. Diarylheptanoid derivatives, also known as curcuminoids (of which curcumin, demethoxycurcumin and bisdemethoxycurcumin are the most important representatives), are the major active constituents of the plant rhizome. Many extracts used in the food and pharmaceutical industries are produced from thermally processed rhizome, when there are significant changes in the composition of the main compounds. Therefore, the aim of the study was to compare how the type of thermal treatment affects the content of curcuminoids and the antioxidant properties of the rhizome. The plant material was subjected to three different methods of thermal processing-microwave heating, boiling and frying in different time intervals. The chemical composition and antioxidant activity of the processed rhizome was evaluated using LC-MS (liquid chromatography-mass spectrometry), HPLC (high-performance liquid chromatography) and spectrophotometric methods (a DPPH test and TPC assay). Obtained results revealed that curcumin was the major curcuminoid present in all samples (113.92 mg/g of the fresh rhizome). Significant correlation between the type and time of the thermal processing and the composition of turmeric samples was revealed. A traditional boiling process lasting for 10 min was the most beneficial process in terms of the curcuminoid content (204 mg/g of curcumin) and antioxidant activity of the samples.
Collapse
Affiliation(s)
- Justyna Zagórska
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland; (J.Z.); (K.I.)
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medical Plants Garden, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland;
| | - Marcin Czop
- Department of Clinical Genetics, Medical University of Lublin, 11 Radziwiłłowska Str., 20-080 Lublin, Poland;
| | - Katarzyna Iłowiecka
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland; (J.Z.); (K.I.)
| | - Wojciech Koch
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland; (J.Z.); (K.I.)
| |
Collapse
|
7
|
Lee YR, Jeong HM, Kim JS, Kim EA, Lee EH, Shim JH. Enzymatic formation of cyclic maltooligosaccharides for the application of quercetin inclusion complex. Carbohydr Polym 2023; 310:120722. [PMID: 36925261 DOI: 10.1016/j.carbpol.2023.120722] [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: 11/16/2022] [Revised: 01/20/2023] [Accepted: 02/15/2023] [Indexed: 02/22/2023]
Abstract
To improve the applicability of quercetin (QCT), we produced a QCT and cycloamylose (CA-QCT) inclusion complex based on the cyclization activity of cyclodextrin glucanotransferase (CGTase; EC 2.4.1.19). The encapsulated QCT was purified using recycling preparative high-performance liquid chromatography, and its formation was analyzed using matrix-assisted laser desorption/ionization-time of flight mass spectrometry. The water solubility of CA-QCT was 55,000-fold higher than that of QCT. CA-QCT had 97 % stability for one week at pH 8 in a 4 °C water bath. According to a 2,2-diphenyl-1-picrylhydrazyl radical scavenging activity assay, CA-QCT activity in aqueous solution was 24 times higher than that of an equal amount of QCT in aqueous solution. In an anti-inflammatory assay using lipopolysaccharide-induced RAW264.7 macrophages, CA-QCT in aqueous solution decreased nitric oxide production in a similar manner to QCT in dimethyl sulfoxide (DMSO). Additionally, even under aqueous conditions, CA-QCT more effectively inhibited the production of inflammatory mediators, such as interleukin-1β, interleukin-6, and cyclooxygenase, compared with QCT dissolved in DMSO.
Collapse
Affiliation(s)
- Ye-Rim Lee
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea; The Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea
| | - Hyun-Mo Jeong
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea; The Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea
| | - Ji-Soo Kim
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea; The Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea
| | - Eun-A Kim
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea; The Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea
| | - Eun-Hyeong Lee
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea; The Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea
| | - Jae-Hoon Shim
- Department of Food Science and Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea; The Korean Institute of Nutrition, Hallym University, Hallymdaehak-gil 1, Chuncheon, Gwangwon-do 24252, Republic of Korea.
| |
Collapse
|
8
|
Cao X, Liu Q, Shi W, Liu K, Deng T, Weng X, Pan S, Yu Q, Deng W, Yu J, Wang Q, Xiao G, Xu X. Microfluidic fabricated bisdemethoxycurcumin thermosensitive liposome with enhanced antitumor effect. Int J Pharm 2023; 641:123039. [PMID: 37225026 DOI: 10.1016/j.ijpharm.2023.123039] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 04/17/2023] [Accepted: 05/05/2023] [Indexed: 05/26/2023]
Abstract
Bisdemethoxycurcumin (BDMC) is the main active ingredient that is isolated from Zingiberaceae plants, wherein it has excellent anti-tumor effects. However, insolubility in water limits its clinical application. Herein, we reported a microfluidic chip device that can load BDMC into the lipid bilayer to form BDMC thermosensitive liposome (BDMC TSL). The natural active ingredient glycyrrhizin was selected as the surfactant to improve solubility of BDMC. Particles of BDMC TSL had small size, homogenous size distribution, and enhanced cultimulative release in vitro. The anti-tumor effect of BDMC TSL on human hepatocellular carcinomas was investigated via 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide method, live/dead staining, and flowcytometry. These results showed that the formulated liposome had a strong cancer cell inhibitory, and presented a dose-dependent inhibitory effect on migration. Further mechanistic studies showed that BDMC TSL combined with mild local hyperthermia could significantly upregulate B cell lymphoma 2 associated X protein levels and decrease B cell lymphoma 2 protein levels, thereby inducing cell apoptosis. The BDMC TSL that was fabricated via microfluidic device were decomposed under mild local hyperthermia, which could beneficially enhance the anti-tumor effect of raw insoluble materials and promote translation of liposome.
Collapse
Affiliation(s)
- Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Qi Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Wenwan Shi
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Kai Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Tianwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Xuedi Weng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Siting Pan
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qingtong Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Wenwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China.
| | - Gao Xiao
- College of Environment and Safety Engineering, Fuzhou University, Fuzhou 350108, Fujian, P. R. China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China; Medicinal function development of new food resources, Jiangsu Provincial Research center, Jiangsu, People's Republic of China.
| |
Collapse
|
9
|
Tamizhmathy M, Gupta U, Shettiwar A, Kumar GS, Daravath S, Aalhate M, Mahajan S, Maji I, Sriram A, Modak C, Rajalakshmi A, Dikundwar AG, Doijad N, Guru SK, Singh PK. Formulation of inclusion complex of Abiraterone acetate with 2-Hydroxypropyl-Beta-Cyclodextrin: physiochemical characterization, molecular docking and bioavailability evaluation. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104321] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2023]
|
10
|
Cao X, Liu Q, Adu-Frimpong M, Shi W, Liu K, Deng T, Yuan H, Weng X, Gao Y, Yu Q, Deng W, Yu J, Wang Q, Xiao G, Xu X. Microfluidic Generation of Near-Infrared Photothermal Vitexin/ICG Liposome with Amplified Photodynamic Therapy. AAPS PharmSciTech 2023; 24:82. [PMID: 36949351 DOI: 10.1208/s12249-023-02539-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2022] [Accepted: 02/12/2023] [Indexed: 03/24/2023] Open
Abstract
Glioma, in which a malignant tumor cell occurs in neural mesenchymal cells, has a rapid progression and poor prognosis, which is still far from desirable in clinical treatments. We developed a lab-on-a-chip (LOC) device for the rapid and efficient preparation of vitexin/indocyanine green (ICG) liposomes. Vitexin could be released from liposome to kill cancer cell, which can potentially improve the glioma therapeutic effect and reduce the treatment time through synergistic photodynamic/photothermal therapies (PDT/PTT). The vitexin/ICG liposome was fabricated via LOC and its physicochemical property and release in vitro were evaluated. 3-(4,5-Dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) method and live/dead staining were used to examine the enhanced antitumor effect of vitexin/ICG liposome in cooperation with PDT/PTT, while the related mechanism was explored by flow cytometry and western blot. The results were as follows: (1) The prepared vitexin/ICG liposome was smaller in size, homogenous in particle size distribution with significant low polydispersity index (PDI), and enhanced cumulative release in vitro. (2) We found that the formulated liposome presented strong cancer cell inhibition and suppression of its migration in a dose-dependent manner. (3) Further mechanistic studies showed that liposome combined with near-infrared irradiation could significantly upregulate levels of B cell lymphoma 2-associated X (Bax) protein and decrease B cell lymphoma 2 (Bcl-2) at protein levels. The vitexin/ICG liposomes prepared based on a simple LOC platform can effectively enhance the solubility of insoluble drugs, and the combined effect of PTT/PDT can effectively increase their antitumor effect, which provides a simple and valid method for the clinical translation of liposomes.
Collapse
Affiliation(s)
- Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Qi Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Michael Adu-Frimpong
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, UK-0215-5321, Ghana
| | - Wenwan Shi
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Kai Liu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Tianwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Hui Yuan
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Xuedi Weng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Yihong Gao
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Qingtong Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Wenwen Deng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China.
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China.
| | - Gao Xiao
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China.
- College of Environment and Safety Engineering, Fuzhou University, Fuzhou, 350108, Fujian, People's Republic of China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.
- Medicinal Function Development of New Food Resources, Jiangsu Provincial Research Center, Zhenjiang, Jiangsu, People's Republic of China.
- School of Life Sciences, Northwestern Polytechnical University, Xi'an, Shanxi, 710072, People's Republic of China.
| |
Collapse
|
11
|
Zagórska J, Czernicka-Boś L, Kukula-Koch W, Iłowiecka K, Koch W. Impact of Thermal Processing on the Selected Biological Activities of Ginger Rhizome-A Review. Molecules 2023; 28:412. [PMID: 36615602 PMCID: PMC9824518 DOI: 10.3390/molecules28010412] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/08/2022] [Accepted: 12/21/2022] [Indexed: 01/06/2023] Open
Abstract
Ginger (Zingiber officinale Rosc.) is a spice, medicinal and cosmetic plant that has been known for centuries. It can be used in dried, fresh, marinated or candied form, and is also an essential ingredient in well-known curry blends. Ginger rhizomes are often freeze-dried as the first step in the preparation of the raw material. Many studies have proved that the composition and biological activity of ginger changes due to thermal processing. Therefore, the aim of the review was to summarize the scientific results on the impact of traditional and unconventional methods of the heat treatment of ginger rhizomes and their influence on the antioxidant and other selected biological activities of the plant. The review of the available scientific data is inconclusive, and it is hard to state unequivocally whether the thermal treatment of the raw material increases or decreases biological activity. Based on the presented literature review, it can be concluded that traditional cooking and microwave processing in general decrease the antioxidant activity of the ginger rhizome, whereas frying, autoclaving, blanching or traditional drying in the sun mostly lead to a significant increase in ginger activity. Interesting data were presented in the works describing the freeze-drying process during which the antioxidant potential of ginger increased.
Collapse
Affiliation(s)
- Justyna Zagórska
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland
| | - Lidia Czernicka-Boś
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland
| | - Wirginia Kukula-Koch
- Department of Pharmacognosy with Medical Plants Garden, Medical University of Lublin, 1 Chodzki Str., 20-093 Lublin, Poland
| | - Katarzyna Iłowiecka
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland
| | - Wojciech Koch
- Department of Food and Nutrition, Medical University of Lublin, 4a Chodzki Str., 20-093 Lublin, Poland
| |
Collapse
|
12
|
Preparation, Physical Characterization, Pharmacokinetics and Anti-Hyperglycemic Activity of Esculetin-Loaded Mixed Micelles. J Pharm Sci 2023; 112:148-157. [PMID: 35780820 DOI: 10.1016/j.xphs.2022.06.022] [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: 03/13/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 11/22/2022]
Abstract
Despite its low water solubility, esculetin (EC) have been described to demonstrate various health benefits. Thus, we sought to develop esculetin-loaded mixed micelles (EC-M) delivery system to purposively improve biological availability and anti-hyperglycemia activity of EC. Thin-film hydration method was employed to fabricate EC-M, amid characterization with transmission electron microscopic analysis (TEM), coupled with physical properties such as particle size (PS), poly-dispersity index (PDI), zeta-potential (ZP) and stability testing. We analyzed in-vitro release and studied EC-M pharmacokinetics in rats. The hyperglycemic mice model was established with streptozotocin (STZ) to evaluate anti-hyperglycemic activity of EC-M. The PS, PDI and ZP of EC-M were 47.97 ± 0.41 nm, 0.189 ± 0.005 and -25.55 ± 0.28 mV, respectively. The release rate of EC-M increased comparable to free EC in the three media. The oral biological availability and half-life of EC-M increased respectively by 3.06 and 1.45 folds compared to free EC. Besides, we observed 46.21% decrease in blood glucose of mice in EC-M group comparable to the model control, wherein, the anti-hyperglycemic effect of EC-M was better compared to free EC. Conclusively, EC-M may ideally serve as a novel approach to enhance biological availability and increased anti-hyperglycemic activity of EC.
Collapse
|
13
|
Ahmad N, Khalid MS, Khan MF, Ullah Z. Beneficial effects of topical 6-gingerol loaded nanoemulsion gel for wound and inflammation management with their comparative dermatokinetic. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
14
|
Shen X, Rong W, Adu-Frimpong M, He Q, Li X, Shi F, Ji H, Toreniyazov E, Xia X, Zhang J, Wang Q, Yu J, Xu X. Preparation, in vitro and in vivo evaluation of pinocembrin-loaded TPGS modified liposomes with enhanced bioavailability and antihyperglycemic activity. Drug Dev Ind Pharm 2022; 48:623-634. [PMID: 36420780 DOI: 10.1080/03639045.2022.2151616] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
PURPOSE To prepare polyethylene glycol succinate-vitamin E modified pinocembrin (PCB)-loaded liposomes (PCBT-liposomes) and evaluate PCBT-liposomal pharmacokinetics and antihyperglycemic activity. SIGNIFICANCE The novel PCBT-liposomes demonstrated a promising application prospect as a nano drug carrier for future research. METHODS Thin film dispersion was used to prepare PCBT-liposomes. We measured a series of characterization, followed by in vitro cumulative release, in vivo pharmacokinetic study, and antihyperglycemic activity evaluation. RESULTS PCBT-liposomes displayed spherical and bilayered nanoparticles with mean particle size (roughly 92 nm), negative zeta potential (about -26.650 mV), high drug encapsulation efficiency (87.32 ± 1.34%) and good storage (at 4 or 25 °C) stability during 48 h after hydration. The cumulative release rate of PCBT-liposomes was markedly higher than free PCB in four different pH media. In vivo investigation showed that PCBT-liposomes could obviously improve oral bioavailability of PCB by 1.96 times, whereas the Cmax, MRT0-t, and T1/2 of PCBT-liposomes were roughly 1.700 ± 0.139 µg·mL-1, 12.695 ± 1.647 h, and 14.244 h, respectively. In terms of biochemical analysis, aspartate amino-transferase (AST), alanine amino-transferase (ALT), interleukin-1 (IL-1), and tumor necrosis factor-α (TNF-α) concentrations in serum of diabetic mice were respectively decreased 28.28%, 17.23%, 17.77%, and 8.08% after PCBT-liposomal treatment. CONCLUSION These results show PCBT-liposomal preparation as an excellent nano-carrier which has the potential to improve water solubility, bioavailability, and antihyperglycemic activity of PCB, amid broadening the application of PCB in the clinical settings.
Collapse
Affiliation(s)
- Xinyi Shen
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Wanjing Rong
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, Ghana
| | - Qing He
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Xiaoxiao Li
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Feng Shi
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Hao Ji
- Jiangsu Tian Sheng Pharmaceutical Co., Ltd., Zhenjiang, China
| | | | - Xiaoli Xia
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Jian Zhang
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Qilong Wang
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Jiangnan Yu
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| | - Ximing Xu
- Department of Pharmaceutics, Pharmacy School, Center for Nano Drug-Gene Delivery and Tissue Engineering, Center for Medicinal Function Development of New Food Resources, Jiangsu Provincial Research, Jiangsu University, Zhenjiang, China
| |
Collapse
|
15
|
Gao Y, Lu Y, Zhang N, Udenigwe CC, Zhang Y, Fu Y. Preparation, pungency and bioactivity of gingerols from ginger ( Zingiber officinale Roscoe): a review. Crit Rev Food Sci Nutr 2022; 64:2708-2733. [PMID: 36135317 DOI: 10.1080/10408398.2022.2124951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
Ginger has been widely used for different purposes, such as condiment, functional food, drugs, and cosmetics. Gingerols, the main pungent component in ginger, possess a variety of bioactivities. To fully understand the significance of gingerols in the food and pharmaceutical industry, this paper first recaps the composition and physiochemical properties of gingerols, and the major extraction and synthesis methods. Furthermore, the pungency and bioactivity of gingerols are reviewed. In addition, the food application of gingerols and future perspectives are discussed. Gingerols, characterized by a 3-methoxy-4-hydroxyphenyl moiety, are divided into gingerols, shogaols, paradols, zingerone, gingerdiones and gingerdiols. At present, gingerols are extracted by conventional, innovative, and integrated extraction methods, and synthesized by chemical, biological and in vitro cell synthesis methods. Gingerols can activate transient receptor potential vanilloid type 1 (TRPV1) and induce signal transduction, thereby exhibiting its pungent properties and bioactivity. By targeted mediation of various cell signaling pathways, gingerols display potential anticancer, antibacterial, blood glucose regulatory, hepato- and renal-protective, gastrointestinal regulatory, nerve regulatory, and cardiovascular protective effects. This review contributes to the application of gingerols as functional ingredients in the food and pharmaceutical industry.
Collapse
Affiliation(s)
- Yuge Gao
- College of Food Science, Southwest University, Chongqing, China
- Westa College, Southwest University, Chongqing, China
| | - Yujia Lu
- Department of Epidemiology, Harvard University T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Na Zhang
- Key Laboratory of Food Science and Engineering of Heilongjiang Province, College of Food Engineering, Harbin University of Commerce, Harbin, China
| | - Chibuike C Udenigwe
- School of Nutrition Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, Ontario, Canada
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, China
| | - Yu Fu
- College of Food Science, Southwest University, Chongqing, China
- Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing, China
| |
Collapse
|
16
|
Yücel Ç, Karatoprak GŞ, Açıkara ÖB, Akkol EK, Barak TH, Sobarzo-Sánchez E, Aschner M, Shirooie S. Immunomodulatory and anti-inflammatory therapeutic potential of gingerols and their nanoformulations. Front Pharmacol 2022; 13:902551. [PMID: 36133811 PMCID: PMC9483099 DOI: 10.3389/fphar.2022.902551] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2022] [Accepted: 07/19/2022] [Indexed: 11/13/2022] Open
Abstract
Ginger (Zingiber officinale Roscoe), a member of the Zingiberaceae family, is one of the most popular spices worldwide, known since ancient times, and used both as a spice and a medicinal plant. The phenolic compounds found in ginger are predominantly gingerols, shogaols, and paradols. Gingerols are the major phenolic compounds found in fresh ginger and contain mainly 6-gingerol as well as 4-, 5-, 8-, 10-, and 12-gingerols. Gingerols possess a wide array of bioactivities, such as antioxidant and anticancer, among others. Regarding the different array of biological activities and published data on the mechanisms underlying its action, the complex interaction between three key events, including inflammation, oxidative stress, and immunity, appears to contribute to a plethora of pharmacological activities of this compound. Among these, the immunomodulatory properties of these compounds, which attract attention due to their effects on the immune system, have been the focus of many studies. Gingerols can alleviate inflammation given their ability to inhibit the activation of protein kinase B (Akt) and nuclear factor kappa B (NF-κB) signaling pathways, causing a decrease in proinflammatory and an increase in anti-inflammatory cytokines. However, given their low bioavailability, it is necessary to develop new and more effective strategies for treatment with gingerols. In order to overcome this problem, recent studies have addressed new drug delivery systems containing gingerols. In this review, the immunomodulatory activities of gingerol and its underlying mechanisms of action combined with the contributions of developed nanodrug delivery systems to this activity will be examined.
Collapse
Affiliation(s)
- Çiğdem Yücel
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Erciyes University, Kayseri, Turkey
| | | | | | - Esra Küpeli Akkol
- Department of Pharmacognosy, Faculty of Pharmacy, Gazi University, Ankara, Turkey
- *Correspondence: Esra Küpeli Akkol, ; Eduardo Sobarzo-Sánchez,
| | - Timur Hakan Barak
- Department of Pharmacognosy, Faculty of Pharmacy, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey
| | - Eduardo Sobarzo-Sánchez
- Department of Organic Chemistry, Faculty of Pharmacy, University of Santiago de Compostela, Santiago de Compostela, Spain
- Instituto de Investigación y Postgrado, Facultad de Ciencias de la Salud, Universidad Central de Chile, Santiago, Chile
- *Correspondence: Esra Küpeli Akkol, ; Eduardo Sobarzo-Sánchez,
| | - Michael Aschner
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Park Avenue Bronx, NY, United States
| | - Samira Shirooie
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| |
Collapse
|
17
|
Upaganlawar A, Polshettiwar S, Raut S, Tagalpallewar A, Pande V. Effective Cancer Management: Inimitable Role of Phytochemical Based Nano- Formulations. Curr Drug Metab 2022; 23:869-881. [PMID: 36065928 DOI: 10.2174/1389200223666220905162245] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 04/07/2022] [Accepted: 04/13/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Global cancer statistics defines the severity of disease even after significant research worldwide. PROBLEM Failure of the currently available treatment approaches, including surgery, radiation therapy and traditional chemotherapy. AIM The aim of this review is to discuss the role of phytochemical based nano-formulations for treatment of cancer. DISCUSSION In the past few decades, phytochemicals have gained popularity for acting as a potential anticancer treatment with low systemic toxicity, especially in terms of cell cycle control and cancer cell killing. Natural resources, with their immense structural variety, serve as a vital source of fresh, therapeutically useful new chemical entities for the treatment of cancer. Vinca alkaloids (VCR), vinblastine, vindesine, vinorelbine, taxanes (PTX), podophyllotoxin and its derivatives (etoposide (ETP), teniposide, camptothecin (CPT) and its derivatives (topotecan, irinotecan), anthracyclines (doxorubicin, daunorubicin, epirubicin, idarubicin, as natural products or their derivatives account for half of all anticancer drugs approved worldwide, and they have been developed utilising the knowledge learned from the natural small molecules or macromolecules. Trabectedin, an epothilone derivative, ixabepilone, and temsirolimus, three new anticancer medications launched in 2007, were derived from microbial origins. Current therapy regimens require selective drug targeting to enhance efficacy against cancer cells while normal cells remain unharmed. Modified medications and systems for drug delivery based on nanotechnology are in the process of being explored and launched in the industry for enhanced therapy and management of cancer, along with promising outcomes. Many obstacles related to cancer cell drug delivery can be overcome by using nano-particulate drug carriers, including enhancing the stability and solubility of the drug, prolonging half-lives of the drug in the blood, decreasing side effects to undesired organs, and increasing medication concentration at the desired site. The scientific initiatives and studies concerning the use of nanotechnology for some selective compounds derived from plants are discussed in this review article. CONCLUSION The present review highlights the phytochemical-based nanoformulations and their strategies in the development of novel systems of drug delivery such as nano-liposomes, functionalized nanoparticles (NPs), and polymer nano-conjugates, SNEDDS (Self nano emulsifying drug delivery system) as this review paper depicts, as well as their rewards over conventional systems of drug delivery, as evidenced by improved biological activity depicted in their in vitro and in vivo anticancer assays.
Collapse
Affiliation(s)
- Aman Upaganlawar
- SNJBs SSDJ College of Pharmacy, Neminagar, Chandwad, Maharashtra, India
| | - Satish Polshettiwar
- School of Pharmacy Dr.Vishwanath Karad MIT World Peace University, Survey No. 124, Kothrud, Pune, Maharashtra 411038, India
| | - Sushil Raut
- Dr. D. Y. Patil Institute of Pharmaceutical Sciences and Research, Pimpri, Pune-India
| | - Amol Tagalpallewar
- School of Pharmacy Dr.Vishwanath Karad MIT World Peace University, Survey No. 124, Kothrud, Pune, Maharashtra 411038, India
| | - Vishal Pande
- N. N. Sattha College of Pharmacy, Ahmednagar, Maharashtra, India
| |
Collapse
|
18
|
Verma M, Fatima S, Ansari IA. Phytofabricated Nanoparticle Formulation for Cancer Treatment: A Comprehensive Review. Curr Drug Metab 2022; 23:818-826. [PMID: 35490313 DOI: 10.2174/1389200223666220427101427] [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: 11/28/2021] [Revised: 03/04/2022] [Accepted: 03/14/2022] [Indexed: 01/05/2023]
Abstract
In recent times, nanotechnology has made significant advances in the field of cancer. The majority of chemotherapeutic drugs do not selectively target cancer cells, and they might cause side effects and damage to healthy cells, resulting in a variety of adverse effects. Having a thorough understanding of nanoparticles may improve drug targeting and administration. The nano-engineering of pharmacological and natural compounds can improve the diagnosis and treatment. Polymeric micelles, liposomes, and dendrimers are examples of innovative cancer therapeutic nano-formulations. It has been demonstrated that quantum dots, nano-suspensions, and gold nanoparticles can improve drug delivery. Nanomedicines may be delivered more effectively, focusing on cancerous cells instead of healthy tissues, which minimizes undesirable side effects and drug resistance to chemotherapeutic agents. However, limited water solubility, low stability, poor absorption, and quick metabolism limit their therapeutic effectiveness. Nanotechnology has generated unique formulations to optimise the potential use of phytochemicals in anticancer therapy. Nanocomposites can improve phytochemical solubility and bioavailability, extend their half-life in circulation, and even transport phytochemicals to specific locations. The progress in using phytochemical-based nanoparticles in cancer treatment is summarized in this paper.
Collapse
Affiliation(s)
- Mahima Verma
- Department of Biosciences, Integral University, IIRC1, Lucknow, India
| | - Shireen Fatima
- Department of Biosciences, Integral University, IIRC1, Lucknow, India
| | | |
Collapse
|
19
|
Promdam N, Khuituan P, Panichayupakaranant P. Effects of standardized [6]-gingerol extracts and [6]-gingerol on isolated ileum and lower esophageal sphincter contractions in mice. Food Chem 2022; 378:132077. [PMID: 35042108 DOI: 10.1016/j.foodchem.2022.132077] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 02/08/2023]
Abstract
Standardized [6]-gingerol extracts were prepared by microwave-assisted extraction using 20% v/v glycerin in ethanol and 20% v/v eutectic mixture of sucrose and citric acid in ethanol as alternative green solvents. The extracts obtained from 20% v/v glycerin in ethanol (GEE) and 20% v/v eutectic mixture of sucrose and citric acid in ethanol (EMSCEE) were standardized by HPLC to contain 17.0 mg/g of [6]-gingerol. The effects of the extracts on mouse ileal contractions via M3 and 5-HT3 receptors as well as lower esophageal sphincter (LES) contraction were determined in vitro relative to the marker compound, [6]-gingerol. [6]-Gingerol, GEE and EMSCEE demonstrated significant and concentration-dependent inhibitory effects on ileal contraction in mice via M3 and 5-HT3 receptors in a noncompetitive manner. In addition, [6]-gingerol and EMSCEE tend to increase the LES tone. These results indicated the potential of GEE and EMSCEE to attenuate nausea and vomiting and might be used as nutraceuticals.
Collapse
Affiliation(s)
- Nantaporn Promdam
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Pissared Khuituan
- Division of Health and Applied Sciences, Faculty of Science, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand; Gut Biology and Microbiota Research Unit, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand
| | - Pharkphoom Panichayupakaranant
- Department of Pharmacognosy and Pharmaceutical Botany, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand; Phytomedicine and Pharmaceutical Biotechnology Excellence Center, Faculty of Pharmaceutical Sciences, Prince of Songkla University, Hat-Yai, Songkhla 90112, Thailand.
| |
Collapse
|
20
|
Liu CM, An L, Wu Z, Ouyang AJ, Su M, Shao Z, Lin Y, Liu X, Jiang Y. 6‑Gingerol suppresses cell viability, migration and invasion via inhibiting EMT, and inducing autophagy and ferroptosis in LPS‑stimulated and LPS‑unstimulated prostate cancer cells. Oncol Lett 2022; 23:187. [PMID: 35527779 PMCID: PMC9073581 DOI: 10.3892/ol.2022.13307] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 03/31/2022] [Indexed: 11/21/2022] Open
Abstract
6-Gingerol is a bioactive compound isolated from Zingiber officinale. 6-Gingerol has been shown to have anticancer effects in numerous types of cancer cell. The mechanisms underlying the anticancer effect of 6-Gingerol in prostate cancer requires investigation. In the present study, the effect on cell viability of 6-Gingerol on LNCaP, PC3 and DU145 prostate cancer cells were determined using the MTT and colony formation assays. 6-Gingerol significantly inhibited cell migration, adhesion and invasion in LPS-stimulated and LPS-unstimulated prostate cancer cells. Furthermore, these changes were accompanied by alterations in the protein expression levels of epithelial-mesenchymal transition biomarkers, including E-cadherin, N-cadherin, Vimentin and zonula occludens-1. 6-Gingerol also induced autophagy by significantly increasing LC3B-II and Beclin-1 protein expression levels in prostate cancer cells. Combining 6-Gingerol with LY294002, an autophagy inhibitor, significantly increased cell survival in DU145 cells. Furthermore, 6-Gingerol significantly decreased the protein expression levels of glutathione (GSH) peroxidase 4 and nuclear factor erythroid 2-related factor 2 in prostate cancer cells. Reactive oxygen species (ROS) levels were significantly increased but GSH levels were decreased following 6-Gingerol treatment in prostate cancer cells. Co-treatment with the ferroptosis inhibitor, ferrostatin-1, significantly increased cell viability and significantly decreased ROS levels in 6-Gingerol-treated cells. These results suggested that 6-Gingerol may have inhibited prostate cell cancer viability via the regulation of autophagy and ferroptosis. In addition, 6-Gingerol inhibited cell migration, adhesion and invasion via the regulation of EMT-related protein expression levels in LPS-stimulated and LPS-unstimulated prostate cancer cells. In conclusion, 6-Gingerol may induce protective autophagy, autophagic cell death and ferroptosis-mediated cell death in prostate cancer cells. These findings may provide a strategy for the treatment and prevention of prostate cancer.
Collapse
Affiliation(s)
- Chi-Ming Liu
- School of Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Lijie An
- School of Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Zhengping Wu
- School of Aesthetic Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Ai-Jun Ouyang
- Department of Pharmacy, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Mengqiao Su
- School of Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Zichen Shao
- School of Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Yi Lin
- School of Aesthetic Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Xiaoyu Liu
- School of Aesthetic Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| | - Yinjie Jiang
- School of Medicine, Yichun University, Yichun, Jiangxi 336000, P.R. China
| |
Collapse
|
21
|
Thangavelu P, Sundaram V, Gunasekaran K, Mujyambere B, Raju S, Kannan A, Arasu A, Krishna K, Ramamoorthi J, Ramasamy S, Velusamy T, Ramalingam S. Development of Optimized Novel Liposome Loaded with 6-gingerol and Assessment of its Therapeutic Activity Against NSCLC In vitro and In vivo Experimental Models. Chem Phys Lipids 2022; 245:105206. [PMID: 35483420 DOI: 10.1016/j.chemphyslip.2022.105206] [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: 12/28/2021] [Revised: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 11/03/2022]
Abstract
6-Gingerol (Gn) is an active compound derived from ginger which possesses various biological activities. The therapeutic applications of Gn are limited due to its hydrophobic nature. To ease its administration, one of the nano-emulsion methods, liposome was selected to encapsulate Gn. Response Surface Methodology (RSM) was used to optimize liposome ratio. 97.2% entrapment efficiency was achieved at the ratio of 1:20:2 (Drug: Lipid: Cholesterol). The optimized liposome attained size below 200 d nm, spherical shape, negative surface charge and showed sustain release upon physical characterization methods such as FESEM, DLS, Zeta potential, Drug release. The signature FTIR peaks of both free Gn and free liposome (FL) were also observed in Lipo-Gn peak. Lipo-Gn showed significant cytotoxic effect on A549 cells (IC50 160.5 ± 0.74µM/ml) as well as inhibits the cell migration. DAPI staining showed higher apoptotic nuclear morphological change in the cells treated with Lipo-Gn, and also Lipo-Gn increased the apoptotic percentage in A549 as 39.89 and 70.32 for 12 and 24h respectively which were significantly more than free Gn. Moreover, the formulation of Lipo-Gn showed significant cell cycle arrest at the G2/M phase compared with free Gn (28.9 and 34.9% in Free Gn vs. 42.7 and 50.1% in Lipo -Gn for 12 and 24hours respectively). Lipo-Gn have been assessed in NSCLC induced BALB/c mice and showed significantly improved pharmacological properties compared to those of free Gn. Thus, Lipo-Gn may be considered for its widening applications against lung cancer.
Collapse
Affiliation(s)
| | - Viswanathan Sundaram
- Bharathiar Cancer Theranostics Research Center - RUSA-2.0, Bharathiar university
| | - Kaavya Gunasekaran
- Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu, India
| | | | - Sowndarya Raju
- Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Arya Kannan
- Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Ashok Arasu
- Department of Biotechnology, Bharathiar University, Coimbatore, Tamil Nadu, India
| | - Kathirvelu Krishna
- DRDO-BU Center for Life Sciences, Bharathiar University Campus, Coimbatore, Tamil Nadu. India.cs
| | - Jayaraj Ramamoorthi
- Theme lead, Flinders NT, Flinders University, Northern Territory 0909, Australia
| | - Sivasamy Ramasamy
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore, Tamil nadu, India
| | | | - Suja Ramalingam
- Department of Biochemistry, Bharathiar University, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
22
|
Anticancer Efficacy of 6-Gingerol with Paclitaxel against Wild Type of Human Breast Adenocarcinoma. Molecules 2022; 27:molecules27092693. [PMID: 35566044 PMCID: PMC9104006 DOI: 10.3390/molecules27092693] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/19/2022] [Accepted: 04/20/2022] [Indexed: 12/20/2022] Open
Abstract
Breast cancer is one of the most common malignant neoplasms, and despite the dynamic development of anticancer therapies, 5-year survival in the metastatic stage is still less than 30%. 6-Gingerol (1-[4′-hydroxy-3′-methoxyphenyl]-5-hydroxy-3-decanone) is a substance contained in ginger, which exhibits anti-cancer properties. Paclitaxel is a cytostatic substance used to treat breast cancer, but its therapeutically effective dose has many adverse effects. The aim of the presented study was to assess the anticancer effect of 6-gingerol and the possibility of increasing the effectiveness of Paclitaxel in the death induction of wild type human breast cancer cells. MCF-7/WT cells were treated with drugs—6-gingerol and paclitaxel at selected concentrations. The mitochondrial activity assay, caspase 7 activity assay, ATP assay, microscopy studies, and RT-PCR assays were performed to evaluate the antitumor activity and mechanism of action of both compounds, alone and in combination. After 72 h of incubation, the mitochondrial activity showed that the combination of 5 nM Paclitaxel with 10 µM 6-Gingerol led to the same decrease in viability as the use of 20 nM Paclitaxel alone; 10 µM 6-Gingerol led to an enhancement of caspase 7 activity, with the highest activity observed after 24 h of incubation. A real-time PCR study showed that 6-Gingerol induces the simultaneous transcription of Bax with TP53 genes in large excess to BCL-2. In contrast, 5 nM Paclitaxel induces TP53 transcription in excess of BCL-2 and Bax. Our results suggest that 6-Gingerol may act as a cell death-inducing agent in cancer cells and, in combination with paclitaxel, and increase the effectiveness of conventional chemotherapy.
Collapse
|
23
|
Arcusa R, Villaño D, Marhuenda J, Cano M, Cerdà B, Zafrilla P. Potential Role of Ginger (Zingiber officinale Roscoe) in the Prevention of Neurodegenerative Diseases. Front Nutr 2022; 9:809621. [PMID: 35369082 PMCID: PMC8971783 DOI: 10.3389/fnut.2022.809621] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Accepted: 02/15/2022] [Indexed: 12/30/2022] Open
Abstract
Ginger is composed of multiple bioactive compounds, including 6-gingerol, 6-shogaol, 10-gingerol, gingerdiones, gingerdiols, paradols, 6-dehydrogingerols, 5-acetoxy-6-gingerol, 3,5-diacetoxy-6-gingerdiol, and 12-gingerol, that contribute to its recognized biological activities. Among them, the major active compounds are 6-shogaol and 6-gingerol. Scientific evidence supports the beneficial properties of ginger, including antioxidant and anti-inflammatory capacities and in contrast, a specific and less studied bioactivity is the possible neuroprotective effect. The increase in life expectancy has raised the incidence of neurodegenerative diseases (NDs), which present common neuropathological features as increased oxidative stress, neuroinflammation and protein misfolding. The structure-activity relationships of ginger phytochemicals show that ginger can be a candidate to treat NDs by targeting different ligand sites. Its bioactive compounds may improve neurological symptoms and pathological conditions by modulating cell death or cell survival signaling molecules. The cognitive enhancing effects of ginger might be partly explained via alteration of both the monoamine and the cholinergic systems in various brain areas. Moreover, ginger decreases the production of inflammatory related factors. The aim of the present review is to summarize the effects of ginger in the prevention of major neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and multiple sclerosis.
Collapse
|
24
|
The interaction between bovine serum albumin and [6]-,[8]- and [10]-gingerol: An effective strategy to improve the solubility and stability of gingerol. Food Chem 2022; 372:131280. [PMID: 34818732 DOI: 10.1016/j.foodchem.2021.131280] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 09/21/2021] [Accepted: 09/28/2021] [Indexed: 12/29/2022]
Abstract
In this study, the binding mechanism between bovine serum albumin (BSA) and three gingerols ([6]-, [8]- and [10]-gingerol) was evaluated to explore an effective strategy for improving solubility and stability of gingerols. The fluorescence analysis suggested gingerols could bind with BSA to form a stable BSA/gingerols complex and [10]-gingerol had the strongest binding affinity (Ka = 4.016 × 104 L/mol) at 298 K. Thermodynamic parameters and molecular modeling validated that hydrophobic interaction and hydrogen bonds were the main driving force for the interaction of BSA/gingerols. Gingerols bound to BSA at site I (subdomain IIA) resulted in a conformational change of BSA with a structure shrinkage, which was responsible for the decrease of surface hydrophobicity. The formation of BSA/gingerols complexes promoted the solubility of [6]-, [8]- and [10]-gingerol increasing by 1.50, 6.04 and 23.50 times, respectively. In addition, the stability and antioxidant capacity of gingerols was significantly improved after binding with BSA.
Collapse
|
25
|
Chavda VP, Vihol D, Mehta B, Shah D, Patel M, Vora LK, Pereira-Silva M, Paiva-Santos AC. Phytochemical-loaded liposomes for anticancer therapy: an updated review. Nanomedicine (Lond) 2022; 17:547-568. [PMID: 35259920 DOI: 10.2217/nnm-2021-0463] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The major obstacles observed in current chemotherapy are severe adverse effects, narrow therapeutic indexes and multidrug resistance. Anticancer phytochemicals are extracted and purified from natural plants, providing alternative therapeutic approaches with recognized biomedical benefits. However, poor bioavailability, high dose requirements and non-specific targeting have made those molecules less effective. To tackle those issues, liposomal nanovesicles for phytochemical delivery are taken into consideration for improving the therapeutic effectiveness by increasing transportation across cell barriers and conferring attractive cancer-specific targeting capabilities. In the present review, the liposomal approaches of anticancer phytochemicals are discussed, and recent advances in these formulations applied to cancer phytotherapy are further reviewed by an informed approach.
Collapse
Affiliation(s)
- Vivek P Chavda
- Department of Pharmaceutics & Pharmaceutical Technology, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Disha Vihol
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Bhavya Mehta
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Dhruvil Shah
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Manan Patel
- Pharmacy Section, L M College of Pharmacy, Ahmedabad, 380009, India
| | - Lalitkumar K Vora
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, BT9 7BL, UK
| | - Miguel Pereira-Silva
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| | - Ana Cláudia Paiva-Santos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal.,REQUIMTE/LAQV, Group of Pharmaceutical Technology, Faculty of Pharmacy, University of Coimbra, Polo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, 3000-548, Portugal
| |
Collapse
|
26
|
Wang Q, Zhang K, Weng W, Chen L, Wei C, Bao R, Adu-Frimpong M, Cao X, Yu Q, Shi F, Toreniyazov E, Ji H, Xu X, Yu J. Liquiritin-hydroxypropyl-beta-cyclodextrin inclusion complex: preparation, characterization, bioavailability and antitumor activity evaluation. J Pharm Sci 2022; 111:2083-2092. [PMID: 35367247 DOI: 10.1016/j.xphs.2022.03.021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 03/28/2022] [Accepted: 03/28/2022] [Indexed: 10/18/2022]
|
27
|
Preparation and Study of Folate Modified Albumin Targeting Microspheres. JOURNAL OF ONCOLOGY 2022; 2022:3968403. [PMID: 35126516 PMCID: PMC8816550 DOI: 10.1155/2022/3968403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/11/2021] [Accepted: 01/07/2022] [Indexed: 11/17/2022]
Abstract
In this study, folate modified bovine serum albumin was successfully synthesized, while preparation of Nintedanib albumin microspheres (ND-FSA NPs) as a carrier was carried out via electrospinning technology. Folate modified albumin was used to enhance the targeting potential of the prepared microspheres. The prepared microspheres had spherical appearance and smooth outer surface. The diameters of microspheres (764.68 ± 88.46 nm) and zeta potential (- 18.38 ± 0.41 mV) were acceptable. The prepared ND-FSA NPs demonstrated a good degree of modification, wherein the modification rate was 28.1%. In vitro release was significantly increased in three different media (double deionized water-DDW, HCl-pH 1.2, and phosphate buffered solution containing 0.5% Tween 80). It is worth noting that incorporation of Nintedanib into folic acid modified albumin microspheres resulted in an enhanced uptake of the drug into MCF-7 breast cancer cells coupled with higher inhibition rate. Altogether, incorporation of Nintedanib into folate modified albumin microspheres is a new approach to improve water solubility and targeting effect of the drug.
Collapse
|
28
|
Can A, Tyler AI, Mackie AR. Potential use of bile salts in lipid self-assembled systems for the delivery of phytochemicals. Curr Opin Colloid Interface Sci 2021. [DOI: 10.1016/j.cocis.2021.101502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
29
|
Zhao S, Wu Y, Hu L. Identification and synthesis of selective cholesterol esterase inhibitor using dynamic combinatorial chemistry. Bioorg Chem 2021; 119:105520. [PMID: 34864280 DOI: 10.1016/j.bioorg.2021.105520] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 11/22/2021] [Accepted: 11/24/2021] [Indexed: 12/15/2022]
Abstract
In this study, the concept of dynamic combinatorial chemistry (DCC) was applied to explore novel cholesterol esterase (CEase) inhibitors. In the presence of enzyme, two substrates (A1H3 and A2H3) were amplified from the dynamic combinatorial library (DCL), which was generated through reversible acylhydrazone formation reaction. In the in vitro biological evaluation, compound A1H3 exhibited not only potent (IC50 in nanomolar range) but also selective inhibition (>120 folds of selectivity for CEase over AChE). Furthermore, the binding pattern and possible binding mechanism were investigated in the kinetic experiment and molecular docking study, respectively.
Collapse
Affiliation(s)
- Shuang Zhao
- School of Pharmacy, Jiangsu University, Zhenjiang, 301 Xuefu Rd., Zhenjiang, China
| | - Yao Wu
- School of Pharmacy, Jiangsu University, Zhenjiang, 301 Xuefu Rd., Zhenjiang, China
| | - Lei Hu
- School of Pharmacy, Jiangsu University, Zhenjiang, 301 Xuefu Rd., Zhenjiang, China.
| |
Collapse
|
30
|
Lugo-Flores MA, Quintero-Cabello KP, Palafox-Rivera P, Silva-Espinoza BA, Cruz-Valenzuela MR, Ortega-Ramirez LA, Gonzalez-Aguilar GA, Ayala-Zavala JF. Plant-Derived Substances with Antibacterial, Antioxidant, and Flavoring Potential to Formulate Oral Health Care Products. Biomedicines 2021; 9:1669. [PMID: 34829898 PMCID: PMC8615420 DOI: 10.3390/biomedicines9111669] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/07/2021] [Accepted: 11/10/2021] [Indexed: 12/26/2022] Open
Abstract
Bacterial diseases and reactive oxygen species can cause dental caries and oral cancer. Therefore, the present review analyzes and discusses the antibacterial and antioxidant properties of synthetic and plant-derived substances and their current and future patents to formulate dental products. The reviewed evidence indicates that chlorhexidine, fluorides, and hydrogen peroxide have adverse effects on the sensory acceptability of oral care products. As an alternative, plant-derived substances have antimicrobial and antioxidant properties that can be used in their formulation. Also, adding plant metabolites favors the sensory acceptability of dental products compared with synthetic compounds. Therefore, plant-derived substances have antibacterial, antioxidant, and flavoring activity with the potential to be used in the formulation of toothpaste, mouth rinses, dentures cleansers-fixatives, and saliva substitutes.
Collapse
Affiliation(s)
- Marco A. Lugo-Flores
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| | - Karen P. Quintero-Cabello
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| | - Patricia Palafox-Rivera
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| | - Brenda A. Silva-Espinoza
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| | - Manuel Reynaldo Cruz-Valenzuela
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| | - Luis Alberto Ortega-Ramirez
- Unidad Académica San Luis Río Colorado, Universidad Estatal de Sonora, Carretera, Sonoyta-San Luis Río Colorado km. 6.5, Parque Industrial, San Luis Río Colorado C.P. 83500, Sonora, Mexico;
| | - Gustavo Adolfo Gonzalez-Aguilar
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| | - Jesus Fernando Ayala-Zavala
- Centro de Investigacion en Alimentacion y Desarrollo, A.C., Carretera Gustavo Enrique Astiazaran Rosas, No. 46, Col. La Victoria, Hermosillo C.P. 83304, Sonora, Mexico; (M.A.L.-F.); (K.P.Q.-C.); (P.P.-R.); (B.A.S.-E.); (M.R.C.-V.); (G.A.G.-A.)
| |
Collapse
|
31
|
Roudsari NM, Lashgari NA, Momtaz S, Roufogalis B, Abdolghaffari AH, Sahebkar A. Ginger: A complementary approach for management of cardiovascular diseases. Biofactors 2021; 47:933-951. [PMID: 34388275 DOI: 10.1002/biof.1777] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/03/2021] [Accepted: 07/26/2021] [Indexed: 12/20/2022]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide. Inflammation and oxidative stress play critical roles in progression of various types of CVD. Broad pharmacological properties of ginger (the rhizome of Zingiber officinale) and its bioactive components have been reported, suggesting that they can be a therapeutic choice for clinical use. Consistent with its rich phenolic content, the anti-inflammatory and antioxidant properties of ginger have been confirmed in many studies. Ginger modifies many cellular processes and in particular was shown to have potent inhibitory effects against nuclear factor kappa B (NF-κB); signal transducer and activator of transcription; NOD-, LRR-, and pyrin domain-containing proteins; toll-like receptors; mitogen-activated protein kinase; and mammalian target of rapamycin signaling pathways. Ginger also blocks pro-inflammatory cytokines and the activation of the immune system. Ginger suppresses the activity of oxidative molecules such as reactive oxygen species, inducible nitric oxide synthase, superoxide dismutase, glutathione, heme oxygenase, and GSH-Px. In this report, we summarize the biochemical pathologies underpinning a variety of CVDs and the effects of ginger and its bioactive components, including 6-shogaol, 6-gingerol, and 10-dehydrogingerdione. The properties of ginger and its phenolic components, mechanism of action, biological functions, side effects, and methods for enhanced cell delivery are also discussed. Together with preclinical and clinical studies, the positive biological effects of ginger and its bioactive components in CVD support the undertaking of further in vivo and especially clinical studies.
Collapse
Affiliation(s)
- Nazanin Momeni Roudsari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Naser-Aldin Lashgari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Saeideh Momtaz
- Medicinal Plants Research Center, Institute of Medicinal Plants, Academic Center for Education, Culture and Research, Tehran, Iran
- Toxicology and Disease Group, Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network, Tehran, Iran
| | - Basil Roufogalis
- Discipline of Pharmacology, School of Medical Sciences, University of Sydney, Sydney, Australia
- National Institute of Complementary Medicine, Western Sydney University, Westmead, Australia
| | - Amir Hossein Abdolghaffari
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Medicinal Plants Research Center, Institute of Medicinal Plants, Academic Center for Education, Culture and Research, Tehran, Iran
- Toxicology and Disease Group, Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Gastrointestinal Pharmacology Interest Group, Universal Scientific Education and Research Network, Tehran, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- School of Medicine, The University of Western Australia, Perth, Australia
| |
Collapse
|
32
|
Wang Q, Wang Y, Xie Y, Adu-Frimpong M, Wei C, Yang X, Cao X, Deng W, Toreniyazov E, Ji H, Xu X, Yu J. Nonionic surfactant vesicles as a novel drug delivery system for increasing the oral bioavailability of Ginsenoside Rb1. FOOD BIOSCI 2021. [DOI: 10.1016/j.fbio.2021.101064] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
|
33
|
Ge Z, Wang Q, Zhu Q, Yusif M, Yu J, Xu X. Improved oral bioavailability, cellular uptake, and cytotoxic activity of zingerone via nano-micelles drug delivery system. J Microencapsul 2021; 38:394-404. [PMID: 34278929 DOI: 10.1080/02652048.2021.1957036] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Herein, a nano-micelle drug delivery system was developed to orally improved zingerone's bioavailability and its antitumor effect. Indeed, zingerone-loaded d-α-tocopheryl polyethylene glycol succinate micelles (ZTMs) were effectively prepared, characterised and assessed. The ZTMs had diameter, polydispersity index, and zeta potential of 50.62 ± 0.25 nm, 0.168 ± 0.006, and -28.07 ± 0.33 mV, respectively, coupled with a high entrapment efficiency (m/m, %) were 94.71 ± 2.02. The release rate of ZTMs in three media was significantly greater than that of free zingerone. Intriguingly, results obtained from pharmacokinetic studies showed that the oral bioavailability of the ZTMs was enhanced by 5.10 times in comparison with the free zingerone. Further, the half inhibitory concentration (IC50) of ZTMs and free zingerone was 7.56 μg/ml and 14.30 μg/ml, respectively, on HepG2 cells. Hence, ZTMs may be used as a potential approach to enrich the solubility, bioavailability, and concomitant anti-proliferative effect of zingerone in vitro.
Collapse
Affiliation(s)
- Zhumei Ge
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qin Zhu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Mukhtar Yusif
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| |
Collapse
|
34
|
The Potential Role of Polyphenols in Modulating Mitochondrial Bioenergetics within the Skeletal Muscle: A Systematic Review of Preclinical Models. Molecules 2021; 26:molecules26092791. [PMID: 34068459 PMCID: PMC8125960 DOI: 10.3390/molecules26092791] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 04/09/2021] [Accepted: 04/11/2021] [Indexed: 12/19/2022] Open
Abstract
Polyphenols are naturally derived compounds that are increasingly being explored for their various health benefits. In fact, foods that are rich in polyphenols have become an attractive source of nutrition and a potential therapeutic strategy to alleviate the untoward effects of metabolic disorders. The last decade has seen a rapid increase in studies reporting on the bioactive properties of polyphenols against metabolic complications, especially in preclinical models. Various experimental models involving cell cultures exposed to lipid overload and rodents on high fat diet have been used to investigate the ameliorative effects of various polyphenols against metabolic anomalies. Here, we systematically searched and included literature reporting on the impact of polyphenols against metabolic function, particularly through the modulation of mitochondrial bioenergetics within the skeletal muscle. This is of interest since the skeletal muscle is rich in mitochondria and remains one of the main sites of energy homeostasis. Notably, increased substrate availability is consistent with impaired mitochondrial function and enhanced oxidative stress in preclinical models of metabolic disease. This explains the general interest in exploring the antioxidant properties of polyphenols and their ability to improve mitochondrial function. The current review aimed at understanding how these compounds modulate mitochondrial bioenergetics to improve metabolic function in preclinical models on metabolic disease.
Collapse
|
35
|
Wang Y, Chen L, Adu‐Frimpong M, Wei C, Weng W, Wang Q, Xu X, Yu J. Preparation, In Vivo and In Vitro Evaluation, and Pharmacodynamic Study of DMY‐Loaded Self‐Microemulsifying Drug Delivery System. EUR J LIPID SCI TECH 2021. [DOI: 10.1002/ejlt.202000369] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Yaping Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Lin Chen
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Michael Adu‐Frimpong
- Department of Applied Chemistry and Biochemistry CK Tedam University of Technology and Applied Science Navrongo, Upper East Region 31011 Ghana
| | - Chunmei Wei
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Wen Weng
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - Xi‐Ming Xu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| | - JiangNan Yu
- Department of Pharmaceutics, School of Pharmacy, Centre for Nano Drug/Gene Delivery and Tissue Engineering Jiangsu University Zhenjiang 212013 P. R. China
| |
Collapse
|
36
|
Improvement of Oral Bioavailability and Anti-Tumor Effect of Zingerone Self-Microemulsion Drug Delivery System. J Pharm Sci 2021; 110:2718-2727. [PMID: 33610568 DOI: 10.1016/j.xphs.2021.01.037] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/14/2021] [Accepted: 01/21/2021] [Indexed: 12/23/2022]
Abstract
This study sought to prepare a self-microemulsion drug delivery system containing zingerone (Z-SMEDDS) to improve the low oral bioavailability of zingerone and anti-tumor effect. Z-SMEDDS was characterized by particle size, zeta potential and encapsulation efficiency, while its pharmacokinetics and anti-tumor effects were also evaluated. Z-SMEDDS had stable physicochemical properties, including average particle size of 17.29 ± 0.07 nm, the zeta potential of -22.81 ± 0.29 mV, and the encapsulation efficiency of 97.96% ± 0.02%. In vitro release studies have shown the release of zingerone released by Z-SMEDDS was significantly higher than free zingerone in different release media. The relative oral bioavailability of Z-SMEDDS was 7.63 times compared with free drug. Meanwhile, the half inhibitory concentration (IC50)of Z-SMEDDS and free zingerone was 8.45 μg/mL and 13.30 μg/mL, respectively on HepG2. This study may provide a preliminary basis for further clinical research and application of Z-SMEDDS.
Collapse
|
37
|
Garcia-Oliveira P, Otero P, Pereira AG, Chamorro F, Carpena M, Echave J, Fraga-Corral M, Simal-Gandara J, Prieto MA. Status and Challenges of Plant-Anticancer Compounds in Cancer Treatment. Pharmaceuticals (Basel) 2021; 14:ph14020157. [PMID: 33673021 PMCID: PMC7918405 DOI: 10.3390/ph14020157] [Citation(s) in RCA: 104] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/03/2021] [Accepted: 02/06/2021] [Indexed: 12/12/2022] Open
Abstract
Nowadays, cancer is one of the deadliest diseases in the world, which has been estimated to cause 9.9 million deaths in 2020. Conventional treatments for cancer commonly involve mono-chemotherapy or a combination of radiotherapy and mono-chemotherapy. However, the negative side effects of these approaches have been extensively reported and have prompted the search of new therapeutic drugs. In this context, scientific community started to look for innovative sources of anticancer compounds in natural sources, including traditional plants. Currently, numerous studies have evaluated the anticancer properties of natural compounds derived from plants, both in vitro and in vivo. In pre-clinical stages, some promising compounds could be mentioned, such as the sulforaphane or different phenolic compounds. On the other hand, some phytochemicals obtained positive results in clinical stages and were further approved for cancer treatment, such as vinca alkaloids or the paclitaxel. Nevertheless, these compounds are not exempt of limitations, such as low solubility, restricted effect on their own, negative side-effects, etc. This review aims to compile the information about the current phytochemicals used for cancer treatment and also promising candidates, main action mechanisms and also reported limitations. In this sense, some strategies to face the limitations have been considered, such as nano-based formulations to improve solubility or chemical modification to reduce toxicity. In conclusion, although more research is still necessary to develop more efficient and safe phytochemical drugs, more of these compounds might be used in future cancer therapies.
Collapse
Affiliation(s)
- Paula Garcia-Oliveira
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Paz Otero
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Antia Gonzalez Pereira
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Franklin Chamorro
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Maria Carpena
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Javier Echave
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
| | - Maria Fraga-Corral
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Centro de Investigação de Montanha (CIMO), Campus de Santa Apolonia, Instituto Politécnico de Bragança, 5300-253 Bragança, Portugal
| | - Jesus Simal-Gandara
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Correspondence: (J.S.-G.); (M.A.P.)
| | - Miguel Angel Prieto
- Nutrition and Bromatology Group, Analytical and Food Chemistry Department, Faculty of Food Science and Technology, University of Vigo, Ourense Campus, E-32004 Ourense, Spain; (P.G.-O.); (P.O.); (A.G.P.); (F.C.); (M.C.); (J.E.); (M.F.-C.)
- Correspondence: (J.S.-G.); (M.A.P.)
| |
Collapse
|
38
|
Weng W, Wang Q, Wei C, Adu-Frimpong M, Toreniyazov E, Ji H, Yu J, Xu X. Mixed micelles for enhanced oral bioavailability and hypolipidemic effect of liquiritin: preparation, in vitro and in vivo evaluation. Drug Dev Ind Pharm 2021; 47:308-318. [PMID: 33494627 DOI: 10.1080/03639045.2021.1879839] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
OBJECTIVES Liquiritin, as one of the main flavonoids in Glycyrrhiza, exhibits extensive pharmacological effects, such as the anti-oxidant, anti-inflammatory, anti-tumor and so on. Herein, the aqueous solubility and oral bioavailability of liquiritin was purposely enhanced via the preparation of the mixed micelles. METHODS The liquiritin-loaded micelles (LLM) were fabricated via thin-film dispersion method. The optimal LLM formulation was evaluated through physical properties including particle size (PS), encapsulation efficiency (EE) and drug loading (DL). In vitro accumulate release as well as in vivo pharmacokinetics were also evaluated. Moreover, the hypolipidemic activity of LLM was observed in the hyperlipidemia mice model. RESULTS The LLM exhibited a homogenous spherical shape with small mean PS, good stability and high encapsulation efficiency. The accumulate release rates in vitro of the LLM were obviously higher than free liquiritin. The oral bioavailability of the formulation was heightened by 3.98 times in comparison with the free liquiritin. More importantly, LLM increased the hypolipidemic and effect of alleviating lipid metabolism disorder in hepatocytes of liquiritin in hyperlipidemia mice model. CONCLUSIONS Collectively, the improved solubility of liquiritin in water coupled with its enhanced oral bioavailability and concomitant hypolipidemic activity could be attributed to the incorporation of the drug into the mixed micelles.
Collapse
Affiliation(s)
- Wen Weng
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Chunmei Wei
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Elmurat Toreniyazov
- Ashkent State Agricultural University (Nukus branch), Nukus, The Republic of Uzbekistan.,Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China
| | - Hao Ji
- Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China.,Jiangsu Tian Sheng Pharmaceutical Co., Ltd, Zhenjiang, People's Republic of China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.,Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, People's Republic of China.,Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, People's Republic of China
| |
Collapse
|
39
|
Ahmad N, Ahmad R, Amir M, Alam MA, Almakhamel MZ, Ali A, Ahmad A, Ashraf K. Ischemic brain treated with 6-gingerol loaded mucoadhesive nanoemulsion via intranasal delivery and their comparative pharmacokinetic effect in brain. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102130] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
40
|
da Silva JA, Sampaio PA, Dulcey LJL, Cominetti MR, Rabello MM, Rolim LA. Preparation and characterization of [6]-gingerol/β-cyclodextrin inclusion complexes. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2020.102103] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
41
|
Wang Z, Liu L, Yin W, Liu Z, Shi L, Tang M. A Novel Drug Delivery System: the Encapsulation of Naringenin in Metal-Organic Frameworks into Liposomes. AAPS PharmSciTech 2021; 22:61. [PMID: 33527250 DOI: 10.1208/s12249-021-01927-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 01/10/2021] [Indexed: 02/08/2023] Open
Abstract
Poorly water-soluble naringenin (NAR) was selected as a model drug and loaded into the porous MOFs for the construction of NAR@ZIF-8 inclusion complex. By film dispersion method, NAR@ZIF-8 was further encapsulated into liposomes to fabricate a novel drug delivery system. Liposomes and a novel drug delivery system were established. Subsequently, the lipid-drug ratio, phospholipid-cholesterol ratio, and hydration temperature were investigated using the Box-Behnken design based the single factor experiment. The prepared liposomes system showed spherical or quasi-spherical shape, uniform particle size distribution, and complete structure. More specifically, the average particle size was 113.2 ± 1.4 nm, and zeta potential was - 7.536 ± 0.264 mV. Moreover, the drug release behaviors of NAR, NAR@ZIF-8, and NAR@ZIF-8 liposomes were explored in vitro. Compared with free NAR and NAR@ZIF-8 which exhibited a burst drug release, NAR@ZIF-8 liposomes showed a more sustained release behavior with 79.86% drug release in 72 h. In vitro cytotoxicity experiments showed that, compared with free NAR and NAR@ZIF-8, NAR@ZIF-8 liposomes exhibited higher inhibition efficiency on lung adenocarcinoma A549 cells and gastric cancer SGC-7901 cells in a concentration-dependent manner.
Collapse
|
42
|
Enhancement of oral bioavailability and hypoglycemic activity of liquiritin-loaded precursor liposome. Int J Pharm 2021; 592:120036. [DOI: 10.1016/j.ijpharm.2020.120036] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 02/06/2023]
|
43
|
Gu M, Lu L, Wei Q, Miao Z, Zhang H, Gao L, Li L. Improved oral bioavailability and anti-chronic renal failure activity of chrysophanol via mixed polymeric micelles. J Microencapsul 2020; 38:47-60. [PMID: 33175576 DOI: 10.1080/02652048.2020.1849440] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
AIMS This study was designed to prepare chrysophanol-loaded micelles (CLM) to improve the oral bioavailability, targetability and anti-chronic renal failure (CRF) activity of chrysophanol (CH). METHODS The preparation of CLM was achieved via thin-film dispersion technique. The in vitro release of CLM compared with free CH was measured in phosphate buffer solution (PBS) containing 0.5%w/v sodium dodecyl sulphate (pH 6.8) while the pharmacokinetic and anti-CRF activity study was also conducted in rats. Moreover, the tissue distribution of CLM was investigated in the mice. RESULTS The CLM had particle size (PS) of 29.64 ± 0.71 nm, and encapsulation efficiency (EE) of 90.48 ± 1.22%w/w. The cumulative release rate of CH from the micellar system was significantly higher than that of the free CH (86%m/m vs. 15%m/m, p < 0.01). In vivo pharmacokinetic studies showed that the bioavailability of CLM after oral administration was substantially improved (about 3.4 times) compared with free drugs (p < 0.01). Also, it was observed that CLM accumulated well in the liver and brain. Moreover, in vitro renal podocytes study showed that CLM had better protection against renal podocyte damage than the free CH. In addition, CLM significantly (p < 0.01) reduced levels of blood urea nitrogen (BUN), kidney injury molecule-1 (Kim-1), and serum creatinine (SCr), which obviously improved kidney damage in rats with CRF. CONCLUSIONS Collectively, these findings suggest that mixed micelles may be used as a promising drug delivery system for oral bioavailability improvement and concomitantly enhance the anti-CRF activity of CH, as well as provide a basis for the clinical application of CH.
Collapse
Affiliation(s)
- Mingjia Gu
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Lidan Lu
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Qingxue Wei
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Zhiwei Miao
- Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, China
| | - Hang Zhang
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Leiping Gao
- Changshu Hospital Affiliated to Nanjing University of Chinese Medicine, Changshu, China
| | - Lejun Li
- Wuxi Hospital Affiliated to Nanjing University of Chinese Medicine, Wuxi, China
| |
Collapse
|
44
|
Zhang H, Chen B, Zhu Y, Sun C, Adu-Frimpong M, Deng W, Yu J, Xu X. Enhanced oral bioavailability of self-assembling curcumin–vitamin E prodrug-nanoparticles by co-nanoprecipitation with vitamin E TPGS. Drug Dev Ind Pharm 2020; 46:1800-1808. [DOI: 10.1080/03639045.2020.1821049] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Affiliation(s)
- Huiyun Zhang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
- School of Chemistry and Chemical Engineering, Yancheng Institute of Technology, Yancheng, China
| | - Baoding Chen
- Department of Ultrasound, The Affiliated Hospital of Jiangsu University, Zhenjiang, PR China
| | - Yuan Zhu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
| | - Congyong Sun
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
| | - Michael Adu-Frimpong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
| | - Wenwen Deng
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, PR China
| |
Collapse
|
45
|
Zhao S, Zhang S, Xu J, Hu L. Synthesis and comparative study of emulsifying and biological properties of triazolated glucolipids. Tetrahedron 2020. [DOI: 10.1016/j.tet.2020.131517] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
|
46
|
Improved oral bioavailability and target delivery of 6-shogaol via vitamin E TPGS-modified liposomes: Preparation, in-vitro and in-vivo characterizations. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2020.101842] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
47
|
Liu J, Wang Q, Omari-Siaw E, Adu-Frimpong M, Liu J, Xu X, Yu J. Enhanced oral bioavailability of Bisdemethoxycurcumin-loaded self-microemulsifying drug delivery system: Formulation design, in vitro and in vivo evaluation. Int J Pharm 2020; 590:119887. [PMID: 32950666 DOI: 10.1016/j.ijpharm.2020.119887] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2020] [Revised: 09/02/2020] [Accepted: 09/13/2020] [Indexed: 12/17/2022]
Abstract
In this study, we sought to overcome the poor solubility and bioavailability of bismethoxycurcumin (BDMC) by fabricating a BDMC-loaded self micro-emulsifying system (BDMC-SMEDDS). Solubility and compatibility tests, pseudo-ternary phase diagrams (PTPDs) as well as d-optimal concept was applied to design the formulation. The assessment of the prepared BDMC-SMEDDS in-vitro mainly included droplet size (DS) and entrapment efficiency (EE) determination, morphology, drug release and stability testing. Besides, the in vivo behavior was also evaluated after oral administration of BDMC-SMEDDS to rats. The optimal formulation was found to compose of Kolliphor EL (K-EL, emulsifier, 645.3 mg), PEG 400 (co-emulsifier, 147.2 mg), ethyl oleate (EO, oil, 207.5 mg) and BDMC (50 mg). The BDMC-SMEDDS with satisfactory stability had a mean size of 21.25 ± 3.23 nm and EE of 98.31 ± 0.32%. Roughly 70% of BDMC was released from BDMC-SMEDDS within 84 h compared with <20% from the free BDMC. More importantly, the in-vivo behavior of BDMC-SMEDDS showed that the AUC(0-12h) and plasma concentration of BDMC increased substantially as compared to the free BDMC. Altogether, BDMC-SMEDDS has the potential to enhance the solubility and bioavailability of BDMC and could be applied in the clinics.
Collapse
Affiliation(s)
- Jian Liu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Emmanuel Omari-Siaw
- Department of Pharmaceutical Sciences, Faculty of Health Sciences, Kumasi Technical University, Kumasi, Ghana
| | - Michael Adu-Frimpong
- Department of Biochemistry and Biotechnology, Kwame Nkrumah University of Science and Technology, Kumasi, Ghana
| | - Jing Liu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, China.
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang 212013, China.
| |
Collapse
|
48
|
A near-infrared azadipyrromethene dye: Photophysical properties under different acidity conditions. INORG CHEM COMMUN 2020. [DOI: 10.1016/j.inoche.2020.107942] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
49
|
Ahmad R, Khan MA, Srivastava A, Gupta A, Srivastava A, Jafri TR, Siddiqui Z, Chaubey S, Khan T, Srivastava AK. Anticancer Potential of Dietary Natural Products: A Comprehensive Review. Anticancer Agents Med Chem 2020; 20:122-236. [DOI: 10.2174/1871520619666191015103712] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 06/21/2019] [Accepted: 07/02/2019] [Indexed: 02/07/2023]
Abstract
Nature is a rich source of natural drug-like compounds with minimal side effects. Phytochemicals
better known as “Natural Products” are found abundantly in a number of plants. Since time immemorial, spices
have been widely used in Indian cuisine as flavoring and coloring agents. Most of these spices and condiments
are derived from various biodiversity hotspots in India (which contribute 75% of global spice production) and
form the crux of India’s multidiverse and multicultural cuisine. Apart from their aroma, flavor and taste, these
spices and condiments are known to possess several medicinal properties also. Most of these spices are mentioned
in the Ayurveda, the indigenous system of medicine. The antimicrobial, antioxidant, antiproliferative,
antihypertensive and antidiabetic properties of several of these natural products are well documented in
Ayurveda. These phytoconstituemts are known to act as functional immunoboosters, immunomodulators as well
as anti-inflammatory agents. As anticancer agents, their mechanistic action involves cancer cell death via induction
of apoptosis, necrosis and autophagy. The present review provides a comprehensive and collective update
on the potential of 66 commonly used spices as well as their bioactive constituents as anticancer agents. The
review also provides an in-depth update of all major in vitro, in vivo, clinical and pharmacological studies done
on these spices with special emphasis on the potential of these spices and their bioactive constituents as potential
functional foods for prevention, treatment and management of cancer.
Collapse
Affiliation(s)
- Rumana Ahmad
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Mohsin A. Khan
- Chancellor, Era University, Sarfarazganj, Hardoi Road, Lucknow-226003, UP, India
| | - A.N. Srivastava
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Anamika Gupta
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Aditi Srivastava
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tanvir R. Jafri
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Zainab Siddiqui
- Department of Pathology, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Sunaina Chaubey
- Department of Biochemistry, Era’s Lucknow Medical College & Hospital, Era University, Sarfarazganj, Lucknow-226003, UP, India
| | - Tahmeena Khan
- Department of Chemistry, Integral University, Dasauli, P.O. Bas-ha, Kursi Road, Lucknow 226026, UP, India
| | - Arvind K. Srivastava
- Department of Food and Nutrition, Era University, Sarfarazganj, Lucknow-226003, UP, India
| |
Collapse
|
50
|
Phytofabrication of Nanoparticles as Novel Drugs for Anticancer Applications. Molecules 2019; 24:molecules24234246. [PMID: 31766544 PMCID: PMC6930546 DOI: 10.3390/molecules24234246] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/30/2019] [Accepted: 11/08/2019] [Indexed: 02/05/2023] Open
Abstract
Cancer is one of the foremost causes of death globally and also the major stumbling block of increasing life expectancy. Although the primary treatment of surgical resection, chemotherapy, and radiotherapy have greatly reduced the mortality of cancer, the survival rate is still low because of the metastasis of tumor, a range of adverse drug reactions, and drug resistance. For all this, it is relevant to mention that a growing amount of research has shown the anticarcinogenic effect of phytochemicals which can modulate the molecular pathways and cellular events include apoptosis, cell proliferation, migration, and invasion. However, their pharmacological potential is hindered by their low water solubility, low stability, poor absorption, and rapid metabolism. In this scenario, the development of nanotechnology has created novel formulations to maximize the potential use of phytochemicals in anticancer treatment. Nanocarriers can enhance the solubility and stability of phytochemicals, prolong their half-life in blood and even achieve site-targeting delivery. This review summarizes the advances in utilizing nanoparticles in cancer therapy. In particular, we introduce several applications of nanoparticles combined with apigenin, resveratrol, curcumin, epigallocatechin-3-gallate, 6-gingerol, and quercetin in cancer treatment.
Collapse
|