1
|
Bakhtiari R, Shiri M, Reza Mohammadi M, Reza Pourmand M, Mirzaie A, Taghiabadi Z. Enhanced antimicrobial effects of carvacrol against methicillin-resistant Staphylococcus aureus strains using niosome formulations. Rev Argent Microbiol 2025:S0325-7541(24)00122-6. [PMID: 39843290 DOI: 10.1016/j.ram.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 09/25/2024] [Accepted: 10/01/2024] [Indexed: 01/24/2025] Open
Abstract
Methicillin-resistant Staphylococcus aureus (MRSA) causes a wide range of infections and contributes to elevated morbidity, mortality, and healthcare costs. Herbal compounds combined with drug delivery systems could be an effective alternative option for treating resistant bacteria. This study evaluates the antimicrobial prowess of carvacrol-loaded niosomes against MRSA strains. In this study, six carvacrol-niosome formulations with different ratios of Span and Tween were prepared. The physicochemical attributes of the optimized synthesized niosomes were assessed using Fourier-transform infrared (FTIR) spectroscopy and scanning electron microscopy (SEM) as well as DLS Zetasizer. Encapsulation efficiency (EE) and in vitro drug release were studied. The antibacterial activity of the synthesized carvacrol-niosomes, in concentrations varying between 7.8 and 1000μg/ml, was evaluated using microdilution broth methods. The optimized niosomes, with a size of 207.3nm and an impressive EE of 91%, exhibited a spherical structure as confirmed by the electron microscopy analysis. Impressively, these carvacrol-niosomes demonstrated superior antimicrobial effectiveness against S. aureus, reducing MIC levels 4-fold to 62.5±0.0μg/ml and MBC to 125±0.0μg/ml, a significant improvement over the 250±0.0μg/ml MIC and 500±0.0μg/ml MBC of free carvacrol. Additionally, while empty niosomes showed minimal cytotoxicity with 88.32±1.32% cell viability at 100μg/ml, free carvacrol led to a marked reduction in viability to 39.46±1.26%. However, niosomes encapsulating carvacrol notably increased cell survival to 59.67±1.62% at this concentration. These findings underscore the enhanced antimicrobial potency of carvacrol when enclosed within niosomes, suggesting its potential as a potent herbal remedy for combating methicillin-resistant S. aureus.
Collapse
Affiliation(s)
- Ronak Bakhtiari
- Department of Pathobiology, Division of Microbiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| | - Maliheh Shiri
- Department of Pathobiology, Division of Microbiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Mohammadi
- Department of Bacteriology, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Reza Pourmand
- Department of Pathobiology, Division of Microbiology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Mirzaie
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Taghiabadi
- Department of Biology, Parand Branch, Islamic Azad University, Parand, Iran; Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
2
|
Nguyen HX, Le NY, Nguyen CN. Quality by design optimization of formulation variables and process parameters for enhanced transdermal delivery of nanosuspension. Drug Deliv Transl Res 2024:10.1007/s13346-024-01733-4. [PMID: 39496992 DOI: 10.1007/s13346-024-01733-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
This investigation aims to fabricate, characterize, and optimize organogel containing andrographolide nanosuspension to enhance transdermal drug delivery into and across the skin in vitro. We identified the critical material attributes (CMAs) and critical process parameters (CPPs) that impact key characteristics of andrographolide nanosuspension using a systematic quality-by-design approach. We prepared andrographolide nanosuspension using the wet milling technique and evaluated various properties of the formulations. The CMAs were types and concentrations of polymers, types and concentrations of surfactants, drug concentration, and lipid concentration. The CPPs were volume of milling media and milling duration. Mean particle size, polydispersity index, encapsulation efficiency, and drug loading capacity as critical quality attributes were selected in the design for the evaluation and optimization of the formulations. Furthermore, we developed and evaluated organogel formulation to carry andrographolide nanosuspension 0.05% w/w. Drug release and permeation studies were conducted to assess the drug release kinetics and transdermal delivery of andrographolide. We presented the alteration in the average particle size, polydispersity index, encapsulation efficiency, drug-loading capacity, and drug release among various formulations to select the optimal parameters. The permeation study indicated that organogel delivered markedly more drug into the receptor fluid and skin tissue than DMSO gel (n = 3, p < 0.05). This enhancement in transdermal drug delivery was demonstrated by cumulative drug permeation after 24 h, steady-state flux, permeability coefficient, and predicted steady-state plasma concentration. Drug quantity in skin layers, total delivery, delivery efficiency, and topical selectivity were also reported. Conclusively, andrographolide nanosuspension-loaded organogel significantly increased transdermal drug delivery in vitro.
Collapse
Affiliation(s)
- Hiep X Nguyen
- College of Pharmacy, California Northstate University, 9700 West Taron Drive, Elk Grove, CA, 95757, USA
- Novoremedy, 2001 Talmage Rd, Ukiah, CA, 95482, USA
| | - Nhi Y Le
- Faculty of Pharmaceutics and Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi, 100000, Vietnam
| | - Chien N Nguyen
- Faculty of Pharmaceutics and Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi, 100000, Vietnam.
- National Institute of Pharmaceutical Technology, Hanoi University of Pharmacy, Hanoi, 100000, Vietnam.
| |
Collapse
|
3
|
Chaurawal N, Quadir SS, Joshi G, Barkat MA, Alanezi AA, Raza K. Development of fucoidan/polyethyleneimine based sorafenib-loaded self-assembled nanoparticles with machine learning and DoE-ANN implementation: Optimization, characterization, and in-vitro assessment for the anticancer drug delivery. Int J Biol Macromol 2024; 279:135123. [PMID: 39208886 DOI: 10.1016/j.ijbiomac.2024.135123] [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: 01/20/2024] [Revised: 08/19/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
This study aims to develop sorafenib-loaded self-assembled nanoparticles (SFB-SANPs) using the combined approach of artificial neural network and design of experiments (ANN-DoE) and to compare it with other machine learning (ML) models. The central composite design (CCD) and ML algorithms were used to screen the effects of concentrations of both the polymers (polyethyleneimine and fucoidan) on the outcome responses, i.e., particle size and entrapment efficiency with defined constraints. The prediction from different ML models (bootstrap forest, K-nearest neighbors, artificial neural network, generalized regression-lasso and support vector machines) were compared with ANN-DoE model. The ANN-DoE model showed better accuracy and predictability and outperformed all the other models. This depicted that the concept of using ANN and DoE combination approach provided the best, uncomplicated and cost-effective way to optimized the nanoformulations. The optimized formulation generated from the ANN-DoE combined model was further evaluated for characterization and anticancer activity. The optimized SFB-SANPs were prepared using the polyelectrolyte complexation method with Polyethyleneimine (PEI) as a cationic polymer and fucoidan (FCD) as an anionic. The SFB-SANPs were nanometric in size (280.4 ± 0.089 nm) and slightly anionic in nature (zeta potential = -6.03 ± 0.92 mV) with an encapsulation efficiency of 95.56 ± 0.30 %. The drug release from SFB-SANPs was controlled and sustained in the cancer microenvironment (pH 5.0). The SFB-SANPs were compatible with red blood cells (RBCs), and the % hemolysis was found to be <5.0 %. The anticancer activity of the SFB-SANPs exhibited an IC50 at 2.017 ± 0.516 μM against MDMB-231 cells, showing a significantly high inhibitory effect on breast cancer cell lines. Therefore, the nanocarriers developed using various ML tools inherit a huge promise in anticancer drug delivery.
Collapse
Affiliation(s)
- Nishtha Chaurawal
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan -305817, India
| | - Sheikh Shahnawaz Quadir
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India
| | - Garima Joshi
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur, Rajasthan 313001, India
| | - Md Abul Barkat
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, 39524, Saudi Arabia
| | - Abdulkareem Ali Alanezi
- Department of Pharmaceutics, College of Pharmacy, University of Hafr Al Batin, 39524, Saudi Arabia
| | - Kaisar Raza
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandarsindri, Ajmer, Rajasthan -305817, India.
| |
Collapse
|
4
|
Sarhan FA, Soliman ME, Hamza MY, El-Gogary RI. Revolutionizing treatment for topical fungal infections: evaluating penetration-enhancer-containing vesicles as a fluconazole delivery system: Ex-vivo and in-vivo dermal testing. Pharm Dev Technol 2024; 29:814-823. [PMID: 39161985 DOI: 10.1080/10837450.2024.2394573] [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: 06/02/2024] [Revised: 08/07/2024] [Accepted: 08/16/2024] [Indexed: 08/21/2024]
Abstract
Fungal infections pose a significant challenge in numerous developing nations and worldwide, necessitating urgent solutions. Oral administration of antifungal medications often leads to severe adverse reactions. Hence, employing topical delivery systems is preferred to ensure efficient dermal delivery of antifungal agents while minimizing side effects. Furthermore, the incorporation of penetration enhancers into nanocarriers loaded with antifungal agents has demonstrated enhanced efficacy in combating mycotic infections. Consequently, ultra-deformable penetration enhancer-containing vesicles (PEVs) were developed to explore this promising approach. In this study, Labrasol® and Transcutol® were used as penetration enhancers in formulating ultra-deformable PEVs containing the antifungal agent Fluconazole (FCZ). The PEVs underwent comprehensive characterization, including measurements of particle size (PS), charge, and entrapment efficiency (EE%). The results revealed that the size of tested PEVs ranged from 100 to 762 nm. All particles exhibited a negative charge, with a minimum zeta potential (ZP) of -38.26 mV, and an intermediate entrapment efficiency (EE%) that reached approximately 40%w/w. Ex-vivo studies demonstrated the ability of PEVs to deliver FCZ to the dermis while minimizing transdermal delivery. The selected formula was tested in-vivo using candidiasis-induced rat model and showed a superiority in its antifungal effect against Candida Albicans compared to the drug control. Stability studies were executed for the selected formula, and revealed good stability shown by the insignificant change in the PS, ZP& EE% over a six-month period.
Collapse
Affiliation(s)
- Fatma A Sarhan
- School of Life and Medical Sciences, University of Hertfordshire Hosted by Global Academic Foundation, Cairo, Egypt
- Department of Pharmaceutics (Physical Properties), Egyptian Drug Authority (EDA) Formerly Known as National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Mahmoud E Soliman
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
- Departement of Pharmaceutics, Egypt-Japan University of Science and Technology (EJUST), Alexandria, Egypt
| | - Manal Yassin Hamza
- Department of Pharmaceutics (Physical Properties), Egyptian Drug Authority (EDA) Formerly Known as National Organization for Drug Control and Research (NODCAR), Cairo, Egypt
| | - Riham I El-Gogary
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Ain Shams University, Cairo, Egypt
| |
Collapse
|
5
|
Rawat PS, Ravi PR, Mahajan RR. Design, pharmacokinetic, and pharmacodynamic evaluation of a lecithin-chitosan hybrid nanoparticle-loaded dual-responsive in situ gel of nebivolol for effective treatment of glaucoma. DISCOVER NANO 2024; 19:156. [PMID: 39331225 PMCID: PMC11436582 DOI: 10.1186/s11671-024-04109-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
In this research work, optimized nebivolol-loaded lecithin-chitosan hybrid nanoparticles (NEB-LCNPs) were prepared using sequential screening and optimization designs. The design of experiments software (DoE) was used to obtain a robust formulation that can improve ocular delivery of the NEB in the treatment of glaucoma. The optimized NEB-LCNPs had a mean particle size of 170.5 ± 5.3 nm and drug loading of 10.5 ± 1.2%. These were further loaded in a dual-responsive in situ gel, designed and reported previously by our group. The NEB-LCNPs loaded in situ gel (NEB-LCNPs-ISG) was characterized for physicochemical properties, rheological behavior, stability, in vitro dissolution, and ocular in vivo studies. The ocular pharmacokinetics showed that NEB-LCNPs-ISG had two-fold higher aqueous humor exposure with AUC0-tlast of 375.4 ng × h/mL and sustained drug concentrations for longer durations (1.7-folds higher duration with a mean residence time of 10.6 h) in comparison to a conventional aqueous suspension of NEB (NEB-Susp). Similarly, the pharmacodynamic study showed that NEB-LCNPs-ISG resulted in a higher percentage reduction in intraocular pressure (% ΔIOP) of 28.1 ± 1.8% × h, which was 2.2-times higher reduction compared to NEB-Susp (74.2 ± 3.2% × h). In addition, the pharmacodynamic effect was more sustained with a mean response time of 11.3 ± 0.2 h, a 2.8-times higher response time compared to NEB-Susp (4.06 ± 0.3 h). These results suggest that NEB-LCNPs-ISG was more effective than the conventional aqueous suspension of NEB in the treatment of glaucoma.
Collapse
Affiliation(s)
- Pradeep Singh Rawat
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana, 500 078, India
| | - Punna Rao Ravi
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana, 500 078, India.
| | - Radhika Rajiv Mahajan
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Hyderabad Campus, Jawahar Nagar, Kapra Mandal, Medchal District, Telangana, 500 078, India
| |
Collapse
|
6
|
Sheng Q, Wang X, Hou Z, Liu B, Jiang M, Ren M, Fu J, He M, Zhang J, Xiang Y, Zhang Q, Zhou L, Deng Y, Shen X. Novel functions of o-cymen-5-ol nanoemulsion in reversing colistin resistance in multidrug-resistant Klebsiella pneumoniae infections. Biochem Pharmacol 2024; 227:116384. [PMID: 38909787 DOI: 10.1016/j.bcp.2024.116384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 06/25/2024]
Abstract
Multidrug resistance (MDR) Klebsiella pneumoniae (K. pneumoniae) is a major emerging threat to human health, and leads to very high mortality rate. The effectiveness of colistin, the last resort against MDR Gram-negative bacteria, is significantly compromised due to the widespread presence of plasmid- or chromosome-mediated resistance genes. In this study, o-cymen-5-ol has been found to greatly restore colistin sensitivity in MDR K. pneumoniae. Importantly, this compound does not impact bacterial viability, induce resistance, or cause any noticeable cell toxicity. Various routes disclosed the potential mechanism of o-cymen-5-ol potentiating colistin activity against MDR K. pneumoniae. These include inhibiting the activity of plasmid-mediated mobile colistin resistance gene (mcr-1), accelerating lipopolysaccharide (LPS) - mediated membrane damage, and promoting the ATP-binding cassette (ABC) transporter pathway. To enhance the administration and bioavailability of o-cymen-5-ol, a nanoemulsion has been designed, which significantly improves the loading efficiency and solubility of o-cymen-5-ol, resulting in antimicrobial potentiation of colistin against K. pneumoniae infection. This study has revealed a new understanding of the o-cymen-5-ol nanoemulsion as a means to enhance the effectiveness of colistin against resistant factors. The finding also suggests that o-cymen-5-ol nanoemulsion could be a promising approach in the development of potential treatments for multidrug-resistant Gram-negative bacterial infections.
Collapse
Affiliation(s)
- Qiushuang Sheng
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Xiao Wang
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Zhaoyan Hou
- Changchun Center for Disease Control and Prevention, Changchun, China
| | - Bin Liu
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Mingquan Jiang
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Mingyue Ren
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Jingchao Fu
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Miao He
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Jingchen Zhang
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Yue Xiang
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Qingbo Zhang
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Lanying Zhou
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun 130103, China
| | - Yanhong Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| | - Xue Shen
- Department of Food Science, College of Food Science and Engineering, Jilin University, Changchun 130062, China; State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine, Jilin University, Changchun 130062, China.
| |
Collapse
|
7
|
Zang J, Zhang L, Guo R, Kong L, Yu Y, Li S, Liu M, Wang J, Zhang Z, Li X, Liu Y. Baicalein loaded liposome with hyaluronic acid and Polyhexamethylene guanidine modification for anti methicillin-resistant Staphylococcus aureus infection. Int J Biol Macromol 2024; 276:133432. [PMID: 38936579 DOI: 10.1016/j.ijbiomac.2024.133432] [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: 12/17/2023] [Revised: 06/15/2024] [Accepted: 06/24/2024] [Indexed: 06/29/2024]
Abstract
Targeting delivery to the infection site and good affinity of vehicle to the bacterial are two main concerns in therapy of bacterial infection, and on-demand release of drug is another important issue. In this work, a liposome drug delivery system (HA/P/BAI-lip) incorporated with baicalein and modified by PHMG and HA was prepared. Several characterizations were conducted to examine the physical properties of liposome. Then it was applied to treatments of MRSA induced dorsal subcutaneous abscess model and the thigh muscle infected model. The presence of guanidine group in HA/P/BAI-lip rendered the liposome satisfactory bacterial target ability and good pH sensitive properties. The lipase secreted by bacterial could promote the hydrolysis of soybean phosphatidylcholine (SPC) in liposome. The modification of HA in HA/P/BAI-lip could lead the drug system to the exact infected site where CD44 was abundant because of inflammation. The low pH microenvironment characteristic of bacterial infection could induce the swelling of liposome following by degradation. Taken together, baicalein could be released selectively at the infected site to exert antibacterial capacity. HA/P/BAI-lip showed impressive antibacterial ability and dramatically decrease the bacterial burden of infection site and alleviate the infiltration of inflammatory cells, facilitating the recovery of infection.
Collapse
Affiliation(s)
- Juan Zang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Lu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Ruibo Guo
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Liang Kong
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Yang Yu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Shutong Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Mo Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Jiahua Wang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Zixu Zhang
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China
| | - Xuetao Li
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China.
| | - Yang Liu
- School of Pharmacy, Liaoning University of Traditional Chinese Medicine, Shengming 1 Road 77, Double D port, Dalian 116600, China.
| |
Collapse
|
8
|
Es-Haghi A, Soltani M, Tabrizi MH, Noghondar MK, Khatamian N, Naeeni NB, Kharaghani M. The effect of EGCG/tyrosol-loaded chitosan/lecithin nanoparticles on hyperglycemia and hepatic function in streptozotocin-induced diabetic mice. Int J Biol Macromol 2024; 267:131496. [PMID: 38626839 DOI: 10.1016/j.ijbiomac.2024.131496] [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: 11/03/2023] [Revised: 04/01/2024] [Accepted: 04/08/2024] [Indexed: 04/21/2024]
Abstract
We aimed to study the potential of epigallocatechin-3-gallate/tyrosol-loaded chitosan/lecithin nanoparticles (EGCG/tyrosol-loaded C/L NPs) in streptozotocin-induced type 2 diabetes mellitus (T2DM) mice. The EGCG/tyrosol-loaded C/L NPs were created using the self-assembly method. Dynamic light scattering, Field Emission Scanning Electron Microscopy, and Fourier transform infrared spectroscopy were utilized to characterize the nanoparticle. Furthermore, in streptozotocin-induced T2DM mice, treatment with EGCG/tyrosol-loaded C/L NPs on fasting blood sugar levels, the expression of PCK1 and G6Pase, and IL-1β in the liver, liver glutathione content, nanoparticle toxicity on liver cells, and liver reactive oxygen species were measured. Our findings showed that EGCG/tyrosol-loaded C/L NPs had a uniform size distribution, and encapsulation efficiencies of 84 % and 89.1 % for tyrosol and EGCG, respectively. The nanoparticles inhibited PANC-1 cells without affecting normal HFF cells. Furthermore, EGCG/tyrosol-loaded C/L NP treatment reduced fasting blood sugar levels, elevated hepatic glutathione levels, enhanced liver cell viability, and decreased reactive oxygen species levels in diabetic mice. The expression of gluconeogenesis-related genes (PCK1 and G6 Pase) and the inflammatory gene IL-1β was downregulated by EGCG/tyrosol-loaded C/L NPs. In conclusion, the EGCG/tyrosol-loaded C/L NPs reduced hyperglycemia, oxidative stress, and inflammation in diabetic mice. These findings suggest that EGCG/tyrosol-loaded C/L NPs could be a promising therapeutic option for type 2 diabetes management.
Collapse
Affiliation(s)
- Ali Es-Haghi
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran.
| | - Mozhgan Soltani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Maryam Karimi Noghondar
- Department of Nursing, Faculty of Nursing and Midwifery, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Niloufar Khatamian
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | | | - Matin Kharaghani
- Department of Biology, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| |
Collapse
|
9
|
Mangla B, Mittal P, Kumar P, Javed S, Ahsan W, Aggarwal G. Development of erlotinib-loaded nanotransferosomal gel for the topical treatment of ductal carcinoma in situ. Nanomedicine (Lond) 2024; 19:855-874. [PMID: 38440976 DOI: 10.2217/nnm-2023-0260] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024] Open
Abstract
Aims: This study was aimed to formulate erlotinib (ERL)-loaded transferosomal gel (ERL@TG) intended for topical application for the treatment of ductal carcinoma in situ. Materials & methods: The optimized process involved a thin-film hydration method to generate ERL-loaded transferosomes (ERL@TFS), which was incorporated into a carbopol gel matrix to generate ERL@TG. The optimized formulation was characterized in vitro followed by cytotoxicity evaluation on MCF-7 breast cancer cell lines and acute toxicity and skin irritation studies was performed in vivo. Results: In a comparative assessment against plain ERL, ERL@TG displayed enhanced efficacy against MCF-7 cell lines, reflected in considerably lower IC50 values with an enhanced safety profile. Conclusion: Optimized ERL@TG was identified as a promising avenue for addressing ductal carcinoma in situ breast cancer.
Collapse
Affiliation(s)
- Bharti Mangla
- Centre for Advanced Formulation & Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Priya Mittal
- Centre for Advanced Formulation & Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Pankaj Kumar
- Centre for Advanced Formulation & Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| | - Shamama Javed
- Department of Pharmaceutics, College of Pharmacy, Jazan University, PO box no. 114, Jazan, Saudi Arabia
| | - Waquar Ahsan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, PO box no. 114, Jazan, Saudi Arabia
| | - Geeta Aggarwal
- Centre for Advanced Formulation & Technology, Delhi Pharmaceutical Sciences and Research University, New Delhi, 110017, India
| |
Collapse
|
10
|
Fayed B, Jagal J, Cagliani R, Kedia RA, Elsherbeny A, Bayraktutan H, Khoder G, Haider M. Co-administration of amoxicillin-loaded chitosan nanoparticles and inulin: A novel strategy for mitigating antibiotic resistance and preserving microbiota balance in Helicobacter pylori treatment. Int J Biol Macromol 2023; 253:126706. [PMID: 37673144 DOI: 10.1016/j.ijbiomac.2023.126706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/25/2023] [Accepted: 09/03/2023] [Indexed: 09/08/2023]
Abstract
Helicobacter pylori (H. pylori) is a causative agent of various gastrointestinal diseases and eradication mainly relies on antibiotic treatment, with (AMX) being a key component. However, rising antibiotic resistance in H. pylori necessitates the use of antibiotics combination therapy, often disrupting gut microbiota equilibrium leading to further health complications. This study investigates a novel strategy utilizing AMX-loaded chitosan nanoparticles (AMX-CS NPs), co-administered with prebiotic inulin to counteract H. pylori infection while preserving microbiota health. Following microbroth dilution method, AMX displayed efficacy against H. pylori, with a MIC50 of 48.34 ± 3.3 ng/mL, albeit with a detrimental impact on Lactobacillus casei (L. casei). The co-administration of inulin (500 μg/mL) with AMX restored L. casei viability while retaining the lethal effect on H. pylori. Encapsulation of AMX in CS-NPs via ionic gelation method, resulted in particles of 157.8 ± 3.85 nm in size and an entrapment efficiency (EE) of 86.44 ± 2.19 %. Moreover, AMX-CS NPs showed a sustained drug release pattern over 72 h with no detectable toxicity on human dermal fibroblasts cell lines. Encapsulation of AMX into CS NPs also reduced its MIC50 against H. pylori, while its co-administration with inulin maintained L. casei viability. Interestingly, treatment with AMX-CS NPs also reduced the expression of the efflux pump gene hefA in H. pylori. This dual treatment strategy offers a promising approach for more selective antimicrobial treatment, minimizing disruption to healthy microbial communities while effectively addressing pathogenic threats.
Collapse
Affiliation(s)
- Bahgat Fayed
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Chemistry of Natural and Microbial Product Department, National Research Centre, Cairo 12622, Egypt
| | - Jayalakshmi Jagal
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Roberta Cagliani
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Reena A Kedia
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Amr Elsherbeny
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom; Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Hulya Bayraktutan
- Division of Molecular Therapeutics and Formulation, School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, United Kingdom; Biodiscovery Institute, School of Medicine, University of Nottingham, Nottingham NG7 2UH, United Kingdom
| | - Ghalia Khoder
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| | - Mohamed Haider
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates; Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| |
Collapse
|
11
|
Sheng Q, Wang N, Zhou Y, Deng X, Hou X, Wang J, Qiu J, Deng Y. A new function of thymol nanoemulsion for reversing colistin resistance in Salmonella enterica serovar Typhimurium infection. J Antimicrob Chemother 2023; 78:2983-2994. [PMID: 37923362 DOI: 10.1093/jac/dkad342] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 10/06/2023] [Indexed: 11/07/2023] Open
Abstract
BACKGROUND Adjuvant addition of approved drugs or foodborne additives to colistin might be a cost-effective strategy to overcome the challenge of plasmid-mediated mobile colistin resistance gene emergence, which poses a threat in the clinic and in livestock caused by infections with Gram-negative bacteria, especially carbapenem-resistant Enterobacteriaceae. METHODS Chequerboard assay was applied to screen the colistin adjuvants from natural compounds. The killing-time curve, combined disc test and membrane permeation assay were conducted to identify the synergy efficacy of thymol and colistin in vitro. Thin-layer chromatography (TLC), LC-MS and fluorescence spectra were used to indicate the interaction of thymol and MCR-1. The potential binding sites were then investigated by molecular simulation dynamics. Finally, a thymol nanoemulsion was prepared with high-pressure homogenization as the clinical dosage form. RESULTS Thymol presented an excellent synergistic effect in vitro with colistin against Salmonella enterica serovar Typhimurium and Escherichia coli bacteria. Thymol addition, forming a complex with MCR-1, might interfere with the efficacy of MCR-1. Moreover, thymol strengthened colistin activity associated with potentiating membrane damage, destroying the biofilm and enhancing reactive oxygen species-mediated oxidative damage. Thymol nanoemulsion combined with colistin remarkably prevented the intestinal damage caused by S. Typhimurium infection, resulting in a survival rate higher than 60%. CONCLUSIONS This study achieved a promising thymol oral formulation as colistin adjuvant to combat S. Typhimurium infection, which could be used to extend the lifespan of colistin in clinical veterinary medicine.
Collapse
Affiliation(s)
- Qiushuang Sheng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Department of Microbiology, Jilin Province Product Quality Supervision and Inspection Institute, Changchun, Jilin, China
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Nan Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yonglin Zhou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xuming Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoning Hou
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Jianfeng Wang
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiazhang Qiu
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Yanhong Deng
- State Key Laboratory for Zoonotic Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
- Department of Respiratory Medicine, Center for Pathogen Biology and Infectious Diseases, Key Laboratory of Organ Regeneration and Transplantation of the Ministry of Education, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
12
|
Kong F, Sun J, Hu Y, Huo W, Li D, Zhang W. Liver-Targeting Composite Nanocarrier Delivery System Based on Chitosan Nanoparticles and Phospholipid Complexes. Assay Drug Dev Technol 2023; 21:357-368. [PMID: 38096118 DOI: 10.1089/adt.2023.051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2023] Open
Abstract
Liver fibrosis is mainly caused by excessive accumulation of extracellular matrix and structural changes in the liver, ultimately leading to cirrhosis if left untreated. Reducing hyaluronan synthesis by inhibiting hyaluronic acid deposition or regulating the expression of hyaluronic synthase can ameliorate liver fibrosis symptoms. In this study, we aimed to improve the bioavailability and liver-targeting capacity of hydroxymethyl coumarin (4-MU) using a newly developed phospholipid complex chitosan nanoparticle (4-MU PC/CNP) optimized using the Box-Behnken design. The composite nanocarrier delivery system was formulated using solvent evaporation technology, and formulation and process parameters were evaluated. Furthermore, 4-MU PC/CNPs and their pharmacokinetics were characterized. The established 4-MU PC/CNPs had an average particle size of 153.07 ± 0.29 nm, a polydispersity index value of 0.383, and a positive zeta potential of ∼35.4 mV. Compared with 4-MUs, 4-MU PC/CNPs exhibited significantly improved water solubility, faster plasma clearance and tissue distribution, and better liver targeting. Pharmacokinetic analysis showed that the oral bioavailability of 4-MU in 4-MU PC/CNPs was significantly higher than that of simple 4-MU. In conclusion, 4-MU PC improved drug lipid (oil-water distribution coefficient of 1.31 ± 0.03) and water solubilities (2.05 times the drug substance). 4-MU PC/CNPs significantly improved 4-MU oral bioavailability, representing a promising approach for enhancing drug solubility. This study demonstrates that the targeting parameters of 4-MU PC/CNPs in the liver were all greater than 1, indicating that they specifically targeted the liver, thereby potentially alleviating liver fibrosis.
Collapse
Affiliation(s)
- Fanming Kong
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Jingmeng Sun
- Department of Pharmacy, The First Hospital of Jilin University, Changchun, China
| | - Yue Hu
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Wenkai Huo
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Dongdong Li
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| | - Weiyu Zhang
- College of Pharmacy, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
13
|
Dawoud MHS, Mannaa IS, Abdel-Daim A, Sweed NM. Integrating Artificial Intelligence with Quality by Design in the Formulation of Lecithin/Chitosan Nanoparticles of a Poorly Water-Soluble Drug. AAPS PharmSciTech 2023; 24:169. [PMID: 37552427 DOI: 10.1208/s12249-023-02609-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 06/25/2023] [Indexed: 08/09/2023] Open
Abstract
The aim of the current study is to explore the potential of artificial intelligence (AI) when integrated with Quality by Design (QbD) approach in the formulation of a poorly water-soluble drug, for its potential use in carcinoma. Silymarin is used as a model drug for its potential effectiveness in liver cancer. A detailed QbD approach was applied. The effect of the critical process parameters was studied on each of the particle size, size distribution, and entrapment efficiency. Response surface designs were applied in the screening and optimization of lecithin/chitosan nanoparticles, to obtain an optimized formula. The release rate was tested, where artificial neural network models were used to predict the % release of the drug from the optimized formula at different time intervals. The optimized formula was tested for its cytotoxicity. A design space was established, with an optimized formula having a molar ratio of 18.33:1 lecithin:chitosan and 38.35 mg silymarin. This resulted in nanoparticles with a size of 161 nm, a polydispersity index of 0.2, and an entrapment efficiency of 97%. The optimized formula showed a zeta potential of +38 mV, with well-developed spherical particles. AI successfully showed high prediction ability of the drug's release rate. The optimized formula showed an enhancement in the cytotoxic effect of silymarin with a decreased IC50 compared to standard silymarin. Lecithin/chitosan nanoparticles were successfully formulated, with deep process and product understanding. Several tools were used as AI which could shift pharmaceutical formulations from experience-dependent studies to data-driven methodologies in the future.
Collapse
Affiliation(s)
- Marwa H S Dawoud
- Department of Pharmaceutics, Faculty of Pharmacy, October University for Modern Sciences and Arts, intersection of 26th of July road and Elwahat road, 6th of October city, Giza, Egypt.
| | - Islam S Mannaa
- Department of Pharmaceutics, Faculty of Pharmacy, October University for Modern Sciences and Arts, intersection of 26th of July road and Elwahat road, 6th of October city, Giza, Egypt
| | - Amira Abdel-Daim
- Department of Biochemistry, Faculty of Pharmacy, October University for Modern Sciences and Arts, Giza, Egypt
| | - Nabila M Sweed
- Department of Pharmaceutics, Faculty of Pharmacy, October University for Modern Sciences and Arts, intersection of 26th of July road and Elwahat road, 6th of October city, Giza, Egypt
| |
Collapse
|
14
|
Yao S, Chen N, Sun X, Wang Q, Li M, Chen Y. Size-Dependence of the Skin Penetration of Andrographolide Nanosuspensions: In Vitro Release-Ex Vivo Permeation Correlation and Visualization of the Delivery Pathway. Int J Pharm 2023:123065. [PMID: 37225025 DOI: 10.1016/j.ijpharm.2023.123065] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 05/04/2023] [Accepted: 05/17/2023] [Indexed: 05/26/2023]
Abstract
Particle size is a key parameter to determine the capacity of nanoparticles to overcome the skin barrier; however, such effect and the possible mechanism remain only partially understood for nanosuspensions. In this work, we examined the skin delivery performance of andrographolide nanosuspensions (AG-NS) ranging in diameter from 250 nm to 1000 nm and analyzed the role of particle size in influencing their ability of skin penetration. The AG-NS with particle sizes of about 250 nm (AG-NS250), 450 nm (AG-NS450), and 1000 nm (AG-NS1000) were successfully prepared by ultrasonic dispersion method and characterized by transmission electron microscopy. The drug release and penetration via the intact and barrier-removed skin were compared by the Franz cell method, and the related mechanisms were probed using laser scanning confocal microscopy (LSCM) via visualization of penetration routes and histopathological study via observation of structural change of the skin. Our finding revealed that drug retention in the skin or its sub-layers was increased with the reduction of particle size, and the drug permeability through the skin also exhibited an obvious dependence on the particle size from 250 nm to 1000 nm. The linear relationship between the in vitro drug release and ex vivo permeation through the intact skin was well established among different preparations and in each preparation, indicating the skin permeation of the drug was mainly determined by the release process. The LSCM indicated that all these nanosuspensions could deliver the drug into the intercellular lipid space, as well as block the hair follicle in the skin, where a similar size dependence was also observed. The histopathological investigation showed that the formulations could make the stratum corneum of the skin loose and swelling without severe irritation. In conclusion, the reduction of particle size of nanosuspension would facilitate topical drug retention mainly via the modulation of drug release.
Collapse
Affiliation(s)
- Sicheng Yao
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Naiying Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Xinxing Sun
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Qiuyue Wang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Mingming Li
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China
| | - Yang Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, Shenyang 110122, China.
| |
Collapse
|
15
|
Lin TC, Yang CY, Wu TH, Tseng CH, Yen FL. Myricetin Nanofibers Enhanced Water Solubility and Skin Penetration for Increasing Antioxidant and Photoprotective Activities. Pharmaceutics 2023; 15:pharmaceutics15030906. [PMID: 36986766 PMCID: PMC10058934 DOI: 10.3390/pharmaceutics15030906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/14/2023] Open
Abstract
Excessive exposure to ultraviolet radiation (UV) can induce oxidative stress through the over-production of reactive oxygen species (ROS) on the skin. Myricetin (MYR), a natural flavonoid compound, significantly inhibited UV-induced keratinocyte damage; however, its bioavailability is limited by its poor water solubility and inefficient skin penetration ability, which subsequently influences its biological activity. The purpose of the study was to develop a myricetin nanofibers (MyNF) system of hydroxypropyl-β-cyclodextrin (HPBCD)/polyvinylpyrrolidone K120 (PVP)-loaded with MYR that would enhance the water solubility and skin penetration by changing the physicochemical characteristics of MYR, including reducing the particle size, increasing the specific surface area, and amorphous transformation. The results also revealed that the MyNF can reduce cytotoxicity in HaCaT keratinocytes when compared with MYR; additionally, MyNF had better antioxidant and photoprotective activity than raw MYR for the UVB-induced HaCaT keratinocytes damage model due to the MyNF increased water solubility and permeability. In conclusion, our results demonstrate that MyNF is a safe, photostable, and thermostable topical ingredient of antioxidant nanofibers to enhance the skin penetration of MYR and prevent UVB-induced skin damage.
Collapse
Affiliation(s)
- Tzu-Ching Lin
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
| | - Chun-Yin Yang
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
| | - Tzu-Hui Wu
- Department of Pharmacy and Master Program, Collage of Pharmacy and Health Care, Tajen University, Pingtung County 90741, Taiwan
| | - Chih-Hua Tseng
- School of Pharmacy, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City 807, Taiwan
- Department of Pharmacy, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung City 801, Taiwan
- Correspondence: (C.-H.T.); (F.-L.Y.); Tel.: +886-7-312-1101 (ext. 2163) (C.-H.T.); +886-7-312-1101 (ext. 2028) (F.-L.Y.)
| | - Feng-Lin Yen
- Drug Development and Value Creation Research Center, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Department of Fragrance and Cosmetic Science, College of Pharmacy, Kaohsiung Medical University, Kaohsiung City 807, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung City 807, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung City 804, Taiwan
- College of Professional Studies, National Pingtung University of Science and Technology, Pingtung County 912, Taiwan
- Correspondence: (C.-H.T.); (F.-L.Y.); Tel.: +886-7-312-1101 (ext. 2163) (C.-H.T.); +886-7-312-1101 (ext. 2028) (F.-L.Y.)
| |
Collapse
|
16
|
Self-Assembled Lecithin-Chitosan Nanoparticles Improved Rotigotine Nose-to-Brain Delivery and Brain Targeting Efficiency. Pharmaceutics 2023; 15:pharmaceutics15030851. [PMID: 36986712 PMCID: PMC10052746 DOI: 10.3390/pharmaceutics15030851] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 02/11/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Rotigotine (RTG) is a non-ergoline dopamine agonist and an approved drug for treating Parkinson’s disease. However, its clinical use is limited due to various problems, viz. poor oral bioavailability (<1%), low aqueous solubility, and extensive first-pass metabolism. In this study, rotigotine-loaded lecithin-chitosan nanoparticles (RTG-LCNP) were formulated to enhance its nose-to-brain delivery. RTG-LCNP was prepared by self-assembly of chitosan and lecithin due to ionic interactions. The optimized RTG-LCNP had an average diameter of 108 nm with 14.43 ± 2.77% drug loading. RTG-LCNP exhibited spherical morphology and good storage stability. Intranasal RTG-LCNP improved the brain availability of RTG by 7.86 fold with a 3.84-fold increase in the peak brain drug concentration (Cmax(brain)) compared to intranasal drug suspensions. Further, the intranasal RTG-LCNP significantly reduced the peak plasma drug concentration (Cmax(plasma)) compared to intranasal RTG suspensions. The direct drug transport percentage (DTP (%)) of optimized RTG-LCNP was found to be 97.3%, which shows effective direct nose-to-brain drug uptake and good targeting efficiency. In conclusion, RTG-LCNP enhanced drug brain availability, showing the potential for clinical application.
Collapse
|
17
|
Thambiliyagodage C, Jayanetti M, Mendis A, Ekanayake G, Liyanaarachchi H, Vigneswaran S. Recent Advances in Chitosan-Based Applications-A Review. MATERIALS (BASEL, SWITZERLAND) 2023; 16:2073. [PMID: 36903188 PMCID: PMC10004736 DOI: 10.3390/ma16052073] [Citation(s) in RCA: 60] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 02/24/2023] [Accepted: 03/01/2023] [Indexed: 05/31/2023]
Abstract
Chitosan derived from chitin gas gathered much interest as a biopolymer due to its known and possible broad applications. Chitin is a nitrogen-enriched polymer abundantly present in the exoskeletons of arthropods, cell walls of fungi, green algae, and microorganisms, radulae and beaks of molluscs and cephalopods, etc. Chitosan is a promising candidate for a wide variety of applications due to its macromolecular structure and its unique biological and physiological properties, including solubility, biocompatibility, biodegradability, and reactivity. Chitosan and its derivatives have been known to be applicable in medicine, pharmaceuticals, food, cosmetics, agriculture, the textile and paper industries, the energy industry, and industrial sustainability. More specifically, their use in drug delivery, dentistry, ophthalmology, wound dressing, cell encapsulation, bioimaging, tissue engineering, food packaging, gelling and coating, food additives and preservatives, active biopolymeric nanofilms, nutraceuticals, skin and hair care, preventing abiotic stress in flora, increasing water availability in plants, controlled release fertilizers, dye-sensitised solar cells, wastewater and sludge treatment, and metal extraction. The merits and demerits associated with the use of chitosan derivatives in the above applications are elucidated, and finally, the key challenges and future perspectives are discussed in detail.
Collapse
Affiliation(s)
- Charitha Thambiliyagodage
- Faculty of Humanities and Sciences, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Madara Jayanetti
- Faculty of Humanities and Sciences, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Amavin Mendis
- Faculty of Humanities and Sciences, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Geethma Ekanayake
- Faculty of Humanities and Sciences, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Heshan Liyanaarachchi
- Faculty of Humanities and Sciences, Sri Lanka Institute of Information Technology, Malabe 10115, Sri Lanka
| | - Saravanamuthu Vigneswaran
- Faculty of Engineering and Information Technology, University of Technology Sydney, P.O. Box 123, Broadway, NSW 2007, Australia
- Faculty of Sciences & Technology (RealTek), Norwegian University of Life Sciences, P.O. Box 5003, N-1432 Ås, Norway
| |
Collapse
|
18
|
Srivastava S, Mathew J, Pandey AC. Baicalein—A review on its molecular mechanism against breast cancer and delivery strategies. Med Chem Res 2023. [DOI: 10.1007/s00044-023-03037-7] [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]
|
19
|
Wang Q, Li M, Sun X, Chen N, Yao S, Feng X, Chen Y. ZIF-8 integrated with polydopamine coating as a novel nano-platform for skin-specific drug delivery. J Mater Chem B 2023; 11:1782-1797. [PMID: 36727421 DOI: 10.1039/d2tb02361j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Metal-organic frameworks (MOFs) are highly promising as a novel class of drug delivery carriers; however, there are few reports about their application in nanoparticle-based formulations for dermal administration. In this work, we developed a novel kind of nanoparticular system based on zeolitic imidazolate framework-8 (ZIF-8) and polydopamine (PDA) modification for improving the dermal delivery of 5-fluorouracil (5-FU). The structures and properties of the prepared nanoparticles were characterized using a variety of analytical methods. Their ex vivo delivery performance in the skin was investigated using Franz cells, and the underlying mechanisms were studied via confocal laser scanning microscopy (CLSM) and hematoxylin-eosin (HE) experiments which were employed to probe the penetration pathway and the interaction between nanoparticles and the skin. The results revealed that both 5-FU@ZIF-8 and ZIF-8@5-FU@PDA had an enhancement effect on the deposition of 5-FU in the skin, and the surface coating of PDA could further reduce drug permeation across the skin, especially in the case of impaired skin, in comparison with the drug solution. The CLSM study using rhodamine 6G as the fluorescent probe to mimic 5-FU indicated that ZIF-8 and ZIF-8@PDA could deliver their payloads into the skin via two pathways, i.e., intercellular and follicular ones, and the follicular route was shown to be particularly important for ZIF-8@PDA, in which the drug and carrier were co-delivered into the skin as an intact particle. This study provides evidence for using ZIF-8 and PDA modification for skin-specific drug delivery and offers an effective avenue to develop novel nanoplatforms for dermal application to treat skin diseases.
Collapse
Affiliation(s)
- Qiuyue Wang
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| | - Mingming Li
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| | - Xinxing Sun
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| | - Naiying Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| | - Sicheng Yao
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| | - Xun Feng
- Department of Sanitary Chemistry, School of Public Health, Shenyang Medical College, No. 146 Yellow River North Street, Shenyang, 110034, China
| | - Yang Chen
- Department of Pharmaceutics, School of Pharmacy, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, China.
| |
Collapse
|
20
|
Wang Y, Pu M, Yan J, Zhang J, Wei H, Yu L, Yan X, He Z. 1,2-Bis(2-aminophenoxy)ethane- N, N, N', N'-tetraacetic Acid Acetoxymethyl Ester Loaded Reactive Oxygen Species Responsive Hyaluronic Acid-Bilirubin Nanoparticles for Acute Kidney Injury Therapy via Alleviating Calcium Overload Mediated Endoplasmic Reticulum Stress. ACS NANO 2023; 17:472-491. [PMID: 36574627 DOI: 10.1021/acsnano.2c08982] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Calcium overload is one of the early determinants of the core cellular events that contribute to the pathogenesis of acute kidney injury (AKI), which include oxidative stress, ATP depletion, calcium overload, and inflammatory response with self-amplifying and interactive feedback loops that ultimately lead to cellular injury and renal failure. Excluding adjuvant therapy, there are currently no approved pharmacotherapies for the treatment of AKI. Using an adipic dihydride linker, we modified the hyaluronic acid polymer chain with a potent antioxidant, bilirubin, to produce an amphiphilic conjugate. Subsequently, we developed a kidney-targeted and reactive oxygen species (ROS)-responsive drug delivery system based on the flash nanocomplexation method to deliver a well-known intracellular calcium chelator, 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (BAPTA-AM, BA), with the goal of rescuing renal cell damage via rapidly scavenging of intracellularly overloaded Ca2+. In the ischemia-reperfusion (I/R) induced AKI rat model, a single dose of as-prepared formulation (BA 100 μg·kg-1) 6 h post-reperfusion significantly reduced renal function indicators by more than 60% within 12 h, significantly alleviated tissular pathological changes, ameliorated tissular oxidative damage, significantly inhibited apoptosis of renal tubular cells and the expression of renal tubular marker kidney injury molecule 1, etc., thus greatly reducing the risk of kidney failure. Mechanistically, the treatment with BA-loaded NPs significantly inhibited the activation of the ER stress cascade response (IRE1-TRAF2-JNK, ATF4-CHOP, and ATF6 axis) and regulated the downstream apoptosis-related pathway while also reducing the inflammatory response. The BA-loaded NPs hold great promise as a potential therapy for I/R injury-related diseases.
Collapse
Affiliation(s)
- Yanan Wang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Minju Pu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Jiahui Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Jingwen Zhang
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Huichao Wei
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Liangmin Yu
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Xuefeng Yan
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| | - Zhiyu He
- Frontiers Science Center for Deep Ocean Multispheres and Earth Systems, Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, Ocean University of China, Qingdao266003, China
- College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao266003, China
| |
Collapse
|
21
|
Ye J, Li R, Cheng J, Liu D, Yang Y, Wang H, Xu X, Li L, Ma P, Liu Y. Comparative Colloidal Stability of Commercial Amphotericin B Nanoformulations Using Dynamic and Static Multiple Light Scattering Techniques. Int J Nanomedicine 2022; 17:6047-6064. [PMID: 36510621 PMCID: PMC9740024 DOI: 10.2147/ijn.s387681] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/30/2022] [Indexed: 12/12/2022] Open
Abstract
Background Amphotericin B (AmB) nanoformulations have been widely used for the treatment of invasive fungal infections in clinical practice, all of which are lyophilized solid dosage forms that improve storage stability. The colloidal stability of reconstituted lyophilized nanoparticles in an injection medium is a critical quality attribute that directly affects their safety and efficacy during clinical use. Methods In the present study, the colloidal stability of commercial AmB nanoformulations, including AmB cholesteryl sulfate complex (AmB-CSC) and AmB liposome (AmB-Lipo), was evaluated using the dynamic (DLS) and static multiple light scattering (SMLS) techniques. Results Compared to the DLS technique, the SMLS technique allows for a more objective and accurate evaluation of the colloidal stability of AmB nanoformulations. The results obtained using the SMLS technique demonstrated that AmB-CSC and AmB-Lipo exhibited excellent colloidal stability in both sterile water and 5% dextrose injection. The disk-like structure of the AmB-CSC nanoparticles more readily adsorbed serum proteins to form protein corona compared to the spherical structure of AmB-Lipo after incubation with serum. Additionally, AmB-CSC and AmB-Lipo can significantly reduce the in vitro cytotoxicity and in vivo nephrotoxicity of AmB, which may be attributed to the good colloidal stability and the improved pharmacokinetic profiles of AmB nanoformulations. Conclusion To the best of our knowledge, this study is the first to compare the colloidal stability of commercial AmB nanoformulations. These findings will provide useful information not only to inform the clinical use of available AmB nanoformulations but also for improving the design and conduct of translational research on novel AmB nanomedicines.
Collapse
Affiliation(s)
- Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Renjie Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Jialing Cheng
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Dongdong Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Xiaoyan Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| | - Panpan Ma
- Beijing Union Second Pharmaceutical Factory, Beijing, People’s Republic of China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, People’s Republic of China
| |
Collapse
|
22
|
Ye J, Li L, Yin J, Wang H, Li R, Yang Y, Guan Y, Xia X, Liu Y. Tumor-targeting intravenous lipid emulsion of paclitaxel: Characteristics, stability, toxicity, and toxicokinetics. J Pharm Anal 2022; 12:901-912. [PMID: 36605580 PMCID: PMC9805944 DOI: 10.1016/j.jpha.2022.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 07/28/2022] [Accepted: 08/17/2022] [Indexed: 11/07/2022] Open
Abstract
Lipid nanoemulsions are promising nanodrug delivery carriers that can improve the efficacy and safety of paclitaxel (PTX). However, no intravenous lipid emulsion of PTX has been approved for clinical treatment, and systemic safety profiles have not yet been reported. Here we outline the development of a PTX-loaded tumor-targeting intravenous lipid emulsion (PTX Emul) and describe its characteristics, colloidal stability, and systemic safety profiles in terms of acute toxicity, long-term toxicity, and toxicokinetics. We also compare PTX Emul with conventional PTX injection. Results showed that PTX Emul exhibited an ideal average particle size (approximately 160 nm) with narrow size distribution and robust colloidal stability under different conditions. Hypersensitivity reaction and hemolysis tests revealed that PTX Emul did not induce hypersensitivity reactions and had no hemolytic potential. In addition, where the alleviated systemic toxicity of PTX Emul may be attributed to the altered toxicokinetic characteristics in beagle dogs, including the decreased AUC and increased plasma clearance and volume of distribution, PTX Emul alleviated acute and long-term toxicity as evidenced by the enhanced the median lethal dose and approximate lethal dose, moderate body weight change, decreased bone marrow suppression and organ toxicity compared with those under PTX injection at the same dose. A fundamental understanding of the systemic safety profiles, high tumor-targeting efficiency, and superior antitumor activity in vivo of PTX Emul can provide powerful evidence of its therapeutic potential as a future treatment for breast cancer.
Collapse
Affiliation(s)
- Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Jiye Yin
- National Beijing Center for Drug Safety Evaluation and Research, Beijing Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Renjie Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China
| | - Yongbiao Guan
- National Beijing Center for Drug Safety Evaluation and Research, Beijing Institute of Pharmacology and Toxicology, Academy of Military Medical Sciences, Beijing, 100850, China,Corresponding author.
| | - Xuejun Xia
- Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Corresponding author.
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China,Corresponding author.
| |
Collapse
|
23
|
Talib WH, Abuawad A, Thiab S, Alshweiat A, Mahmod AI. Flavonoid-based nanomedicines to target tumor microenvironment. OPENNANO 2022. [DOI: 10.1016/j.onano.2022.100081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
24
|
Khan MM, Zaidi SS, Siyal FJ, Khan SU, Ishrat G, Batool S, Mustapha O, Khan S, Din FU. Statistical optimization of co-loaded rifampicin and pentamidine polymeric nanoparticles for the treatment of cutaneous leishmaniasis. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.104005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
25
|
Acceleration of Wound Healing in Rats by Modified Lignocellulose Based Sponge Containing Pentoxifylline Loaded Lecithin/Chitosan Nanoparticles. Gels 2022; 8:gels8100658. [PMID: 36286159 PMCID: PMC9601342 DOI: 10.3390/gels8100658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/12/2022] [Accepted: 10/13/2022] [Indexed: 11/04/2022] Open
Abstract
Dressing wounds accelerates the re-epithelialization process and changes the inflammatory environment towards healing. In the current study, a lignocellulose sponge containing pentoxifylline (PTX)-loaded lecithin/chitosan nanoparticles (LCNs) was developed to enhance the wound healing rate. Lecithin/chitosan nanoparticles were obtained by the solvent-injection method and characterized in terms of morphology, particle size distribution, and zeta potential. The lignocellulose hydrogels were functionalized through oxidation/amination and freeze-dried to obtain sponges. The prepared sponge was then loaded with LCNs/PTX to control drug release. The nanoparticle containing sponges were characterized using FTIR and SEM analysis. The drug release study from both nanoparticles and sponges was performed in PBS at 37 °C at different time points. The results demonstrated that PTX has sustained release from lignocellulose hydrogels. The wound healing was examined using a standard rat model. The results exhibited that PTX loaded hydrogels could achieve significantly accelerated and enhanced healing compared to the drug free hydrogels and the normal saline treatment. Histological examination of the healed skin confirmed the visual observations. Overall speaking, the in vivo assessment of the developed sponge asserts its suitability as wound dressing for treatment of chronic skin wounds.
Collapse
|
26
|
Development and Evaluation of a Novel Diammonium Glycyrrhizinate Phytosome for Nasal Vaccination. Pharmaceutics 2022; 14:pharmaceutics14102000. [PMID: 36297436 PMCID: PMC9612344 DOI: 10.3390/pharmaceutics14102000] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 09/14/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
The objective of the present research was to formulate diammonium glycyrrhizinate (DG) into phytosomes (DG-P) to induce nasal immune responses and enhance absorption. Plackett- Burman design was used for process optimization, incorporating specific formulation and process variables to obtain the optimal parameters. Fourier transform infrared spectroscopy (FTIR), X-ray power diffraction (P-XRD), and transmission electron microscopy (TEM) were used for characterization. The adjuvant activity of the DG-P was evaluated by using bone marrow dendritic cells. In vitro nasal mucosal permeation and in situ nasal perfusion were also investigated to evaluate nasal absorption. The DG phytosomes were in the size range of 20~30 nm and zeta-potential range of −30~−40 mV. DG-P demonstrated 4.2-fold increased solubility in n-octanol. Coculturing bone marrow dendritic cells with DG-P led to enhanced dendritic cell maturation. Apparent permeability coefficient of the phytosomal formulation was almost four times higher than that of free DG determined by ex vivo permeation studies on excised porcine mucosa. In situ nasal perfusion studies in rats demonstrated that the nasal absorption of DG-P was significantly higher than that of free DG. Conclusively, the results confirmed that DG-P have potential for use as an adjuvant for nasal vaccine.
Collapse
|
27
|
Tang Q, Yi Y, Chen Y, Zhuang Z, Wang F, Zhang L, Wei S, Zhang Y, Wang Y, Liu L, Liu Q, Jiang C. A green and highly efficient method to deliver hydrophilic polyphenols of Salvia miltiorrhiza and Carthamus tinctorius for enhanced anti-atherosclerotic effect via metal-phenolic network. Colloids Surf B Biointerfaces 2022; 215:112511. [PMID: 35483256 DOI: 10.1016/j.colsurfb.2022.112511] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/05/2022] [Accepted: 04/17/2022] [Indexed: 10/18/2022]
Abstract
Salvia miltiorrhiza and Carthamus tinctorius are traditional Chinese medicines that have been widely used for the treatment of cardiovascular disease. Salvianic acid A (SAA), salvianic acid B (SAB), protocatechuic aldehyde (PCA) and hydroxysafflor yellow A (HSYA) are the major hydrophilic polyphenols of Salvia miltiorrhiza and Carthamus tinctorius, all of which have been documented as active compounds for the prevention and treatment of atherosclerosis (AS). However, high aqueous solubility, low permeability and poor stability properties of the four hydrophilic polyphenols might influence their bioavailability and thus hinder their clinical potential. In this work, we introduced a green and highly efficient method for the efficient delivery of the four hydrophilic components via metal-phenolic network. The four coordination polymers of SAA, SAB, PCA and HSYA were successfully fabricated, and confirmed by UV-vis, FTIR, XPS, ICP-MS and dynamic light scattering analysis. We found all of them displayed potent antioxidant activity, good biocompatibility and stability. Impressively, the four coordination polymers showed remarkably enhanced anti-atherosclerotic effect compared with free drugs. Collectively, metal-phenolic network-based coordination polymer might show great potential for safe and efficient delivery of the hydrophilic polyphenols of Salvia miltiorrhiza and Carthamus tinctorius for anti-atherosclerotic therapy.
Collapse
Affiliation(s)
- Qingfa Tang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Yankui Yi
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Yao Chen
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Ziming Zhuang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Feng Wang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Lu Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Shenkun Wei
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Yusheng Zhang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Yueqiusha Wang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Li Liu
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China
| | - Qiang Liu
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China.
| | - Cuiping Jiang
- Guangdong Provincial Key Laboratory of Chinese Medicine Pharmaceutics, School of Traditional Chinese Medicine, Southern Medical University, Guangzhou Avenue North 1838, Guangzhou 510515, PR China; Zhujiang Hospital, Southern Medical University, 253 Gongye Middle Avenue, Haizhu District, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
28
|
Li J, Xiang H, Zhang Q, Miao X. Polysaccharide-Based Transdermal Drug Delivery. Pharmaceuticals (Basel) 2022; 15:ph15050602. [PMID: 35631428 PMCID: PMC9146969 DOI: 10.3390/ph15050602] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/04/2022] Open
Abstract
Materials derived from natural plants and animals have great potential for transdermal drug delivery. Polysaccharides are widely derived from marine, herbal, and microbial sources. Compared with synthetic polymers, polysaccharides have the advantages of non-toxicity and biodegradability, ease of modification, biocompatibility, targeting, and antibacterial properties. Currently, polysaccharide-based transdermal drug delivery vehicles, such as hydrogel, film, microneedle (MN), and tissue scaffolds are being developed. The addition of polysaccharides allows these vehicles to exhibit better-swelling properties, mechanical strength, tensile strength, etc. Due to the stratum corneum’s resistance, the transdermal drug delivery system cannot deliver drugs as efficiently as desired. The charge and hydration of polysaccharides allow them to react with the skin and promote drug penetration. In addition, polysaccharide-based nanotechnology enhances drug utilization efficiency. Various diseases are currently treated by polysaccharide-based transdermal drug delivery devices and exhibit promising futures. The most current knowledge on these excellent materials will be thoroughly discussed by reviewing polysaccharide-based transdermal drug delivery strategies.
Collapse
Affiliation(s)
- Jingyuan Li
- Marine College, Shandong University, Weihai 264209, China; (J.L.); (H.X.); (Q.Z.)
- SDU-ANU Joint Science College, Shandong University, Weihai 264209, China
| | - Hong Xiang
- Marine College, Shandong University, Weihai 264209, China; (J.L.); (H.X.); (Q.Z.)
| | - Qian Zhang
- Marine College, Shandong University, Weihai 264209, China; (J.L.); (H.X.); (Q.Z.)
| | - Xiaoqing Miao
- Marine College, Shandong University, Weihai 264209, China; (J.L.); (H.X.); (Q.Z.)
- Weihai Changqing Ocean Science Technology Co., Ltd., Weihai 264209, China
- Correspondence: ; Tel.: +86-19806301068
| |
Collapse
|
29
|
Manna S, Jana S. Marine Polysaccharides in Tailor- Made Drug Delivery. Curr Pharm Des 2022; 28:1046-1066. [DOI: 10.2174/1381612828666220328122539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 02/11/2022] [Indexed: 01/09/2023]
Abstract
Abstract:
Marine sources have attracted much interest as an emerging source of biomaterials in drug delivery applications. Amongst all other marine biopolymers, polysaccharides have been the mostly investigated class of biomaterials. The low cytotoxic behavior, in combination with the newly explored health benefits of marine polysaccharides has made it one of the prime research areas in the pharmaceutical and biomedical fields. In this review, we focused on all available marine polysaccharides, including their classification based on biological sources. The applications of several marine polysaccharides in recent years for tissue-specific novel drug delivery including gastrointestinal, brain tissue, transdermal, ocular, liver, and lung have also been discussed here. The abundant availability in nature, cost-effective extraction, and purification process along with a favorable biodegradable profile will encourage researchers to continue investigating marine polysaccharides for exploring newer applications in targeting specific delivery of therapeutics.
Collapse
Affiliation(s)
- Sreejan Manna
- Department of Pharmaceutical Technology, Brainware University, Barasat, Kolkata, West Bengal -700125, India
| | - Sougata Jana
- Department of Pharmaceutics, Gupta College of Technological Sciences, Ashram More, G.T. Road, Asansol-713301, West Bengal, India
- Department of Health and Family Welfare, Directorate of Health Services, Kolkata, India
| |
Collapse
|
30
|
Zarei B, Tabrizi MH, Rahmati A. PEGylated Lecithin-Chitosan Nanoparticle-Encapsulated Alphα-Terpineol for In Vitro Anticancer Effects. AAPS PharmSciTech 2022; 23:94. [PMID: 35314914 DOI: 10.1208/s12249-022-02245-5] [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: 11/30/2021] [Accepted: 02/28/2022] [Indexed: 12/11/2022] Open
Abstract
The aim of this study was to fabrication PEGylated lecithin-chitosan nanoparticles (PLC-NPs) as alphα-Terpineol's (αT-PLC-NPs) delivery system and examine its anti-cancer effects. αT-PLC-NPs were synthesized by self-assembling method; after characterization, entrapment efficiency of α-T was measured by HPLC procedure. MTT test was conducted for cytotoxicity evaluation. Chick chorioallantoic membrane (CAM) and quantitative polymerase chain reaction (qPCR) analysis were used to determine the angiogenesis properties, and qPCR, flow cytometry, and acridine orange and propidium iodide (AO/PI) staining were used to evaluate the pro-apoptotic effects of αT-PLC-NPs. Finally, the anti-inflammatory and antibacterial activity of the αT-PLC-NPs was also evaluated. αT-PLC-NPs with a size of 220.8 nm, polydispersity index (PDI) of 0.3, zeta potential of +29.03 mV, and encapsulation efficiency of 82% showed higher inhibitory effect on MCF7 cells (IC50: 750 μg/mL) compared to HFF cells (above 1000 μg/mL). Decreased angiogenesis indices and embryonic growth factors in CAM assay, decreased expression of VEGF and VEGF-R genes, and decreased cell migration showed the inhibitory effect of αT-PLC-NPs on angiogenesis. Increased expression of P53, P21, and caspase9 genes, as well as the results of AO/PI staining along with increasing the number of SubG1 phase cells in flow cytometry, confirmed the pro-apoptotic effects of αT-PLC-NPs. Also, its anti-inflammatory effects were demonstrated by inhibiting the expression of pro-inflammatory cytokines (TNF-α and IL-6). The inhibitory power of αT-PLC-NPs in suppressing gram-positive and negative bacterial strains was demonstrated by disk diffusion (DD), minimum inhibitory concentration (MIC), and minimum bactericidal concentration (MBC) methods. PLC-NPs are a promising carrier for α-T transfer for preclinical studies.
Collapse
|
31
|
Maher Zahran E, Mohamad SA, Yahia R, Badawi AM, Sayed AM, Ramadan Abdelmohsen U. Anti-otomycotic potential of nanoparticles of Moringa oleifera leaf extract: an integrated in vitro, in silico and phase 0 clinical study. Food Funct 2022; 13:11083-11096. [DOI: 10.1039/d2fo02382b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The work flow of the study.
Collapse
Affiliation(s)
- Eman Maher Zahran
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, Universities Zone, 61111 New Minia City, Egypt
| | - Soad A. Mohamad
- Department of Pharmaceutics and Clinical pharmacy, Faculty of Pharmacy, Deraya University, Universities Zone, 61111 New Minia City, Egypt
| | - Ramadan Yahia
- Department of Microbiology, Faculty of Pharmacy, Deraya University, Universities Zone, 61111 New Minia City, Egypt
| | - Ahmed M. Badawi
- Department of Otorhinolaryngology, Faculty of Medicine, Minia University, Egypt
| | - Ahmed M. Sayed
- Department of Pharmacognosy, Faculty of Pharmacy, Nahda University, 62513 Beni-Suef, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, AlMaaqal University, 61014 Basra, Iraq
| | - Usama Ramadan Abdelmohsen
- Department of Pharmacognosy, Faculty of Pharmacy, Deraya University, Universities Zone, 61111 New Minia City, Egypt
- Department of Pharmacognosy, Faculty of Pharmacy, Minia University, 61519Minia, Egypt
| |
Collapse
|
32
|
Despotopoulou D, Lagopati N, Pispas S, Gazouli M, Demetzos C, Pippa N. The technology of transdermal delivery nanosystems: from design and development to preclinical studies. Int J Pharm 2021; 611:121290. [PMID: 34788674 DOI: 10.1016/j.ijpharm.2021.121290] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/18/2022]
Abstract
Transdermal administration has gained much attention due to the remarkable advantages such as patient compliance, drug escape from first-pass elimination, favorable pharmacokinetic profile and prolonged release properties. However, the major limitation of these systems is the limited skin penetration of the stratum corneum, the skin's most important barrier, which protects the body from the insertion of substances from the environment. Transdermal drug delivery systems are aiming to the disruption of the stratum corneum in order for the active pharmaceutical ingredients to enter successfully the circulation. Therefore, nanoparticles are holding a great promise because they can act as effective penetration enhancers due to their small size and other physicochemical properties that will be analyzed thoroughly in this report. Apart from the investigation of the physicochemical parameters, a comparison between the different types of nanoparticles will be performed. The complexity of skin anatomy and the unclear mechanisms of penetration should be taken into consideration to reach some realistic conclusions regarding the way that the described parameters affect the skin permeability. To the best of the authors knowledge, this is among the few reports on the literature describing the technology of transdermal delivery systems and how this technology affects the biological activity.
Collapse
Affiliation(s)
- Despoina Despotopoulou
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Panepistimioupolis Zografou 15771, National and Kapodistrian University of Athens, Athens, Greece
| | - Nefeli Lagopati
- Department of Histology and Embryology, Medical School, National Kapodistrian University of Athens, Greece
| | - Stergios Pispas
- Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece
| | - Maria Gazouli
- Department of Basic Medical Science, Laboratory of Biology, School of Medicine National and Kapodistrian University of Athens, Greece
| | - Costas Demetzos
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Panepistimioupolis Zografou 15771, National and Kapodistrian University of Athens, Athens, Greece
| | - Natassa Pippa
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Panepistimioupolis Zografou 15771, National and Kapodistrian University of Athens, Athens, Greece; Theoretical and Physical Chemistry Institute, National Hellenic Research Foundation, 48 Vassileos Constantinou Avenue, 11635 Athens, Greece.
| |
Collapse
|
33
|
Abd-Rabou AA, Abdelaziz AM, Shaker OG, Ayeldeen G. Metformin-loaded lecithin nanoparticles induce colorectal cancer cytotoxicity via epigenetic modulation of noncoding RNAs. Mol Biol Rep 2021; 48:6805-6820. [PMID: 34468912 DOI: 10.1007/s11033-021-06680-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Colorectal cancer (CRC) is major aliment around the word, with a cumulative rate of mortality. Metformin (MT) was recently approved as anticancer drug against solid tumors, such as CRC. Resistance to MT therapy remains to be a challenging matter facing the development of possible anti-cancer strategy. To circumvent this problem, MT nano-encapsulation has been introduced to sensitize resistant cancer cells. The purpose of the current study is to explore the MT's aptitude encapsulated in lecithin (LC) and chitosan (CS) nanoparticles to inhibit CRC proliferation through modulations of long noncoding RNAs (lncRNAs), micro RNAs (miRNAs), and some biochemical markers. METHODS AND RESULTS Cytotoxic screenings of the newly synthesized MT-based regimens; MT, MT-LC NPs (NP1), MT-CS NPs (NP2), and MT-LC-CS NPs (NP3) against colorectal cancerous Caco-2 and HCT116 cell lines versus normal WI-38 cells were performed. The epigenetic mechanistic effects of these proposed regimens on lncRNAs and miRNAs were investigated. Additionally, some protein levels were assessed in CRC cells upon treatments; YKL-40, PPARγ, E-cadherin (ECN), and VEGF. We resulted that NP1 recorded the highest significant cytotoxic effect on CRC cells. HCT116 cells were more sensitive to the NP1 compared to Caco-2 cells. Intriguingly, it was suggested that NP1 tackled the CRC cells through down-regulation of the H19, HOTTIP, HULC, LINC00641, miR-200, miR-92a, miR-21, YKL-40, PPARγ, and VEGF expressions, as well as up-regulation of the miR-944 and ECN expressions. CONCLUSIONS We concluded that the NP1 can potentially be cytotoxic to CRC cells in-vitro by modulating noncoding RNA.
Collapse
Affiliation(s)
- Ahmed A Abd-Rabou
- Medical Research Division, Hormones Department, National Research Center, Dokki, Giza, 12622, Egypt.
- Stem Cell Lab., Centre of Excellence for Advanced Science, National Research Center, Dokki, Giza, 12622, Egypt.
| | - Ahmed M Abdelaziz
- Ahmed Mahr Teaching Hospital (AMTH), Cairo, Egypt
- Supplementary General Sciences, Future University, Cairo, Egypt
| | - Olfat G Shaker
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| | - Ghada Ayeldeen
- Medical Biochemistry and Molecular Biology Department, Faculty of Medicine, Cairo University, Cairo, Egypt
| |
Collapse
|
34
|
Ye J, Li R, Yang Y, Dong W, Wang Y, Wang H, Sun T, Li L, Shen Q, Qin C, Xu X, Liao H, Jin Y, Xia X, Liu Y. Comparative colloidal stability, antitumor efficacy, and immunosuppressive effect of commercial paclitaxel nanoformulations. J Nanobiotechnology 2021; 19:199. [PMID: 34225762 PMCID: PMC8256566 DOI: 10.1186/s12951-021-00946-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 06/24/2021] [Indexed: 12/20/2022] Open
Abstract
Background Standard chemotherapy with taxanes, such as paclitaxel (PTX), remains the mainstay of systemic treatment of triple-negative breast cancer. Nanotechnology-based formulations have gradually replaced PTX injection and are widely used in China. However, no studies have compared the colloidal stability, antitumor efficacy, and safety of commercial PTX nanoformulations. Additionally, the desire to evaluate preclinical antitumor efficacy in human-derived tumor cells led to the widespread application of immunodeficient mouse models that likely contributed to the neglect of nanomedicines-immune system interactions. The present study investigated the colloidal stability, antitumor efficacy and safety, and nanomedicines-host immune system interactions of PTX nanoformulations. A further comparative analysis was performed to evaluate the clinical potential. Results Compared with liposome, PTX emulsion and PTX nanoparticle exhibited favorable colloidal stability. PTX emulsion was superior in inducing apoptosis and had a more pronounced inhibitory effect on 4T1-tumor spheroids compared with PTX liposome and PTX nanoparticle. Although PTX emulsion exhibited superior in vitro antitumor effect, no significant differences in the in vivo antitumor efficacy were found among the three types of PTX nanoformulations in an immunocompetent orthotopic 4T1 murine triple-negative breast cancer model. All PTX nanoformulations at maximum tolerated dose (MTD) induced lymphopenia and immunosuppression, as evidenced by the reduction of T cell subpopulations and inhibition of the dendritic cells maturation. Conclusions The MTD PTX nanomedicines-induced lymphopenia and immunosuppression may weaken the lymphocyte-mediated antitumor cellular immune response and partly account for the lack of differences in the in vivo antitumor outcomes of PTX nanoformulations. Understanding of what impacts PTX nanomedicines has on the immune system may be critical to improve the design and conduct of translational research of PTX nanomedicines in monotherapy or combination therapy with immunotherapy. Graphic abstract ![]()
Collapse
Affiliation(s)
- Jun Ye
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Renjie Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Yanfang Yang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Wujun Dong
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Yujie Wang
- Beijing Wehand-Bio Pharmaceutical Co. Ltd., Beijing, 102600, People's Republic of China
| | - Hongliang Wang
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Tong Sun
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Lin Li
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Qiqi Shen
- Beijing Wehand-Bio Pharmaceutical Co. Ltd., Beijing, 102600, People's Republic of China
| | - Caiyun Qin
- Beijing Wehand-Bio Pharmaceutical Co. Ltd., Beijing, 102600, People's Republic of China
| | - Xiaoyan Xu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Hengfeng Liao
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Yiqun Jin
- Beijing Wehand-Bio Pharmaceutical Co. Ltd., Beijing, 102600, People's Republic of China
| | - Xuejun Xia
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China.,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China
| | - Yuling Liu
- State Key Laboratory of Bioactive Substance and Function of Natural Medicines, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, 1 Xiannongtan Street, Beijing, 100050, People's Republic of China. .,Beijing Key Laboratory of Drug Delivery Technology and Novel Formulation, Institute of Materia Medica, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100050, People's Republic of China.
| |
Collapse
|
35
|
Cai P, Lu Y, Yin Z, Wang X, Zhou X, Li Z. Baicalein ameliorates osteoporosis via AKT/FOXO1 signaling. Aging (Albany NY) 2021; 13:17370-17379. [PMID: 34198266 PMCID: PMC8312461 DOI: 10.18632/aging.203227] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Accepted: 05/18/2021] [Indexed: 01/08/2023]
Abstract
In this study, we used bioinformatics and an in vitro cellular model of glucocorticoid-induced osteoporosis to investigate mechanisms underlying the beneficial effects of baicalein (BN) against osteoporosis. STITCH database analysis revealed 30 BN-targeted genes, including AKT1, CCND1, MTOR, and PTEN. Functional enrichment analysis demonstrated that BN-targeted genes were enriched in 49 Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways. MIRWALK2.0 database analysis identified 110 enriched KEGG pathways related to osteoporosis. A Venn diagram demonstrated that 26 KEGG pathways were common between osteoporosis and BN-targeted genes. The top 5 common KEGG pathways were prostate cancer, bladder cancer, glioma, pathways in cancer, and melanoma. BN-targeted genes in the top 5 shared KEGG pathways were involved in PI3K-AKT, MAPK, p53, ErbB, and mTOR signaling pathways. In addition, glucocorticoid-induced osteoporosis in MC3T3-E1 cells was partially reversed by BN through inhibition of AKT, which, by upregulating FOXO1, enhanced expression of bone turnover markers (ALP, OCN, Runx2, and Col 1) and extracellular matrix mineralization. These findings demonstrate that BN suppresses osteoporosis via an AKT/FOXO1 signaling pathway.
Collapse
Affiliation(s)
- Pan Cai
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Yan Lu
- Department of Laboratory Medicine, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China
| | - Xiuhui Wang
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Xiaoxiao Zhou
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| | - Zhuokai Li
- Department of Orthopedics, Shanghai University of Medicine and Health Sciences Affiliated Zhoupu Hospital, Shanghai, China
| |
Collapse
|
36
|
Pramanik S, Sali V. Connecting the dots in drug delivery: A tour d'horizon of chitosan-based nanocarriers system. Int J Biol Macromol 2020; 169:103-121. [PMID: 33338522 DOI: 10.1016/j.ijbiomac.2020.12.083] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/26/2020] [Accepted: 12/11/2020] [Indexed: 01/09/2023]
Abstract
One of the most promising pharmaceutical research areas is developing advanced delivery systems for controlled and sustained drug release. The drug delivery system (DDS) can be designed to strengthen the pharmacological and therapeutic characteristics of different medicines. Natural polymers have resolved numerous commencing hurdles, which hindered the clinical implementation of traditional DDS. The naturally derived polymers furnish various advantages such as biodegradability, biocompatibility, inexpensiveness, easy availability, and biologically identifiable moieties, which endorse cellular activity in contrast to synthetic polymers. Among them, chitosan has recently been in the spotlight for devising safe and efficient DDSs due to its superior properties such as minimal toxicity, bio-adhesion, stability, biodegradability, and biocompatibility. The primary amino group in chitosan shows exceptional qualities such as the rate of drug release, anti-microbial properties, the ability to cross-link with various polymers, and macrophage activation. This review intends to provide a glimpse into different practical utilization of chitosan as a drug carrier. The first segment of the review will give cognizance into the source of extraction and chitosan's remarkable properties. Further, we have endeavored to provide recent literature pertaining to chitosan applications in various drug delivery systems via different administration routes along with current patented chitosan formulations.
Collapse
Affiliation(s)
- Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India; Department of Polymeric Medical Devices, Medical Devices Engineering, Biomedical Technology Wing, Sree Chitra Tirunal Institute for Medical Sciences & Technology, Thiruvananthapuram, Kerala 695011, India.
| | - Vaishnavi Sali
- C.U. Shah College of Pharmacy, SNDT Women's University, Sir Vithaldas Thakersay, Santacruz West, Juhu, Mumbai, Maharashtra 400049, India
| |
Collapse
|