1
|
Jiang X, Gong M, Jia Y, Adu-Frimpong M, Wang X, Hua Q, Li T, Li J, Pan P, Toreniyazov E, Yu J, Cao X, Wang Q, Xu X. Preparation, in vitro and in vivo evaluation and anti-renal injury effects of Niazimicin-loaded mixed polymeric micelles. J Pharm Sci 2025; 114:103703. [PMID: 39988296 DOI: 10.1016/j.xphs.2025.103703] [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: 11/13/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/25/2025]
Abstract
BACKGROUND Chronic Kidney Disease (CKD) has become one of the major life-threatening conditions. Moringa seeds have been reported to exhibit renoprotective effects, with Niazimicin as its characteristic component. OBJECTIVE To investigate the anti-renal injury effects of Niazimicin and its mixed micelles (N-M) that composed of monomethyl ether poly (ethylene glycol)-polycaprolactone (mPEG-PCL) and polyethylene glycolated chitosan (PEG-CS) on adenine-induced CKD mice. METHODS PEG-CS was prepared via formaldehyde linkage method. The thin film dispersion method was employed for the preparation of N-M before it was characterized in vivo and in vitro. The anti-renal injury effects were evaluated by analyzing the serum levels of creatinine (Cr), p-Cresol sulphate (pCs), indole sulphate (IS) and hematoxylin-eosin (HE)-stained sections of hepatic and renal pathological tissues in CKD mice. RESULTS The N-M were spherical micelles of uniform size and highly dispersed with particle size of 42.94 ± 0.58 nm, encapsulation efficiency (EE) of 97.73 ± 2.33% and drug loading (DL) of 16.17 ± 0.28%, as well as good stability, and a very low critical micelle concentration (CMC) value of 0.00731 mg/mL. The N-M had a delayed-release effect and higher oral bioavailability compared to Niazimicin. CONCLUSION In CKD mice, Niazimicin exhibited an anti-renal injury effect, while the renoprotective effect of N-M was superior to that of Niazimicin.
Collapse
Affiliation(s)
- Xia Jiang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Mingie Gong
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Yue Jia
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School Chemical and Biochemical Sciences, C.K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, UK, 0215-5321, Ghana
| | - Xiaowen Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Qinyang Hua
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Tingyuan Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Jiaying Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | - Pengfei Pan
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China
| | | | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| | - Xia Cao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| | - Qilong Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, Jiangsu, CN, PR China; Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, CN, PR China.
| |
Collapse
|
2
|
Nguyen TD, Khanal S, Lee E, Choi J, Bohara G, Rimal N, Choi DY, Park S. Astaxanthin-loaded brain-permeable liposomes for Parkinson's disease treatment via antioxidant and anti-inflammatory responses. J Nanobiotechnology 2025; 23:78. [PMID: 39905399 DOI: 10.1186/s12951-025-03104-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Parkinson's disease is a neurodegenerative disorder characterized by cardinal motor symptoms resulting from the loss of dopaminergic neurons in the substantia nigra pars compacta. Although current medications may alleviate its symptoms, Parkinson's disease remains incurable. Astaxanthin is an antioxidant and anti-inflammatory agent; however, its high susceptibility to oxidative degradation and low aqueous solubility limit its therapeutic efficacy. This study aimed to improve the pharmaceutical properties and neuroprotective effects of astaxanthin for Parkinson's disease treatment by using lactoferrin-conjugated astaxanthin-loaded liposomes (Lf-ASX-LPs). We successfully formulated Lf-ASX-LPs with high encapsulation efficiency (97.6%) and favorable physical characteristics (diameter: 109.8 ± 1.1 nm; polydispersity index: 0.18 ± 0.01; zeta potential: - 9.5 ± 1.1 mV). Lf-functionalized liposomes demonstrated enhanced cellular uptake and permeation in a Transwell® study, showing a 16.7-fold higher internalization in SH-SY5Y cells over 24 h than those without Lf conjugation. Additionally, Lf functionalization enhanced brain penetration ability, as demonstrated by a biodistribution study using nude mice, compared to LPs without Lf conjugation. In vitro, Lf-ASX-LPs reduced cell loss by 20.1% and oxygen species by 30.0%, ameliorated the reduction in mitochondrial membrane potential under 1-methyl-4-phenylpyridinium-induced toxicity by 40.1%, and reduced extracellular nitric oxide levels under lipopolysaccharide-induced toxicity by 32.0%, indicating cytoprotective and antioxidant effects. In a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced Parkinson's disease mouse model, treatment with Lf-ASX-LPs resulted in 5.0-, 3.1-, and 5.6-fold increases in dopamine levels, TH+ fiber density, and TH+ neurons, respectively, restoring dopaminergic neuron damage. The developed formulation also alleviated behavioral impairment and neuroinflammation, reducing astrocyte and microglial activation in the striatum and substantia nigra of the MPTP-treated animals. Thus, our formulation of Lf-ASX-LPs represents a promising strategy for providing neuroprotection and retarding Parkinson's disease progression.
Collapse
Affiliation(s)
- Thai-Duong Nguyen
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Shristi Khanal
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Eunhee Lee
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Jinsol Choi
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, Republic of Korea
| | - Ganesh Bohara
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Nikesh Rimal
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea
| | - Dong-Young Choi
- College of Pharmacy, Yeungnam University, 280 Daehak-Ro, Gyeongsan, Gyeongbuk, 38541, Republic of Korea.
| | - Soyeun Park
- College of Pharmacy, Keimyung University, 1095 Dalgubeoldae-Ro, Dalseo-Gu, Daegu, 42601, Republic of Korea.
| |
Collapse
|
3
|
Koirala P, Bhattarai P, Sriprablom J, Zhang R, Nirmal S, Nirmal N. Recent progress of functional nano-chitosan in pharmaceutical and biomedical applications: An updated review. Int J Biol Macromol 2025; 285:138324. [PMID: 39638188 DOI: 10.1016/j.ijbiomac.2024.138324] [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: 07/17/2024] [Revised: 11/28/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Chitosan is a deacylated derivative of chitin, which is a naturally occurring polysaccharide found in the shells of crustaceans. Chitosan's biocompatibility, physicochemical and mechanical properties qualify it as an excellent candidate for biomedical and pharmaceutical applications. Furthermore, the nanoengineering of chitosan enhances its functional and desirable properties for various applications. Additional functionalization of nano-chitosan is possible using various crosslinkers via chemical modification of hydroxyl or amino groups chitosan. This advanced functionalized nano-chitosan enables drug stability, site-specific delivery, controlled release, and sustainable pharmacodynamic properties. It is also used as a regenerative medicine for wound healing, bone and dental repair, biosensing and disease detection, tissue engineering, etc. Therefore, this review discusses the functionalization of nano-chitosan. A brief discussion is provided on the applications of nano-chitosan in the pharmaceutical industry such as drug carriers, targeted delivery, and controlled release, as well as in the biomedical industry, including wound healing, bone, and dental repair. Additionally, the disease detection using nano-chitosan has been investigated. Lastly, future perspectives and concluding remarks are presented.
Collapse
Affiliation(s)
- Pankaj Koirala
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Prabina Bhattarai
- Department of Health and Human Development, Montana State University, Bozeman, MT, USA
| | - Jiratthitikan Sriprablom
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Run Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Queensland 4072, Australia
| | - Siddhesh Nirmal
- Maharashtra Institute of Technology, Chatrapati Sambhaji Nagar, Aurangabad, Maharashtra 431010, India
| | - Nilesh Nirmal
- Institute of Nutrition, Mahidol University, 999 Phutthamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand.
| |
Collapse
|
4
|
Žigrayová D, Mikušová V, Mikuš P. Advances in Chitosan Derivatives: Preparation, Properties and Applications in Pharmacy and Medicine. Gels 2024; 10:701. [PMID: 39590057 PMCID: PMC11593520 DOI: 10.3390/gels10110701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/21/2024] [Accepted: 10/25/2024] [Indexed: 11/28/2024] Open
Abstract
Chitosan (CS) derivatives have been extensively investigated to enhance the physicochemical and biological properties of CS, such as its solubility, biocompatibility, and bioactivity, which are required in various areas of pharmacy and medicine. The present work emphasizes the ongoing research and development in this field, suggesting that the further exploration of CS derivatives could lead to innovative solutions that benefit society. The physicochemical properties, biological activities, methods of preparation, advantages, limitations, intended application areas, and realized practical implementations of particular CS derivatives are summarized and discussed herein. Despite the numerous promising attributes of CS derivatives as reported in this paper, however, challenges like target selectivity, standardization (purity, chitosan structural variability), and cost-effectiveness still need addressing for widespread implementation, especially in drug delivery. Therefore, basic research studies still prevail in CS drug delivery systems. However, for specific applications such as wound healing and tissue engineering, implementations of CS derivatives in practice are found to be more frequent. To obtain a more complex view of the topic, information from the scientific papers reviewed is supplemented with information from actual patents and clinical studies. Both basic research advances and the most successful and important medical implementations of CS derivatives are discussed concerning further challenges and future perspectives.
Collapse
Affiliation(s)
- Dominika Žigrayová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia; (D.Ž.); (V.M.)
| | - Veronika Mikušová
- Department of Galenic Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia; (D.Ž.); (V.M.)
| | - Peter Mikuš
- Department of Pharmaceutical Analysis and Nuclear Pharmacy, Faculty of Pharmacy, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
- Faculty of Pharmacy, Toxicological and Antidoping Center, Comenius University Bratislava, Odbojárov 10, 83232 Bratislava, Slovakia
| |
Collapse
|
5
|
Xu X, Zhang X, Li R, Yang X, Fu P, Feng R, Sun X, Wang Z, Yu J, Cao X, Yu Q, Wang Q, Liu S, Yang X, Zhu Y, Shi W, Deng W. Platelet Membrane-Coated Curcumin-PLGA Nanoparticles Promote Astrocyte-Neuron Transdifferentiation for Intracerebral Hemorrhage Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2311128. [PMID: 38888124 DOI: 10.1002/smll.202311128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 03/14/2024] [Indexed: 06/20/2024]
Abstract
Intracerebral hemorrhage (ICH) is a hemorrhagic disease with high mortality and disability rates. Curcumin is a promising drug for ICH treatment due to its multiple biological activities, but its application is limited by its poor watersolubility and instability. Herein, platelet membrane-coated curcumin polylactic-co-glycolic acid (PLGA) nanoparticles (PCNPs) are prepared to achieve significantly improved solubility, stability, and sustained release of curcumin. Fourier transform infrared spectra and X-ray diffraction assays indicate good encapsulation of curcumin within nanoparticles. Moreover, it is revealed for the first time that curcumin-loaded nanoparticles can not only suppress hemin-induced astrocyte proliferation but also induce astrocytes into neuron-like cells in vitro. PCNPs are used to treat rat ICH by tail vein injection, using in situ administration as control. The results show that PCNPs are more effective than curcumin-PLGA nanoparticles in concentrating on hemorrhagic lesions, inhibiting inflammation, suppressing astrogliosis, promoting neurogenesis, and improving motor functions. The treatment efficacy of intravenously administered PCNPs is comparable to that of in situ administration, indicating a good targeting effect of PCNPs on the hemorrhage site. This study provides a potent treatment for hemorrhagic injuries and a promising solution for efficient delivery of water-insoluble drugs using composite materials of macromolecules and cell membranes.
Collapse
Affiliation(s)
- Ximing Xu
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Xinyu Zhang
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Ran Li
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Xiufen Yang
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Peng Fu
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Ruijie Feng
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Xuan Sun
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Zhe Wang
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Jiangnan Yu
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Xia Cao
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Qingtong Yu
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Qilong Wang
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Sitong Liu
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Xiaoxia Yang
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Yuan Zhu
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| | - Wentao Shi
- Central Laboratory, Gaochun Hospital Affiliated to Jiangsu University, Jiangsu University, Nanjing, Jiangsu Province, 211300, China
| | - Wenwen Deng
- School of Pharmacy, Jiangsu University, The International Institute on Natural Products and Stem Cells (iNPS), Key Lab for Drug Delivery & Tissue Regeneration, Jiangsu Provincial Research Center for Medicinal Function Development of New Food Resources, Zhenjiang, 202013, China
| |
Collapse
|
6
|
Khayyal MT, Teaima MH, Marzouk HM, -Hazek RME, Behnam F, Behnam D. Comparative Pharmacokinetic Study of Standard Astaxanthin and its Micellar Formulation in Healthy Male Volunteers. Eur J Drug Metab Pharmacokinet 2024; 49:467-475. [PMID: 38748358 PMCID: PMC11199261 DOI: 10.1007/s13318-024-00898-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 06/26/2024]
Abstract
BACKGROUND AND OBJECTIVE Astaxanthin is a naturally occurring carotenoid with high anti-oxidant properties, but it is a very lipophilic compound with low oral bioavailability. This study was conducted to compare the pharmacokinetic parameters of a novel astaxanthin preparation based on micellar solubilization technology, NovaSOL® 400-mg capsules (Test product), and those of astaxanthin 400-mg capsules (reference product), after single oral dose administration to healthy male adults. METHODS A single oral dose (400 mg equivalent to 8 mg astaxanthin) of test and reference astaxanthin were administered with 240 mL of water to 12 volunteers according to crossover design, in two phases, with a washout period of 1 week in between. Blood samples were collected at hourly intervals for the first 12 h, then at 24.0, 48.0, and 72.0 h after administration. Aliquots of plasma were centrifuged and the clear supernatant was injected into the high performance liquid chromatography-diode array detection (HPLC-DAD) system. Plasma concentration of astaxanthin versus time profiles were constructed, and the primary pharmacokinetic parameters, maximum concentration (Cmax), area under concentration time curve from time of administration (0) to time (t) [AUC0-t] or to infinity ∞, [AUC0-∞], half-life (T½) and time to reach Cmax (Tmax) were calculated. RESULTS The test micellar astaxanthin reached a Cmax of 7.21 µg/ml after 3.67 h compared to only 3.86 µg/ml after 8.5 h for the reference native astaxanthin. CONCLUSION Micellar formulation of astaxanthin is capable of producing a high concentration of astaxanthin in plasma in a shorter time, thereby expected to provide faster potential therapeutic efficacy.
Collapse
Affiliation(s)
- Mohamed T Khayyal
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Mahmoud H Teaima
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Center of Applied Research and advanced Studies (CARAS), Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hoda M Marzouk
- Department of Pharmaceutical Analytical Chemistry, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, ET-11562, Egypt
- Center of Applied Research and advanced Studies (CARAS), Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Rania M El -Hazek
- National Center for Radiation Research and Technology, Atomic Energy Authority, Cairo, Egypt
| | | | | |
Collapse
|
7
|
Khan MKA, Abdulhameed AS, Alshahrani H, Algburi S. Chitosan/functionalized fruit stones as a highly efficient adsorbent biomaterial for adsorption of brilliant green dye: Comprehensive characterization and statistical optimization. Int J Biol Macromol 2024; 263:130465. [PMID: 38423427 DOI: 10.1016/j.ijbiomac.2024.130465] [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: 12/03/2023] [Revised: 01/02/2024] [Accepted: 02/24/2024] [Indexed: 03/02/2024]
Abstract
In this research, a highly efficient adsorbent biomaterial (hereinafter, CTS/PPS-HS) of chitosan/functionalized fruit stones (peach and plum) with H2SO4 was produced for the adsorption of brilliant green (BG) dye from aquatic systems. The developed biomaterial was characterized by several techniques like SEM-EDX, FTIR, XRD, BET, and pHpzc. To systematically optimize the adsorption performance of CTS/PPS-HS, the Box-Behnken design (BBD) based on response surface methodology (RSM) was attained. The factors considered for optimization included A: CTS/PPS-HS dosage (0.02-0.08 g), B: pH (4-10), and C: removal time (10-60 min). The pseudo-first-order and Langmuir isotherm models exhibited excellent agreement with the experimental results of BG adsorption by CTS/PPS-HS. The outstanding adsorption capacity (409.63 mg/g) of CTS/PPS-HS was obtained. The remarkable adsorption of BG onto CTS/PPS-HS can be primarily attributed to electrostatic forces between the acidic sites of CTS/PPS-HS and the BG cations, accompanied by interactions such as π-π, Yoshida H-bonding, n-π, and H-bond interactions. The current data underscores the significant potential inherent in combining biomass with CTS polymer to create an exceptionally effective adsorbent biomaterial tailored for the elimination of cationic dyes.
Collapse
Affiliation(s)
- Mohammad K A Khan
- Department of Mechanical Engineering, College of Engineering, Najran University, Najran, Saudi Arabia
| | - Ahmed Saud Abdulhameed
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Anbar, Ramadi, Iraq; College of Engineering, University of Warith Al-Anbiyaa, Karbala, Iraq.
| | - Hassan Alshahrani
- Department of Mechanical Engineering, College of Engineering, Najran University, Najran, Saudi Arabia
| | - Sameer Algburi
- College of Engineering Technology, Al-Kitab University, Kirkuk 36015, Iraq
| |
Collapse
|
8
|
Gong L, Zhu J, Yang Y, Qiao S, Ma L, Wang H, Zhang Y. Effect of polyethylene glycol on polysaccharides: From molecular modification, composite matrixes, synergetic properties to embeddable application in food fields. Carbohydr Polym 2024; 327:121647. [PMID: 38171672 DOI: 10.1016/j.carbpol.2023.121647] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/21/2023] [Accepted: 11/25/2023] [Indexed: 01/05/2024]
Abstract
Polyethylene glycol (PEG) is a flexible, water-soluble, non-immunogenic, as well as biocompatible polymer, and it could synergize with polysaccharides for food applications. The molecular modification strategies, including covalent bond interactions (amino groups, carboxyl groups, aldehyde groups, tosylate groups, etc.), and non-covalent bond interactions (hydrogen bonding, electrostatic interactions, etc.) on PEG molecular chains are discussed. Its versatile structure, group modifiability, and amphiphilic block buildability could improve the functions of polysaccharides (e.g., chitosan, cellulose, starch, alginate, etc.) and adjust the properties of combined PEG/polysaccharides with outstanding chain tunability and matrix processability owing to plasticizing effects, compatibilizing effects, steric stabilizing effects and excluded volume effects by PEG, for achieving the diverse performance targets. The synergetic properties of PEG/polysaccharides with remarkable architecture were summarized, including mechanical properties, antibacterial activity, antioxidant performance, self-healing properties, carrier and delivery characteristics. The PEG/polysaccharides with excellent combined properties and embeddable merits illustrate potential applications including food packaging, food intelligent indication/detection, food 3D printing and nutraceutical food absorption. Additionally, prospects (like food innovation and preferable nutrient utilization) and key challenges (like structure-effectiveness-applicability relationship) for PEG/polysaccharides are proposed and addressed for food fields.
Collapse
Affiliation(s)
- Linshan Gong
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Juncheng Zhu
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Yuxin Yang
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Shihao Qiao
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Liang Ma
- College of Food Science, Southwest University, Chongqing 400715, PR China
| | - Hongxia Wang
- College of Food Science, Southwest University, Chongqing 400715, PR China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China; Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, PR China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, PR China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China.
| | - Yuhao Zhang
- College of Food Science, Southwest University, Chongqing 400715, PR China; Chongqing Key Laboratory of Speciality Food Co-Built by Sichuan and Chongqing, Chongqing 400715, PR China; Key Laboratory of Quality and Safety Control of Citrus Fruits, Ministry of Agriculture and Rural Affairs, Southwest University, Chongqing, 400712, PR China; Key Laboratory of Luminescence Analysis and Molecular Sensing (Southwest University), Ministry of Education, Chongqing 400715, PR China; Key Laboratory of Condiment Supervision Technology for State Market Regulation, Chongqing 401121, PR China.
| |
Collapse
|
9
|
Nguyen HT, Le AT, Nguyen TT, Huy TQ, Nguyen TTT. The role of chitosan in enhancing the solubility and antibacterial activity of emodin against drug-resistant bacteria. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:109-125. [PMID: 37843984 DOI: 10.1080/09205063.2023.2268964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 10/02/2023] [Indexed: 10/18/2023]
Abstract
Similar to most anthraquinone compounds, the pharmacological properties of emodin are limited because of its low water solubility. In this study, the formulation of chitosan and emodin (EMD/CS) was prepared by a bottom-up method with precipitation and sonication steps in order to enhance the solubility of emodin. Thanks to the interactions of oxygen-and nitrogen-containing groups in chitosan with emodin molecules, the solubility of emodin in the formulation was remarkably increased to 0.5 mg/mL. The EMD/CS particles were well dispersed and distributed in a range of sub-micrometer with an average particle size of 342 nm. The EMD/CS formulation exhibited synergic antibacterial activity of emodin and chitosan, against drug-resistant bacterial strains, namely Methicillin-resistant Staphylococcus aureus (MRSA) and Escherichia coli O157:H7 (E. coli O157:H7). When the compositions of emodin and chitosan increased, the antibacterial effectiveness of the EMD/CS formulation increased. The EMD/CS formulation with compositions of 0.5 mg/mL of emodin and 9.0 mg/mL of chitosan could significantly inhibit the proliferation of E. coli O157:H7. Meanwhile, the EMD/CS formulation with a lower concentration of emodin (0.4 mg/mL) and chitosan (7.2 mg/mL) could cause an extermination effect on MRSA. The enhanced solubility of EMD/CS formulation suggests that this formulation can be a potential candidate for the treatment of infectious diseases caused by drug-resistant bacterial pathogens.
Collapse
Affiliation(s)
- Hue Thi Nguyen
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, Vietnam
| | - Anh-Tuan Le
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, Vietnam
| | | | - Tran Quang Huy
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, Vietnam
- Faculty of Biomedical Sciences, Phenikaa University, Hanoi, Vietnam
| | - Thuy Thi Thu Nguyen
- Phenikaa University Nano Institute (PHENA), Phenikaa University, Hanoi, Vietnam
| |
Collapse
|
10
|
Li S, Wang Q, Duan X, Pei Z, He Z, Guo W, Han L. A glutathione-responsive PEGylated nanogel with doxorubicin-conjugation for cancer therapy. J Mater Chem B 2023; 11:11612-11619. [PMID: 38038224 DOI: 10.1039/d3tb01731a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
The complexity, degradability, and stability of drug delivery systems are crucial factors for clinical application. Herein, a glutathione (GSH)-responsive polyethylene glycol (PEG)ylated nanogel conjugated with doxorubicin (Dox) was prepared based on a linker with disulfide bonds, PEG, and Dox using a one-pot method. FT-IR and UV-vis analyses confirmed that all raw materials were incorporated in the Dox-conjugated nanogel structure. Dynamic light scattering (DLS) and transmission electron microscopy (TEM) results showed that the particle size of the Dox-conjugated nanogel was at the nanoscale and could be responsively disrupted in high GSH concentration. The in vitro accumulative Dox release rate from the nanogel reached 88% in PBS with 5 mg mL-1 GSH on day 4. Moreover, H22 cell viability and apoptosis experiments revealed that the nanogel effectively inhibited tumor cell growth. In vivo tracking and cell uptake experiments demonstrated that the nanogel accumulated and persisted in tumor tissues for 5 days and was distributed into cell nuclei at 6 h. Furthermore, H22-bearing mice experiments showed that the tumor size of the Dox-conjugated nanogel group was the smallest (287 mm3) compared to that of the free Dox (558 mm3) and 0.9% NaCl (2700 mm3) groups. Meanwhile, the body weight of mice as well as the H&E and TUNEL tissue section staining of organs and tumor tissues from the mice illustrated that the nanogel could significantly prevent side effects and induce tumor cell apoptosis. Taken together, compared with free Dox, the Dox-conjugated nanogel exhibited higher therapeutic efficacy and lower side effects in normal tissues, making it a potential novel nanomedicine for cancer.
Collapse
Affiliation(s)
- Shufen Li
- School of Pharmacy, Changzhi Medical College, Changzhi 046000, China.
- Department of Physiology, Changzhi Medical College, Changzhi, 046000, China.
| | - Qiang Wang
- School of Pharmacy, Changzhi Medical College, Changzhi 046000, China.
| | - Xiao Duan
- School of Pharmacy, Changzhi Medical College, Changzhi 046000, China.
- The Stem Cell and Tissue Engineering Research Center, Changzhi Medical College, Changzhi, 046000, China
| | - Zhen Pei
- Department of Physiology, Changzhi Medical College, Changzhi, 046000, China.
| | - Zhipeng He
- Department of Gastrointestinal Surgery, Heji Hospital Affiliated to Changzhi Medical College, Changzhi Medical College, Changzhi, 046000, China.
| | - Wei Guo
- Department of Gastrointestinal Surgery, Heji Hospital Affiliated to Changzhi Medical College, Changzhi Medical College, Changzhi, 046000, China.
| | - Lingna Han
- Department of Physiology, Changzhi Medical College, Changzhi, 046000, China.
| |
Collapse
|
11
|
Sato H, Yamada K, Miyake M, Onoue S. Recent Advancements in the Development of Nanocarriers for Mucosal Drug Delivery Systems to Control Oral Absorption. Pharmaceutics 2023; 15:2708. [PMID: 38140049 PMCID: PMC10747340 DOI: 10.3390/pharmaceutics15122708] [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: 10/30/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/24/2023] Open
Abstract
Oral administration of active pharmaceutical ingredients is desirable because it is easy, safe, painless, and can be performed by patients, resulting in good medication adherence. The mucus layer in the gastrointestinal (GI) tract generally acts as a barrier to protect the epithelial membrane from foreign substances; however, in the absorption process after oral administration, it can also disturb effective drug absorption by trapping it in the biological sieve structured by mucin, a major component of mucus, and eliminating it by mucus turnover. Recently, functional nanocarriers (NCs) have attracted much attention due to their immense potential and effectiveness in the field of oral drug delivery. Among them, NCs with mucopenetrating and mucoadhesive properties are promising dosage options for controlling drug absorption from the GI tracts. Mucopenetrating and mucoadhesive NCs can rapidly deliver encapsulated drugs to the absorption site and/or prolong the residence time of NCs close to the absorption membrane, providing better medications than conventional approaches. The surface characteristics of NCs are important factors that determine their functionality, owing to the formation of various kinds of interactions between the particle surface and mucosal components. Thus, a deeper understanding of surface modifications on the biopharmaceutical characteristics of NCs is necessary to develop the appropriate mucosal drug delivery systems (mDDS) for the treatment of target diseases. This review summarizes the basic information and functions of the mucosal layer, highlights the recent progress in designing functional NCs for mDDS, and discusses their performance in the GI tract.
Collapse
Affiliation(s)
- Hideyuki Sato
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (H.S.); (K.Y.)
| | - Kohei Yamada
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (H.S.); (K.Y.)
| | - Masateru Miyake
- Business Integrity and External Affairs, Otsuka Pharmaceutical Co., Ltd., 2-16-4 Konan, Minato-ku, Tokyo 108-8242, Japan;
| | - Satomi Onoue
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (H.S.); (K.Y.)
| |
Collapse
|
12
|
Liu X, Zhang J, Chen Z, Xiao J, Zhou A, Fu Y, Cao Y. Cluster-determinant 36 (CD36) mediates intestinal absorption of dietary astaxanthin and affects its secretion. Food Res Int 2023; 173:113328. [PMID: 37803639 DOI: 10.1016/j.foodres.2023.113328] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 10/08/2023]
Abstract
The functional activity of dietary astaxanthin is closely related to its absorption, and the absorption of dietary carotenoids mainly mediated by transmembrane transport protein (TTP) has become the mainstream research direction in recent years. However, the main TTP mediating astaxanthin absorption and its potential mechanisms are still unclear. Hence, based on the preliminary screening results, this study aims to elucidate the role of cluster-determinant 36 (CD36) mediating astaxanthin absorption from the perspective of expression levels through in vitro cell model, in situ single-pass intestinal perfusion model and in vivo mice model. The results showed that astaxanthin uptake was significantly increased by 45.13% in CD36 overexpressing cells and decreased by 20.92% in the case of sulfo-N-succinimidyl oleate (SSO) inhibition. A similar trend also appeared in the duodenum and jejunum by in situ model. Moreover, astaxanthin uptake in the small intestine of CD36 knockout mice was significantly reduced by 88.22%. Furthermore, the inhibition or knockout of CD36 suppressed the expression of other transporters (SR-BI and NPC1L1). Interestingly, CD36 was also involved in the downstream secretion pathway, which is manifested by interfering with the expression of related proteins (ERK1/2, MTP, ApoB48, and ApoAI). Therefore, these results indicate the important role of CD36 in astaxanthin transmembrane transport for the first time, providing vital exploration way for the absorption of dietary fat-soluble substances.
Collapse
Affiliation(s)
- Xiaojuan Liu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China.
| | - Junlin Zhang
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Zhiqing Chen
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Jie Xiao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Aimei Zhou
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| | - Yongshui Fu
- Institute of Blood Transfusion, Guangzhou Blood Center, Guangzhou 510095, China
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou 510642, China
| |
Collapse
|
13
|
Ghosh A, Banik S, Yamada K, Misaka S, Prud’homme RK, Sato H, Onoue S. Stabilized Astaxanthin Nanoparticles Developed Using Flash Nanoprecipitation to Improve Oral Bioavailability and Hepatoprotective Effects. Pharmaceutics 2023; 15:2562. [PMID: 38004541 PMCID: PMC10675309 DOI: 10.3390/pharmaceutics15112562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/25/2023] [Accepted: 10/25/2023] [Indexed: 11/26/2023] Open
Abstract
In this study, we developed stabilized astaxanthin (AX) nanoparticles (sNP/AX) to improve the physicochemical properties, oral bioavailability, and hepatoprotection of AX. A flash nanoprecipitation technique was used with a multi-inlet vortex mixer to prepare the sNP/AX. Vitamins E (VE) and C (VC) were used as co-stabilizers with poloxamer 407 as a stabilizer to inhibit the oxidative degradation of AX during sNP/AX formation and storage. VC stabilized AX in the aqueous phase during the preparation, whereas VE markedly improved the storage stability of sNP/AX, as evidenced by the AX contents remaining at 94 and 81% after 12 weeks of storage at 4 °C and 25 °C, respectively. The mean sNP/AX diameter was 215 nm, which resulted in higher AX release properties than those of crystalline AX. Rats, orally administered sNP/AX (33.2 mg AX/kg), exhibited higher systemic exposure to AX, whereas oral absorption in the crystalline AX group was negligible. In the rat hepatic injury model, oral pretreatment with sNP/AX (33.2 mg AX/kg) markedly attenuated hepatic damage, as shown by the histopathological analysis and reduced levels of plasma biomarkers for hepatic injury. These findings suggest that strategically including antioxidative additives in the sNP/AX has the potential to improve the physicochemical and nutraceutical properties of AX.
Collapse
Affiliation(s)
- Antara Ghosh
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (A.G.); (S.B.); (K.Y.); (H.S.)
| | - Sujan Banik
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (A.G.); (S.B.); (K.Y.); (H.S.)
| | - Kohei Yamada
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (A.G.); (S.B.); (K.Y.); (H.S.)
| | - Shingen Misaka
- Department of Bioregulation and Pharmacological Medicine, School of Medicine, Fukushima Medical University, 1 Hikarigaoka, Fukushima 960-1295, Japan;
| | - Robert K. Prud’homme
- Department of Chemical and Biological Engineering, A301 EQUAD, Princeton University, Princeton, NJ 80544, USA;
| | - Hideyuki Sato
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (A.G.); (S.B.); (K.Y.); (H.S.)
| | - Satomi Onoue
- Laboratory of Biopharmacy, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan; (A.G.); (S.B.); (K.Y.); (H.S.)
| |
Collapse
|
14
|
Guo M, Cui W, Li Y, Fei S, Sun C, Tan M, Su W. Microfluidic fabrication of size-controlled nanocarriers with improved stability and biocompatibility for astaxanthin delivery. Food Res Int 2023; 170:112958. [PMID: 37316049 DOI: 10.1016/j.foodres.2023.112958] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/25/2023] [Accepted: 05/10/2023] [Indexed: 06/16/2023]
Abstract
Improving the stability of astaxanthin (AST) is a vital way to enhance its oral bioavailability. In this study, a microfluidic strategy for the preparation of astaxanthin nano-encapsulation system was proposed. Thanks to the precise control of microfluidic and the rapid preparation ability of the Mannich reaction, the resulting astaxanthin nano-encapsulation system (AST-ACNs-NPs) was obtained with average sizes of 200 nm, uniform spherical shape and high encapsulation rate of 75%. AST was successfully doped into the nanocarriers, according to the findings of the DFT calculation, fluorescence spectrum, Fourier transform spectroscopy, and UV-vis absorption spectroscopy. Compared with free AST, AST-ACNs-NPs showed better stability under the conditions of high temperature, pH and UV light with<20% activity loss rate. The nano-encapsulation system containing AST could significantly reduce the hydrogen peroxide produced by reactive oxygen species, keep the potential of the mitochondrial membrane at a healthy level, and improve the antioxidant ability of H2O2-induced RAW 264.7 cells. These results indicated that microfluidics-based astaxanthin delivery system is an effective solution to improve the bioaccessibility of bioactive substances and has potential application value in food industry.
Collapse
Affiliation(s)
- Meng Guo
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Weina Cui
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Yuanchao Li
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China.
| | - Siyuan Fei
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Chaofan Sun
- College of Science, Northeast Forestry University, Harbin 150040, Heilongjiang, China
| | - Mingqian Tan
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China
| | - Wentao Su
- School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, Liaoning, China; Academy of Food Interdisciplinary Science, Dalian Polytechnic University, Dalian 116034, Liaoning, China; National Engineering Research Center of Seafood, Dalian 116034, Liaoning, China; State Key Laboratory of Marine Food Processing and Safety Control, Dalian Polytechnic University, Dalian 116034, Liaoning, China.
| |
Collapse
|
15
|
Zhang D, He J, Cui J, Wang R, Tang Z, Yu H, Zhou M. Oral Microalgae-Nano Integrated System against Radiation-Induced Injury. ACS NANO 2023; 17:10560-10576. [PMID: 37253200 DOI: 10.1021/acsnano.3c01502] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
The increasing applications of ionizing radiation in society raise the risk of radiation-induced intestinal and whole-body injury. Astaxanthin is a powerful antioxidant to reduce the reactive oxygen generated from radiation and the subsequent damage. However, the oral administration of astaxanthin remains challenging owing to its low solubility and poor bioavailability. Herein, we facilely construct an orally used microalgae-nano integrated system (SP@ASXnano) against radiation-induced intestinal and whole-body injury, combining natural microalgae Spirulina platensis (SP) with astaxanthin nanoparticles (ASXnano). SP and ASXnano show complementation in drug delivery to improve distribution in the intestine and blood. SP displays limited gastric drug loss, prolonged intestinal retention, constant ASXnano release, and progressive degradation. ASXnano improves drug solubility, gastric stability, cell uptake, and intestinal absorption. SP and ASXnano have synergy in many aspects such as anti-inflammation, microbiota protection, and fecal short-chain fatty acid up-regulation. In addition, the system is ensured with biosafety for long-term administration. The system organically combines the properties of microalgae and nanoparticles, which was expected to expand the medical application of SP as a versatile drug delivery platform.
Collapse
Affiliation(s)
- Dongxiao Zhang
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jian He
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Jiarong Cui
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Ruoxi Wang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
| | - Zhe Tang
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Hongyu Yu
- Department of Gastroenterology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200120, China
| | - Min Zhou
- Department of Surgery, The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
- Zhejiang University-University of Edinburgh Institute (ZJU-UoE Institute), Zhejiang University School of Medicine, Zhejiang University, Haining, 314400, China
- Institute of Translational Medicine, Zhejiang University, Hangzhou, 310009, China
- Cancer Center, Zhejiang University, Hangzhou, 310058, China
- Research Center for Life Science and Human Health, Binjiang Institute of Zhejiang University, Hangzhou, 310053, China
| |
Collapse
|
16
|
Abdelazim K, Ghit A, Assal D, Dorra N, Noby N, Khattab SN, El Feky SE, Hussein A. Production and therapeutic use of astaxanthin in the nanotechnology era. Pharmacol Rep 2023:10.1007/s43440-023-00488-y. [PMID: 37179259 PMCID: PMC10182848 DOI: 10.1007/s43440-023-00488-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 05/15/2023]
Abstract
Astaxanthin (AXT) is a red fat-soluble pigment found naturally in aquatic animals, plants, and various microorganisms and can be manufactured artificially using chemical catalysis. AXT is a xanthophyll carotenoid with a high potential for scavenging free radicals. Several studies have investigated AXT efficacy against diseases such as neurodegenerative, ocular, skin, and cardiovascular hypertension, diabetes, gastrointestinal and liver diseases, and immuno-protective functions. However, its poor solubility, low stability to light and oxygen, and limited bioavailability are major obstacles hindering its wide applications as a therapeutic agent or nutritional supplement. Incorporating AXT with nanocarriers holds great promise in enhancing its physiochemical properties. Nanocarriers are delivery systems with several benefits, including surface modification, bioactivity, and targeted medication delivery and release. Many approaches have been applied to enhance AXT's medicinal effect, including solid lipid nanoparticles, nanostructured lipid carriers (NLCs) and polymeric nanospheres. AXT nano-formulations have demonstrated a high antioxidant and anti-inflammatory effect, significantly affecting cancer in different organs. This review summarizes the most recent data on AXT production, characterization, biological activity, and therapeutic usage, focusing on its uses in the nanotechnology era.
Collapse
Affiliation(s)
- Karim Abdelazim
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Amr Ghit
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
- Department of Medicine and Aging Sciences, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
- Center for Advanced Studies and Technology, "G. d'Annunzio" University of Chieti-Pescara, Chieti, Italy
| | - Dina Assal
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
- Department of Biology, Biotechnology Program, American University in Cairo, Cairo, Egypt
| | - Neamat Dorra
- Department of Microbiology and Immunology, Faculty of Pharmacy, Sinai University, Arish, Egypt
| | - Nehad Noby
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt
| | - Sherine N Khattab
- Chemistry Department, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Shaymaa Essam El Feky
- Radiation Sciences Department, Medical Research Institute, University of Alexandria, Alexandria, Egypt
| | - Ahmed Hussein
- Department of Biotechnology, Institute of Graduate Studies and Research, Alexandria University, Alexandria, Egypt.
| |
Collapse
|
17
|
Anwer MK, Ahmed MM, Aldawsari MF, Iqbal M, Soliman GA, Aljuffali IA. Eluxadoline-Loaded Eudragit Nanoparticles for Irritable Bowel Syndrome with Diarrhea: Formulation, Optimization Using Box-Behnken Design, and Anti-Diarrheal Activity. Pharmaceutics 2023; 15:pharmaceutics15051460. [PMID: 37242700 DOI: 10.3390/pharmaceutics15051460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 05/04/2023] [Accepted: 05/04/2023] [Indexed: 05/28/2023] Open
Abstract
Eluxadoline (ELD), a recently approved drug, exhibits potential therapeutic effects in the management and treatment of IBS-D. However, its applications have been limited due to poor aqueous solubility, leading to a low dissolution rate and oral bioavailability. The current study's goals are to prepare ELD-loaded eudragit (EG) nanoparticles (ENPs) and to investigate the anti-diarrheal activity on rats. The prepared ELD-loaded EG-NPs (ENP1-ENP14) were optimized with the help of Box-Behnken Design Expert software. The developed formulation (ENP2) was optimized based on the particle size (286 ± 3.67 nm), PDI (0.263 ± 0.01), and zeta potential (31.8 ± 3.18 mV). The optimized formulation (ENP2) exhibited a sustained release behavior with maximum drug release and followed the Higuchi model. The chronic restraint stress (CRS) was successfully used to develop the IBS-D rat model, which led to increased defecation frequency. The in vivo studies revealed a significant reduction in defecation frequency and disease activity index by ENP2 compared with pure ELD. Thus, the results demonstrated that the developed eudragit-based polymeric nanoparticles can act as a potential approach for the effective delivery of eluxadoline through oral administration for irritable bowel syndrome diarrhea treatment.
Collapse
Affiliation(s)
- Md Khalid Anwer
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Mohammed F Aldawsari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Muzaffar Iqbal
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
- Bioavailability Laboratory, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Gamal A Soliman
- Department of Pharmacology and Toxicology, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
- Department of Pharmacology, College of Veterinary Medicine, Cairo University, Giza 12613, Egypt
| | - Ibrahim A Aljuffali
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
18
|
Xue Y, Liao Y, Wang H, Li S, Gu Z, Adu-Frimpong M, Yu J, Xu X, Smyth HDC, Zhu Y. Preparation and evaluation of astaxanthin-loaded 2-hydroxypropyl-beta-cyclodextrin and Soluplus® nanoparticles based on electrospray technology. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2023; 103:3628-3637. [PMID: 36840513 DOI: 10.1002/jsfa.12527] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Revised: 02/20/2023] [Accepted: 02/25/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Astaxanthin is a type of food-derived active ingredient with antioxidant, antidiabetic and non-toxicity functions, but its poor solubility and low bioavailability hinder further application in food industry. In the present study, through inclusion technologies, micellar solubilization and electrospray techniques, we prepared astaxanthin nanoparticles before optimizing the formulation to regulate the physical and chemical properties of micelles. We accomplished the preparation of astaxanthin nanoparticle delivery system based on single needle electrospray technology through use of 2-hydroxypropyl-β-cyclodextrin and Soluplus® to improveme the release behavior of the nanocarrier. RESULTS Through this experiment, we successfully prepared astaxanthin nanoparticles with a particle size of approximately 80 nm, which was further verified with scanning electron microscopy and transmission electron microscopy. Furthermore, the encapsulation of astaxanthin molecules into the carrier nanoparticles was verified via the results of attenuated total reflectance intensity and X-ray powder diffraction techniques. The in vitro release behavior of astaxanthin nanoparticles was different in media that contained 0.5% Tween 80 (pH 1.2, 4.5 and 6.8) buffer solution and distilled water. Also, we carried out a pharmacokinetic study of astaxanthin nanoparticles, in which it was observed that astaxanthin nanoparticle showed an effect of immediate release and significant improved bioavailability. CONCLUSION 2-hydroxypropyl-β-cyclodextrin and Soluplus® were used in the present study as a hydrophilic nanocarrier that could provide a simple way of encapsulating natural function food with repsect to improving the solubility and bioavailability of poorly water-soluble ingredients. © 2023 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yuanyuan Xue
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Youwu Liao
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Haiqiao Wang
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Shuang Li
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Zhengqing Gu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Michael Adu-Frimpong
- Department of Biochemistry and Forensic Sciences, School of Chemical and Biochemical Sciences, C. K. Tedam University of Technology and Applied Sciences (CKT-UTAS), Navrongo, Ghana
| | - Jiangnan Yu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Ximing Xu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| | - Hugh D C Smyth
- College of Molecular Pharmaceutics & Drug Delivery, The University of Texas at Austin, Austin, TX, USA
| | - Yuan Zhu
- Department of Pharmaceutics, School of Pharmacy, Center for Nano Drug/Gene Delivery and Tissue Engineering, Jiangsu University, Zhenjiang, China
| |
Collapse
|
19
|
Cai L, Gan M, Regenstein JM, Luan Q. Improving the biological activities of astaxanthin using targeted delivery systems. Crit Rev Food Sci Nutr 2023; 64:6902-6923. [PMID: 36779336 DOI: 10.1080/10408398.2023.2176816] [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: 02/14/2023]
Abstract
The antioxidant and anti-inflammatory properties of astaxanthin (AST) enable it to protect against oxidative stress-related and inflammatory diseases with a range of biological effects. These activities provide the potential to develop healthier food products. Therefore, it would be beneficial to design delivery systems for AST to overcome its low stability, control its release, and/or improve its bioavailability. This review discusses the basis for AST's various biological activities and the factors limiting these activities, including stability, solubility, and bioavailability. It also discusses the different systems available for the targeted delivery of AST and their applications in enhancing the biological activity of AST. These include systems that are candidates for preventive and therapeutic effects, which include nerves, liver, and skin, particularly for possible cancer reduction. Targeted delivery of AST to specific regions of the gastrointestinal tract, or more selectively to target tissues and cells, can be achieved using targeted delivery systems to increase the biological activities of AST.
Collapse
Affiliation(s)
- Luyun Cai
- Ningbo Innovation Center, College of Biosystems Engineering and Food Science, Zhejiang University, Ningbo, Zhejiang, China
| | - Miaoyu Gan
- Ningbo Innovation Center, College of Biosystems Engineering and Food Science, Zhejiang University, Ningbo, Zhejiang, China
| | - Joe M Regenstein
- Department of Food Science, Cornell University, Ithaca, New York, USA
| | - Qian Luan
- Ningbo Innovation Center, College of Biosystems Engineering and Food Science, Zhejiang University, Ningbo, Zhejiang, China
| |
Collapse
|
20
|
He J, Xie Y, Zhong J, Chen W, Fang S, Chen X, Peng S, Liu W, Liu C. Improving shikonin solubility and stability by encapsulation in natural surfactant-coated shikonin nanoparticles. J Food Sci 2023; 88:825-836. [PMID: 36625167 DOI: 10.1111/1750-3841.16445] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 11/21/2022] [Accepted: 12/15/2022] [Indexed: 01/11/2023]
Abstract
It is significant to develop a colloidal delivery system to improve the water solubility, stability, and bioavailability of shikonin, which is a hydrophobic plant polyphenol with a variety of physiological activities. In this study, three kinds of natural surfactants (saponin, sophorolipid, and rhamnolipid) were used to prepare shikonin nanoparticles by the pH-driven method. The physicochemical and structural properties of the shikonin nanoparticles were characterized, including particle size, zeta potential, and morphology. The encapsulation efficiencies of shikonin nanoparticles coated with saponin and sophorolipid were 97.6% and 97.3%, respectively, which were much higher than that of rhamnolipid-coated shikonin nanoparticles (19.0%). Shikonin nanoparticles coated with saponin and sophorolipid showed good resistance to heat and light and maintained long-term stability during storage. Moreover, shikonin nanoparticles coated with saponin and sophorolipid improved their in vitro-bioavailability. PRACTICAL APPLICATION: These article results are of great importance for improving the stability and bioavailability of shikonin in functional foods, dietary supplements, or pharmaceutical preparations. Moreover, this study provided theoretical and practical guides for further research of shikonin nanoparticles and may promote the development of natural colloidal delivery systems.
Collapse
Affiliation(s)
- Jie He
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Youfa Xie
- Jiangzhong Pharmaceutical Co., Ltd., Nanchang, Jiangxi, P. R. China
| | - Junzhen Zhong
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Wenrong Chen
- Sirio Pharma Co., Ltd., Shantou, Guangdong, P. R. China
| | - Suqiong Fang
- Sirio Pharma Co., Ltd., Shantou, Guangdong, P. R. China
| | - Xing Chen
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Shengfeng Peng
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, P. R. China
| | - Wei Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, P. R. China.,National R&D Center for Freshwater Fish Processing, Jiangxi Normal University, Nanchang, Jiangxi, P. R. China
| | - Chengmei Liu
- State Key Laboratory of Food Science and Technology, Nanchang University, Nanchang, Jiangxi, P. R. China
| |
Collapse
|
21
|
Chen Q, Riviere JE, Lin Z. Toxicokinetics, dose-response, and risk assessment of nanomaterials: Methodology, challenges, and future perspectives. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1808. [PMID: 36416026 PMCID: PMC9699155 DOI: 10.1002/wnan.1808] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/10/2022] [Accepted: 04/12/2022] [Indexed: 11/24/2022]
Abstract
The rapid growth of nanomaterial applications has raised safety concerns for human health. A number of studies have been conducted to assess the toxicokinetics, toxicology, dose-response, and risk assessment of different nanomaterials using in vitro and in vivo animal and human models. However, current studies cannot meet the demand for efficient assessment of toxicokinetics, dose-response relationships, or the toxicological risk arising from the rapidly increasing number of newly synthesized nanomaterials. In this article, we review the methods for conducting toxicokinetics, hazard identification, dose-response, exposure, and risk assessment studies of nanomaterials, identify the knowledge gaps, and discuss the challenges remaining. We provide the rationale behind the appropriate design of nanomaterial plasma toxicokinetic and tissue distribution studies, including caveats on the interpretation and correlation of in vitro and in vivo toxicology studies. The potential of using physiologically based pharmacokinetic (PBPK) models to extrapolate toxicokinetic and toxicity findings from in vitro to in vivo and from animals to humans is discussed, and the knowledge gaps of PBPK modeling for nanomaterials are identified. While challenges still exist, there has been progress in the toxicokinetics, hazard identification, and risk assessment of nanomaterials in the past two decades. Recent advancements in the field are highlighted with relevant examples. We also share latest guidelines as well as our perspectives on future studies needed to characterize the toxicokinetics, toxicity, and dose-response relationship in support of nanomaterial risk assessment. This article is categorized under: Toxicology and Regulatory Issues in Nanomedicine > Toxicology of Nanomaterials Toxicology and Regulatory Issues in Nanomedicine > Regulatory and Policy Issues in Nanomedicine.
Collapse
Affiliation(s)
- Qiran Chen
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| | - Jim E. Riviere
- 1Data Consortium, Kansas State University, Olathe, Kansas, USA
- Center for Chemical Toxicology Research and Pharmacokinetics, Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| | - Zhoumeng Lin
- Department of Environmental and Global Health, College of Public Health and Health Professions, University of Florida, Gainesville, Florida, USA
- Center for Environmental and Human Toxicology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
22
|
Imam SS, Gilani SJ, Bin Jumah MN, Rizwanullah M, Zafar A, Ahmed MM, Alshehri S. Harnessing Lipid Polymer Hybrid Nanoparticles for Enhanced Oral Bioavailability of Thymoquinone: In Vitro and In Vivo Assessments. Polymers (Basel) 2022; 14:3705. [PMID: 36145851 PMCID: PMC9504729 DOI: 10.3390/polym14183705] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Revised: 08/23/2022] [Accepted: 09/02/2022] [Indexed: 11/24/2022] Open
Abstract
The clinical application of phytochemicals such as thymoquinone (THQ) is restricted due to their limited aqueous solubility and oral bioavailability. Developing mucoadhesive nanocarriers to deliver these natural compounds might provide new hope to enhance their oral bioavailability. Herein, this investigation aimed to develop THQ-loaded lipid-polymer hybrid nanoparticles (THQ-LPHNPs) based on natural polymer chitosan. THQ-LPHNPs were fabricated by the nanoprecipitation technique and optimized by the 3-factor 3-level Box−Behnken design. The optimized LPHNPs represented excellent properties for ideal THQ delivery for oral administration. The optimized THQ-LPHNPs revealed the particles size (PS), polydispersity index (PDI), entrapment efficiency (%EE), and zeta potential (ZP) of <200 nm, <0.25, >85%, and >25 mV, respectively. THQ-LPHNPs represented excellent stability in the gastrointestinal milieu and storage stability in different environmental conditions. THQ-LPHNPs represented almost similar release profiles in both gastric as well as intestinal media with the initial fast release for 4 h and after that a sustained release up to 48 h. Further, the optimized THQ-LPHNPs represent excellent mucin binding efficiency (>70%). Cytotoxicity study revealed much better anti-breast cancer activity of THQ-LPHNPs compared with free THQ against MDA-MB-231 and MCF-7 breast cancer cells. Moreover, ex vivo experiments revealed more than three times higher permeation from the intestine after THQ-LPHNPs administration compared to the conventional THQ suspension. Furthermore, the THQ-LPHNPs showed 4.74-fold enhanced bioavailability after oral administration in comparison with the conventional THQ suspension. Therefore, from the above outcomes, mucoadhesive LPHNPs might be suitable nano-scale carriers for enhanced oral bioavailability and therapeutic efficacy of highly lipophilic phytochemicals such as THQ.
Collapse
Affiliation(s)
- Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| | - Sadaf Jamal Gilani
- Department of Basic Health Sciences, Preparatory Year, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - May Nasser Bin Jumah
- Biology Department, College of Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Environment and Biomaterial Unit, Health Sciences Research Center, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
- Saudi Society for Applied Science, Princess Nourah bint Abdulrahman University, Riyadh 11671, Saudi Arabia
| | - Md. Rizwanullah
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Ameeduzzafar Zafar
- Department of Pharmaceutics, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia
| | - Mohammed Muqtader Ahmed
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
23
|
Nanocarrier System: State-of-the-Art in Oral Delivery of Astaxanthin. Antioxidants (Basel) 2022; 11:antiox11091676. [PMID: 36139750 PMCID: PMC9495775 DOI: 10.3390/antiox11091676] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 11/17/2022] Open
Abstract
Astaxanthin (3,3′-dihydroxy-4,4′-diketo-β-β carotene), which belongs to the xanthophyll class, has shown potential biological activity in in vitro and in vivo models including as a potent antioxidant, anti-lipid peroxidation and cardiovascular disease prevention agent. It is mainly extracted from an alga, Haematococcus pluvialis. As a highly lipid-soluble carotenoid, astaxanthin has been shown to have poor oral bioavailability, which limits its clinical applications. Recently, there have been several suggestions and the development of various types of nano-formulation, loaded with astaxanthin to enhance their bioavailability. The employment of nanoemulsions, liposomes, solid lipid nanoparticles, chitosan-based and PLGA-based nanoparticles as delivery vehicles of astaxanthin for nutritional supplementation purposes has proven a higher oral bioavailability of astaxanthin. In this review, we highlight the pharmacological properties, pharmacokinetics profiles and current developments of the nano-formulations of astaxanthin for its oral delivery that are believed to be beneficial for future applications. The limitations and future recommendations are also discussed in this review.
Collapse
|
24
|
Kazmi I, Al-Abbasi FA, Imam SS, Afzal M, Nadeem MS, Altayb HN, Alshehri S. Formulation of Piperine Nanoparticles: In Vitro Breast Cancer Cell Line and In Vivo Evaluation. Polymers (Basel) 2022; 14:polym14071349. [PMID: 35406223 PMCID: PMC9003416 DOI: 10.3390/polym14071349] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/16/2022] [Accepted: 03/22/2022] [Indexed: 12/22/2022] Open
Abstract
Piperine (PPN), one of the most investigated phytochemicals, is known to have excellent therapeutic efficacy against a variety of ailments including breast cancer. However, its physicochemical properties such as poor aqueous solubility restrict its clinical application. Therefore, the present investigation was designed to develop PPN encapsulated lipid polymer hybrid nanoparticles (PPN-LPHNPs) to overcome the limitation. The developed PPN-LPHNPs were optimized by the three-factor, three-level Box−Behnken design (33-BBD). The optimized PPN-LPHNPs were then evaluated for their drug release profile, cytotoxicity assay against MDA-MB-231 and MCF-7 cells, and gastrointestinal stability as well as colloidal stability. In addition, the optimized PPN-LPHNPs were evaluated for ex vivo intestinal permeation and in vivo pharmacokinetic in albino Wistar rats. As per the results, the optimized PPN-LPHNPs showed a small average particles size of <160 nm with a low (<0.3) polydispersity index, and highly positive surface charge (>+20 mV). PPN-LPHNPs revealed excellent gastrointestinal as well as colloidal stability and sustained release profiles up to 24 h. Furthermore, PPN-LPHNPs revealed excellent cytotoxicity against both MDA-MB-231 and MCF-7 cancer cells compared to the free PPN. Moreover, animal studies revealed that the PPN-LPHNPs exhibited a 6.02- and 4.55-fold higher intestinal permeation and relative oral bioavailability, respectively, in comparison to the conventional PPN suspension. Thus, our developed LPHNPs present a strong potential for improved delivery of PPN.
Collapse
Affiliation(s)
- Imran Kazmi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.-A.); (M.S.N.); (H.N.A.)
- Correspondence: or (I.K.); (S.S.I.); Tel.: +966-543970731 (I.K.); +966-554543058 (S.S.I.)
| | - Fahad A. Al-Abbasi
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.-A.); (M.S.N.); (H.N.A.)
| | - Syed Sarim Imam
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
- Correspondence: or (I.K.); (S.S.I.); Tel.: +966-543970731 (I.K.); +966-554543058 (S.S.I.)
| | - Muhammad Afzal
- Department of Pharmacology, College of Pharmacy, Jouf University, Sakaka 72341, Saudi Arabia;
| | - Muhammad Shahid Nadeem
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.-A.); (M.S.N.); (H.N.A.)
| | - Hisham N. Altayb
- Department of Biochemistry, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; (F.A.A.-A.); (M.S.N.); (H.N.A.)
| | - Sultan Alshehri
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
25
|
Abuhassira-Cohen Y, Livney YD. Enhancing bioavailability of encapsulated hydrophobic nutraceuticals: Insights from in-vitro, in-vivo and clinical studies. Curr Opin Food Sci 2022. [DOI: 10.1016/j.cofs.2022.100832] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
26
|
Recent Advancement in Chitosan-Based Nanoparticles for Improved Oral Bioavailability and Bioactivity of Phytochemicals: Challenges and Perspectives. Polymers (Basel) 2021; 13:polym13224036. [PMID: 34833334 PMCID: PMC8617804 DOI: 10.3390/polym13224036] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/06/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
The excellent therapeutic potential of a variety of phytochemicals in different diseases has been proven by extensive studies throughout history. However, most phytochemicals are characterized by a high molecular weight, poor aqueous solubility, limited gastrointestinal permeability, extensive pre-systemic metabolism, and poor stability in the harsh gastrointestinal milieu. Therefore, loading of these phytochemicals in biodegradable and biocompatible nanoparticles (NPs) might be an effective approach to improve their bioactivity. Different nanocarrier systems have been developed in recent decades to deliver phytochemicals. Among them, NPs based on chitosan (CS) (CS-NPs), a mucoadhesive, non-toxic, and biodegradable polysaccharide, are considered the best nanoplatform for the oral delivery of phytochemicals. This review highlights the oral delivery of natural products, i.e., phytochemicals, encapsulated in NPs prepared from a natural polymer, i.e., CS, for improved bioavailability and bioactivity. The unique properties of CS for oral delivery such as its mucoadhesiveness, non-toxicity, excellent stability in the harsh environment of the GIT, good solubility in slightly acidic and alkaline conditions, and ability to enhance intestinal permeability are discussed first, and then the outcomes of various phytochemical-loaded CS-NPs after oral administration are discussed in detail. Furthermore, different challenges associated with the oral delivery of phytochemicals with CS-NPs and future directions are also discussed.
Collapse
|
27
|
Yang M, Kimchi ET, Staveley-O’Carroll KF, Li G. Astaxanthin Prevents Diet-Induced NASH Progression by Shaping Intrahepatic Immunity. Int J Mol Sci 2021; 22:11037. [PMID: 34681695 PMCID: PMC8541356 DOI: 10.3390/ijms222011037] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/08/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023] Open
Abstract
Dietary change leads to a precipitous increase in non-alcoholic fatty liver disease (NAFLD) from simple steatosis to the advanced form of non-alcoholic steatohepatitis (NASH), affecting approximately 25% of the global population. Although significant efforts greatly advance progress in clarifying the pathogenesis of NAFLD and identifying therapeutic targets, no therapeutic agent has been approved. Astaxanthin (ASTN), a natural antioxidant product, exerts an anti-inflammation and anti-fibrotic effect in mice induced with carbon tetrachloride (CCl4) and bile duct ligation (BDL); thus, we proposed to further investigate the potential effect of ASTN on a diet-induced mouse NASH and liver fibrosis, as well as the underlying cellular and molecular mechanisms. By treating pre-development of NASH in mice induced with a choline-deficient, L-amino acid-defined, high-fat diet (CDAHFD), we have demonstrated that oral administration ASTN preventively ameliorated NASH development and liver fibrosis by modulating the hepatic immune response, liver inflammation, and oxidative stress. Specifically, ASTN treatment led to the reduction in liver infiltration of monocyte-derived macrophages, hepatic stellate cell (HSC) activation, oxidative stress response, and hepatocyte death, accompanied by the decreased hepatic gene expression of proinflammatory cytokines such as TNF-α, TGF-β1, and IL-1β. In vitro studies also demonstrated that ASTN significantly inhibited the expression of proinflammatory cytokines and chemokine CCL2 in macrophages in response to lipopolysaccharide (LPS) stimulation. Overall, in vivo and in vitro studies suggest that ASTN functions as a promising therapeutic agent to suppress NASH and liver fibrosis via modulating intrahepatic immunity.
Collapse
Affiliation(s)
- Ming Yang
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA; (M.Y.); (E.T.K.)
| | - Eric T. Kimchi
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA; (M.Y.); (E.T.K.)
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
| | - Kevin F. Staveley-O’Carroll
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA; (M.Y.); (E.T.K.)
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
| | - Guangfu Li
- Department of Surgery, University of Missouri, Columbia, MO 65212, USA; (M.Y.); (E.T.K.)
- Harry S. Truman Memorial VA Hospital, Columbia, MO 65201, USA
- Department of Molecular Microbiology and Immunology, University of Missouri, Columbia, MO 65212, USA
| |
Collapse
|