1
|
Zhang M, Wang HY, Zhang YQ. A new method to immobilize urease in silk fibroin membrane by unidirectional nanopore dehydration. Biotechnol Prog 2025; 41:e3502. [PMID: 39238226 DOI: 10.1002/btpr.3502] [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: 04/28/2024] [Revised: 08/05/2024] [Accepted: 08/23/2024] [Indexed: 09/07/2024]
Abstract
The immobilization of free enzymes is crucial for enhancing their stability in different environments, enabling reusability, and expanding their applications. However, the development of a straightforward immobilization method that offers stability, high efficiency, biocompatibility, and modifiability remains a significant challenge. Silk fibroin (SF) is a good carrier for immobilized enzymes and drugs. Here, we employed urease as a model enzyme and utilized our developed technology called unidirectional nanopore dehydration (UND) to efficiently dehydrate a regenerated SF solution containing urease in a single step, resulting in the preparation of a highly functionalized SF membrane immobilizing urease (UI-SFM). The preparation process of UI-SFM is based on an all-water system, which is mild, green and able to efficiently and stably immobilize urease in the membranes, maintaining 92.7% and 82.8% relative enzyme activity after 30 days of storage in dry and hydrated states, respectively. Additionally, we performed additional post-treatments, including stretching and cross-linking with polyethylene glycol diglycidyl ether (PEGDE), to obtain two more robust immobilized urease membranes (UI-SFMs and UI-SFMc). The thermal and storage stability of these two membranes were significantly improved, and the recovery ratio of enzyme activity reached more than 90%. After 10 repetitions of the enzymatic reaction, the activity recovery of UI-SFMs and UI-SFMc remained at 92% and 88%, respectively. The results suggest that both UND-based and post-treatment-developed membranes exhibit excellent urease immobilization capabilities. Furthermore, the enzyme immobilization method offers a straightforward and versatile approach for efficient and stable enzyme immobilization, while its flexible modifiability caters to diverse application requirements.
Collapse
Affiliation(s)
- Meng Zhang
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, National Engineering Laboratory for Modern Silk, Soochow University, Suzhou, People's Republic of China
| | - Hai-Yan Wang
- Stomatology Department, The People's Hospital of Suzhou New District, Suzhou, People's Republic of China
| | - Yu-Qing Zhang
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, National Engineering Laboratory for Modern Silk, Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
2
|
Kovshova T, Malinovskaya J, Kotova J, Gorshkova M, Vanchugova L, Osipova N, Melnikov P, Vadekhina V, Nikitin A, Ermolenko Y, Gelperina S. Core-Shell PLGA Nanoparticles: In Vitro Evaluation of System Integrity. Biomolecules 2024; 14:1601. [PMID: 39766308 PMCID: PMC11674307 DOI: 10.3390/biom14121601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/06/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The objective of this study was to compare the properties of core-shell nanoparticles with a PLGA core and shells composed of different types of polymers, focusing on their structural integrity. The core PLGA nanoparticles were prepared either through a high-pressure homogenization-solvent evaporation technique or nanoprecipitation, using poloxamer 188 (P188), a copolymer of divinyl ether with maleic anhydride (DIVEMA), and human serum albumin (HSA) as the shell-forming polymers. The shells were formed through adsorption, interfacial embedding, or conjugation. For dual fluorescent labeling, the core- and shell-forming polymers were conjugated with Cyanine5, Cyanine3, and rhodamine B. The nanoparticles had negative zeta potentials and sizes ranging from 100 to 250 nm (measured using DLS) depending on the shell structure and preparation technique. The core-shell structure was confirmed using TEM and fluorescence spectroscopy, with the appearance of FRET phenomena due to the donor-acceptor properties of the labels. All of the shells enhanced the cellular uptake of the nanoparticles in Gl261 murine glioma cells. The integrity of the core-shell structures upon their incubation with the cells was evidenced by intracellular colocalization of the fluorescent labels according to the Manders' colocalization coefficients. This comprehensive approach may be useful for the selection of the optimal preparation method even at the early stages of the core-shell nanoparticle development.
Collapse
Affiliation(s)
- Tatyana Kovshova
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Julia Malinovskaya
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Julia Kotova
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Marina Gorshkova
- Laboratory of Polyelectrolyte Chemistry and Biomedical Polymers, Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences, Leninsky Prosp. 29, Moscow 119991, Russia; (M.G.); (L.V.)
| | - Lyudmila Vanchugova
- Laboratory of Polyelectrolyte Chemistry and Biomedical Polymers, Topchiev Institute of Petrochemical Synthesis, Russian Academy of Sciences, Leninsky Prosp. 29, Moscow 119991, Russia; (M.G.); (L.V.)
| | - Nadezhda Osipova
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Pavel Melnikov
- Department of Fundamental and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow 119034, Russia; (P.M.); (V.V.)
| | - Veronika Vadekhina
- Department of Fundamental and Applied Neurobiology, V. Serbsky Federal Medical Research Centre of Psychiatry and Narcology of the Ministry of Health of the Russian Federation, Kropotkinskiy per. 23, Moscow 119034, Russia; (P.M.); (V.V.)
| | - Alexey Nikitin
- Laboratory of Biomedical Nanomaterials, National University of Science and Technology (MISIS), Leninsky Prosp. 4, Moscow 119049, Russia;
| | - Yulia Ermolenko
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| | - Svetlana Gelperina
- Faculty of Chemical and Pharmaceutical Technologies and Biomedical Preparations, D. Mendeleev University of Chemical Technology of Russia, Miusskaya pl. 9, Moscow 125047, Russia; (J.M.); (J.K.); (N.O.); (Y.E.)
| |
Collapse
|
3
|
Ouyang Z, Chen X, Wang Z, Xu Y, Deng Z, Xing L, Zhang L, Hu M, Li H, Lian T, Gao F, Liu C, Zhou Y, Sun L, Wang YC, Liu D. Azithromycin-loaded PLGA microspheres coated with silk fibroin ameliorate inflammation and promote periodontal tissue regeneration. Regen Biomater 2024; 12:rbae146. [PMID: 39791015 PMCID: PMC11717352 DOI: 10.1093/rb/rbae146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/06/2024] [Accepted: 12/08/2024] [Indexed: 01/12/2025] Open
Abstract
Periodontitis, a widespread inflammatory disease, is the major cause of tooth loss in adults. While mechanical periodontal therapy benefits the periodontal disease treatment, adjunctive periodontal therapy is also necessary. Topically applied anti-inflammatory agents have gained considerable attention in periodontitis therapy. Although azithromycin (AZM) possesses excellent anti-inflammatory properties, its bioavailability is limited owing to poor water solubility and the absence of sustained release mechanisms. Herein, we synthesized biodegradable microspheres (AZM@PLGA-SF) for sustained AZM release to locally ameliorate periodontal inflammation and facilitate periodontal tissue regeneration. AZM was encapsulated in poly (lactic-co-glycolic acid) (PLGA) microspheres (AZM@PLGA) using single emulsion-solvent evaporation, followed by surface coating with silk fibroin (SF) via electrostatic adsorption, reducing the initial burst release of AZM. In vivo, local treatment with AZM@PLGA-SF microspheres significantly reduced periodontal inflammation and restored periodontal tissue to healthy levels. Mechanically, the formulated microspheres regulated the periodontal inflammatory microenvironment by reducing the levels of pro-inflammatory cytokines (tumor necrosis factor -α, interleukin [IL]-6, interferon-γ, IL-2, and IL-17A) in gingival crevicular fluid and promoted the expression of anti-inflammatory cytokines (IL-4 and IL-10). AZM@PLGA-SF microspheres demonstrated excellent biological safety. Therefore, we introduce an anti-inflammatory therapy for periodontitis with substantial potential for mitigating periodontal inflammation and encouraging the repair and regeneration of periodontal tissues.
Collapse
Affiliation(s)
- Zhaoguang Ouyang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction & Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, Guangzhou 510013, PR China
| | - Xiaoyu Chen
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Zhengyang Wang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Yue Xu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Zhe Deng
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, Hunan 410208, PR China
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MA 21205, USA
| | - Liangyu Xing
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Li Zhang
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Meilin Hu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Haocong Li
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Tengye Lian
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Feng Gao
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Chunyi Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Yangyang Zhou
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
| | - Lu Sun
- Department of Periodontics and Oral Medicine, University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
- Periodontal and Implant Microsurgery Academy (PiMA), University of Michigan School of Dentistry, Ann Arbor, MI 48105, USA
| | - Ying ChengYao Wang
- Department of Operative Dentistry and Endodontics, Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin 300041, PR China
- Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin 300041, PR China
| | - Dayong Liu
- Department of Endodontics, Tianjin Medical University School and Hospital of Stomatology & Tianjin Key Laboratory of Oral Soft and Hard Tissues Restoration and Regeneration, Tianjin 300070, PR China
- Tianjin Medical University Institute of Stomatology, Tianjin 300070, PR China
- School and Hospital of Stomatology, Hebei Medical University & Hebei Key Laboratory of Stomatology & Hebei Clinical Research Center for Oral Diseases, Shijiazhuang, Hebei 050011, PR China
| |
Collapse
|
4
|
Alves LP, Oliveira KDS, dos Santos ACG, de Melo DF, Moreira LMCDC, Oshiro Junior JA, da Silva DTC, Cavalcanti ALDM, Damasceno BPGDL. Cellulose Acetate Microparticles Synthesized from Agave sisalana Perrine for Controlled Release of Simvastatin. Polymers (Basel) 2024; 16:1898. [PMID: 39000753 PMCID: PMC11243862 DOI: 10.3390/polym16131898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/14/2024] [Accepted: 06/22/2024] [Indexed: 07/17/2024] Open
Abstract
Simvastatin (SIM) is widely prescribed to treat hyperlipidemia, despite its limitations, such as a short half-life and low oral bioavailability. To overcome these drawbacks, the development of a controlled-release formulation is desirable. This study aims to develop a microparticulate system based on cellulose acetate (ACT) obtained from Agave sisalana Perrine to promote a controlled SIM release. SIM-loaded microparticles (SMP) were prepared using the solvent emulsification-evaporation method. Several parameters were evaluated, including particle size, surface charge, morphology, encapsulation efficiency, thermochemical characteristics, crystallinity, and in vitro release profile. ACT exhibited favorable flow properties after acetylation, with a degree of substitution values superior to 2.5, as confirmed by both the chemical route and H-NMR, indicating the formation of cellulose triacetate. The obtained SMP were spherical with an average size ranging from 1842 to 1857 nm, a zeta potential of -4.45 mV, and a high SIM incorporation efficiency (98%). Thermal and XRD analyses revealed that SIM was homogeneously dispersed into the polymeric matrix in its amorphous state. In vitro studies using dialysis bags revealed that the controlled SIM release from microparticles was higher under simulated intestinal conditions and followed the Higuchi kinetic model. Our results suggest that ACT-based microparticles are a promising system for SIM delivery, which can improve its bioavailability, and result in better patient compliance.
Collapse
Affiliation(s)
- Larissa Pereira Alves
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - Kevin da Silva Oliveira
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - Ana Cláudia Gonçalves dos Santos
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - Demis Ferreira de Melo
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - Lívia Maria Coelho de Carvalho Moreira
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - João Augusto Oshiro Junior
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
| | - Dayanne Tomaz Casimiro da Silva
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - Airlla Laana de Medeiros Cavalcanti
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| | - Bolívar Ponciano Goulart de Lima Damasceno
- Graduate Program of Pharmaceutical Sciences, Paraíba State University, Campina Grande 58429-600, PB, Brazil; (L.P.A.); (K.d.S.O.); (D.F.d.M.); (L.M.C.d.C.M.); (J.A.O.J.); (D.T.C.d.S.); (B.P.G.d.L.D.)
- Laboratory of Development and Characterization of Pharmaceutical Products, Department of Pharmacy, Paraíba State University, Campina Grande 58429-600, PB, Brazil;
| |
Collapse
|
5
|
Xiong H, Zhang P, Wang D, Zhou Z, Sun J, Diao M. A silk-based hydrogel containing dexamethasone and lipoic acid microcrystals for local delivery to the inner ear. Colloids Surf B Biointerfaces 2024; 237:113855. [PMID: 38513298 DOI: 10.1016/j.colsurfb.2024.113855] [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: 02/05/2024] [Revised: 03/13/2024] [Accepted: 03/15/2024] [Indexed: 03/23/2024]
Abstract
Local drug delivery has been exploited recently to treat hearing loss, as this method can both bypass the blood-labyrinth barrier and provide sustained drug release. Combined drug microcrystals (MCs) offer additional advantages for sensorineural hearing loss treatment via intratympanic (IT) injection due to their shape effect and combination strategy. In this study, to endow viscous effects of hydrogels, nonspherical dexamethasone (DEX) and lipoic acid (LA) MCs were incorporated into silk fibroin (SF) hydrogels, which were subsequently administered to the tympanic cavity to investigate their pharmaceutical properties. First, we prepared DEX and LA MCs by a traditional precipitation technique followed by SF hydrogel incorporation (SF+DEX+LA). After characterization of the physicochemical features, including morphology, rheology, and dissolution, both a suspension of combined DEX and LA MCs (DEX+LA) and SF+DEX+LA were administered to guinea pigs by IT injection, after which the pharmacokinetics, biodegradation and biocompatibility were evaluated. To our surprise, compared to the DEX+LA group, the pharmacokinetics of the SF+DEX+LA hydrogel group did not improve significantly, which may be ascribed to their nonspherical shape and deposition effects of the drugs MCs. The cochlear tissue in each group displayed good morphology, with no obvious inflammatory reactions. This combined MC suspension has the clear advantages of no vehicle, easy scale-up preparation, and good biocompatibility and outcomes, which paves the way for practical treatment of hearing loss via local drug delivery.
Collapse
Affiliation(s)
- Haixia Xiong
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China
| | - Peili Zhang
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China
| | - Dongcheng Wang
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Jianjun Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Peking University International Hospital, Beijing 102206, China.
| | - Mingfang Diao
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital of Beijing, Beijing 100048, China.
| |
Collapse
|
6
|
Deng Q, Lin P, Gu H, Zhuang X, Wang F. Silk Protein-Based Nanoporous Microsphere for Controllable Drug Delivery through Self-Assembly in Ionic Liquid System. Biomacromolecules 2024; 25:1527-1540. [PMID: 38307005 DOI: 10.1021/acs.biomac.3c01104] [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: 02/04/2024]
Abstract
Ionic liquids (ILs) showed a promising application prospect in the field of biomedicine due to their unique recyclability, modifiability, and structure adjustability. In this study, nanoporous microsphere of silk protein and blending with poly(d,l-lactic acid) as model drug delivery was fabricated, respectively, through an IL-induced self-assembly method. Their morphology, structure, and thermal properties were comparably investigated through scanning electron microscopy, transmission electron microscopy, Fourier transform infrared spectroscopy, X-ray photoelectron spectroscopy, differential scanning calorimetry, X-ray diffraction, and thermogravimetric analyses, and the interaction mechanisms were also discussed to elucidate the effect of structure on drug delivery kinetics. The pure protein exhibited a bigger nanopore size in the microsphere compared to the composite one, facilitating more effective drug loading up to 88.7%. However, drug release was over 53.5% for the composite during initial 4 h, while pure protein was only about half of the composite. Both of them exhibited sustained slow release after 24 h and anticancer efficacy. Furthermore, the favorable compatibility between drug and microsphere vehicle was found and experienced improved thermal stability upon encapsulation, which could protect the drug molecules in high temperature at 200 °C. When the protein and its composite self-assembled to microspheres in ILs due to electrostatic and hydrophobic interaction, the drug could be infiltrated into the nanoporous matrix through biophysical action, and the protein structure displayed reversible transition during delivery. The sustained slow release from pure SF was attributed to the high β-sheet block action and strong drug-protein interactions, whose strength could be tuned through blending poly(d,l-lactic acid) with protein. These findings indicated that the SF-based nanoporous microspheres formed from IL self-assembled system are an ideal and potential drug delivery vehicle which can be incorporated into various biomaterials in the future.
Collapse
Affiliation(s)
- Qianqian Deng
- Center of Analysis and Testing, Nanjing Normal University, Nanjing 210023, China
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Ping Lin
- Center of Analysis and Testing, Nanjing Normal University, Nanjing 210023, China
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Hanling Gu
- Center of Analysis and Testing, Nanjing Normal University, Nanjing 210023, China
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Xincheng Zhuang
- Center of Analysis and Testing, Nanjing Normal University, Nanjing 210023, China
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| | - Fang Wang
- Center of Analysis and Testing, Nanjing Normal University, Nanjing 210023, China
- School of Chemistry and Materials Science, Nanjing Normal University, Nanjing 210023, China
| |
Collapse
|
7
|
Wang HY, Zhang Y, Zhang M, Zhang YQ. Functional modification of silk fibroin from silkworms and its application to medical biomaterials: A review. Int J Biol Macromol 2024; 259:129099. [PMID: 38176506 DOI: 10.1016/j.ijbiomac.2023.129099] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 12/26/2023] [Accepted: 12/26/2023] [Indexed: 01/06/2024]
Abstract
Silk fibroin (SF) from the silkworm Bombyx mori is a fibrous protein identified as a widely suitable biomaterial due to its biocompatibility, tunable degradation, and mechanical strength. Various modifications of SF protein can give SF fibers new properties and functions, broadening their applications in textile and biomedical industries. A diverse array of functional modifications on various forms of SF has been reported. In order to provide researchers with a more systematic understanding of the types of functional modifications of SF protein, as well as the corresponding applications, we comprehensively review the different types of functional modifications, including transgenic modification, modifications with chemical groups or biologically active substance, cross-linking and copolymerization without chemical reactions, their specific modification methods and applications. Furthermore, recent applications of SF in various medical biomaterials are briefly discussed.
Collapse
Affiliation(s)
- Hai-Yan Wang
- Obstetrical department, The People's Hospital of Suzhou New District, Suzhou, China
| | - Yun Zhang
- Obstetrical department, The People's Hospital of Suzhou New District, Suzhou, China
| | - Meng Zhang
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, China
| | - Yu-Qing Zhang
- Silk Biotechnology Laboratory, School of Biology and Basic Medical Sciences, Soochow University, Suzhou, China.
| |
Collapse
|
8
|
Hu J, Jiang Z, Zhang J, Yang G. Application of silk fibroin coatings for biomaterial surface modification: a silk road for biomedicine. J Zhejiang Univ Sci B 2023; 24:943-956. [PMID: 37961798 PMCID: PMC10646393 DOI: 10.1631/jzus.b2300003] [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: 01/02/2023] [Accepted: 04/18/2023] [Indexed: 11/15/2023]
Abstract
Silk fibroin (SF) as a natural biopolymer has become a popular material for biomedical applications due to its minimal immunogenicity, tunable biodegradability, and high biocompatibility. Nowadays, various techniques have been developed for the applications of SF in bioengineering. Most of the literature reviews focus on the SF-based biomaterials and their different forms of applications such as films, hydrogels, and scaffolds. SF is also valuable as a coating on other substrate materials for biomedicine; however, there are few reviews related to SF-coated biomaterials. Thus, in this review, we focused on the surface modification of biomaterials using SF coatings, demonstrated their various preparation methods on substrate materials, and introduced the latest procedures. The diverse applications of SF coatings for biomedicine are discussed, including bone, ligament, skin, mucosa, and nerve regeneration, and dental implant surface modification. SF coating is conducive to inducing cell adhesion and migration, promoting hydroxyapatite (HA) deposition and matrix mineralization, and inhibiting the Notch signaling pathway, making it a promising strategy for bone regeneration. In addition, SF-coated composite scaffolds can be considered prospective candidates for ligament regeneration after injury. SF coating has been proven to enhance the mechanical properties of the substrate material, and render integral stability to the dressing material during the regeneration of skin and mucosa. Moreover, SF coating is a potential strategy to accelerate nerve regeneration due to its dielectric properties, mechanical flexibility, and angiogenesis promotion effect. In addition, SF coating is an effective and popular means for dental implant surface modification to promote osteogenesis around implants made of different materials. Thus, this review can be of great benefit for further improvements in SF-coated biomaterials, and will undoubtedly contribute to clinical transformation in the future.
Collapse
Affiliation(s)
- Jinxing Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Jing Zhang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Disease, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
9
|
Wang X, Zhou Z, Yu C, He K, Sun L, Kou Y, Zhang M, Zhang Z, Luo P, Wen L, Chen G. A prestin-targeting peptide-guided drug delivery system rearranging concentration gradient in the inner ear: An improved strategy against hearing loss. Eur J Pharm Sci 2023; 187:106490. [PMID: 37295658 DOI: 10.1016/j.ejps.2023.106490] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 05/23/2023] [Accepted: 06/06/2023] [Indexed: 06/12/2023]
Abstract
Hearing loss is mainly due to outer hair cell (OHC) damage in three cochlear turns. Local administration via the round window membrane (RWM) has considerable otological clinical potential in bypassing the blood-labyrinth barrier. However, insufficient drug distribution in the apical and middle cochlear turns results in unsatisfactory efficacy. We functionalized poly (lactic-co-glycolic acid) nanoparticles (PLGA NPs) with targeting peptide A665, which specifically bound to prestin, a protein uniquely expressed in OHCs. The modification facilitated the cellular uptake and RWM permeability of NPs. Notably, the guide of A665 towards OHCs enabled more NPs perfusion in the apical and middle cochlear turns without decreasing accumulation in the basal cochlear turn. Subsequently, curcumin (CUR), an appealing anti-ototoxic drug, was encapsulated in NPs. In aminoglycoside-treated guinea pigs with the worst hearing level, CUR/A665-PLGA NPs, with superior performance to CUR/PLGA NPs, almost completely preserved the OHCs in three cochlear turns. The lack of increased low-frequencies hearing thresholds further confirmed that the delivery system with prestin affinity mediated cochlear distribution rearrangement. Good inner ear biocompatibility and little or no embryonic zebrafish toxicity were observed throughout the treatment. Overall, A665-PLGA NPs act as desirable tools with sufficient inner ear delivery for improved efficacy against severe hearing loss.
Collapse
Affiliation(s)
- Xinrui Wang
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Zeming Zhou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Chong Yu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Kerui He
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Lifang Sun
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Yuwei Kou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China
| | - Ming Zhang
- Guangdong Sunho Pharmaceutical Co. Ltd, Zhongshan 528437, China
| | - Zhifeng Zhang
- State Key Laboratory for Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 000853, China
| | - Pei Luo
- State Key Laboratory for Quality Research in Chinese Medicine, Macau University of Science and Technology, Macau 000853, China
| | - Lu Wen
- School of Pharmacy, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| | - Gang Chen
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery & Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System & Class III Laboratory of Modern Chinese Medicine Preparation & Key Laboratory of Modern Chinese Medicine of Education Department of Guangdong Province, Center for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou 510006, China.
| |
Collapse
|
10
|
Wang X, Xiong H, Zhang P, Liu Y, Gao C, Zhou Z, Sun J, Diao M. Intratympanic microcrystals of dexamethasone and lipoic acid for the treatment of cisplatin-induced inner ear injury. Colloids Surf B Biointerfaces 2023; 223:113191. [PMID: 36739674 DOI: 10.1016/j.colsurfb.2023.113191] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/29/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Steroids (anti-inflammatory drugs) combined with antioxidants are frequently prescribed to treat cisplatin (CP)-induced hearing loss in the clinic. Compared to systemic administration of free drugs, local drug delivery systems offer better therapeutic qualities and patient compliance since they not only can bypass the blood-labyrinth barrier but also can perform sustained release. In this work, dexamethasone (DEX) and lipoic acid (LA) non-spherical microcrystals (MCs) were prepared without complicated chemical modification. Following a series of physical characterizations, including morphology, stability and injectability, dissolution and round window membrane distribution of MCs, DEX MCs, LA MCs and the simple mixture of DEX MCs + LA MCs (combination group) were administered in guinea pigs by intratympanic injection. We found that LA MCs enabled improvement of DEX absorption in the combination group compared to a single dose. In addition, no significant morphological changes or inflammatory responses were observed in cochlear tissue, indicating good biocompatibility. Finally, the combination group also demonstrated synergistic therapeutic effect for protecting hair cells against CP-induced damage. The local co delivery of DEX MCs and LA MCs offers a new strategy for the treatment of CP-induced inner ear injury since they provide sustained anti-inflammatory and antioxidant effects simultaneously.
Collapse
Affiliation(s)
- Xiangxiang Wang
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Haixia Xiong
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Peili Zhang
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China
| | - Ya Liu
- Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; National Clinical Medical Research Center for Otolaryngology Diseases, Beijing 100048, China
| | - Chang Gao
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China.
| | - Jianjun Sun
- Department of Otorhinolaryngology Head and Neck Surgery, Peking University International Hospital, Beijing 102206, China.
| | - Mingfang Diao
- Department of Otolaryngology, School of Medicine, South China University of Technology, Guangzhou 510006, China; Department of Endoscopic Ear Surgery, Senior Department of Otorhinolaryngology Head and Neck Surgery, The Sixth Medical Center of PLA General Hospital, Beijing 100048, China; National Clinical Medical Research Center for Otolaryngology Diseases, Beijing 100048, China.
| |
Collapse
|
11
|
Han CS, Kang JH, Kim YJ, Kim DW, Park CW. Inhalable Nano-Dimpled Microspheres Containing Budesonide-PLGA for Improved Aerodynamic Performance. Int J Nanomedicine 2022; 17:3405-3419. [PMID: 35945926 PMCID: PMC9357420 DOI: 10.2147/ijn.s372582] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Accepted: 07/24/2022] [Indexed: 11/23/2022] Open
Abstract
Introduction Dry powder inhalations are an attractive pharmaceutical dosage form. They are environmentally friendly, portable, and physicochemical stable compared to other inhalation forms like pressurized metered-dose inhalers and nebulizers. Sufficient drug deposition of DPIs into the deep lung is required to enhance the therapeutic activity. Nanoscale surface roughness in microparticles could improve aerosolization and aerodynamic performance. This study aimed to prepare microspheres with nanoscale dimples and confirm the effect of roughness on inhalation efficiency. Methods The dimpled-surface on microspheres (MSs) was achieved by oil in water (O/W) emulsion-solvent evaporation by controlling the stirring rate. The physicochemical properties of MSs were characterized. Also, in vitro aerodynamic performance of MSs was evaluated by particle image velocimetry and computational fluid dynamics. Results The particle image velocimetry results showed that dimpled-surface MSs had better aerosolization, about 20% decreased X-axial velocity, and a variable angle, which could improve the aerodynamic performance. Furthermore, it was confirmed that the dimpled surface of MSs could cause movement away from the bronchial surface, which helps the MSs travel into the deep lung using computational fluid dynamics. Conclusion The dimpled-surface MSs showed a higher fine particle fraction value compared to smooth-surface MSs in the Andersen Cascade Impactor, and surface roughness like dimples on microspheres could improve aerosolization and lung deposition.
Collapse
Affiliation(s)
- Chang-Soo Han
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Ji-Hyun Kang
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Young-Jin Kim
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
| | - Dong-Wook Kim
- College of Pharmacy, Wonkwang University, Iksan, 54538, Republic of Korea
| | - Chun-Woong Park
- College of Pharmacy, Chungbuk National University, Cheongju, 28160, Republic of Korea
- Correspondence: Chun-Woong Park, College of Pharmacy, Chungbuk National University, 194-21, Osongsangmyeong 1-ro, Heungdeok-gu, Cheongju, Chungbuk, 28160, Republic of Korea, Tel +82-43-261-3330, Fax +82-43-268-2732, Email
| |
Collapse
|
12
|
Yuan X, Yuan Z, Wang Y, Wan Z, Wang X, Yu S, Han J, Huang J, Xiong C, Ge L, Cai Q, Zhao Y. Vascularized pulp regeneration via injecting simvastatin functionalized GelMA cryogel microspheres loaded with stem cells from human exfoliated deciduous teeth. Mater Today Bio 2022; 13:100209. [PMID: 35198958 PMCID: PMC8841886 DOI: 10.1016/j.mtbio.2022.100209] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/20/2022] [Accepted: 01/27/2022] [Indexed: 12/20/2022] Open
|
13
|
Sharma K, Porat Z, Gedanken A. Designing Natural Polymer-Based Capsules and Spheres for Biomedical Applications-A Review. Polymers (Basel) 2021; 13:4307. [PMID: 34960858 PMCID: PMC8708131 DOI: 10.3390/polym13244307] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 11/29/2021] [Accepted: 12/06/2021] [Indexed: 12/12/2022] Open
Abstract
Natural polymers, such as polysaccharides and polypeptides, are potential candidates to serve as carriers of biomedical cargo. Natural polymer-based carriers, having a core-shell structural configuration, offer ample scope for introducing multifunctional capabilities and enable the simultaneous encapsulation of cargo materials of different physical and chemical properties for their targeted delivery and sustained and stimuli-responsive release. On the other hand, carriers with a porous matrix structure offer larger surface area and lower density, in order to serve as potential platforms for cell culture and tissue regeneration. This review explores the designing of micro- and nano-metric core-shell capsules and porous spheres, based on various functions. Synthesis approaches, mechanisms of formation, general- and function-specific characteristics, challenges, and future perspectives are discussed. Recent advances in protein-based carriers with a porous matrix structure and different core-shell configurations are also presented in detail.
Collapse
Affiliation(s)
- Kusha Sharma
- Department of Chemistry, Bar-Ilan Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel;
| | - Ze’ev Porat
- Department of Civil and Environmental Engineering, Ben-Gurion University of the Negev, Be’er Sheva 84105, Israel
- Department of Chemistry, Nuclear Research Center-Negev, Be’er Sheva 84190, Israel
| | - Aharon Gedanken
- Department of Chemistry, Bar-Ilan Institute for Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 52900, Israel;
| |
Collapse
|
14
|
Antitumor Activity of Rosmarinic Acid-Loaded Silk Fibroin Nanoparticles on HeLa and MCF-7 Cells. Polymers (Basel) 2021; 13:polym13183169. [PMID: 34578069 PMCID: PMC8467615 DOI: 10.3390/polym13183169] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 09/15/2021] [Accepted: 09/16/2021] [Indexed: 12/24/2022] Open
Abstract
Rosmarinic acid (RA), one of the most important polyphenol-based antioxidants, has drawn increasing attention because of its remarkable bioactive properties, including anti-inflammatory, anticancer and antibacterial activities. The aim of this study was to synthesize and characterize RA-loaded silk fibroin nanoparticles (RA-SFNs) in terms of their physical–chemical features and composition, and to investigate their antitumor activity against human cervical carcinoma and breast cancer cell lines (HeLa and MCF-7). Compared with the free form, RA bioavailability was enhanced when the drug was adsorbed onto the surface of the silk fibroin nanoparticles (SFNs). The resulting particle diameter was 255 nm, with a polydispersity index of 0.187, and the Z-potential was −17 mV. The drug loading content of the RA-SFNs was 9.4 wt.%. Evaluation of the in vitro drug release of RA from RA-SFNs pointed to a rapid release in physiological conditions (50% of the total drug content was released in 0.5 h). Unloaded SFNs exhibited good biocompatibility, with no significant cytotoxicity observed during the first 48 h against HeLa and MCF-7 cancer cells. In contrast, cell death increased in a concentration-dependent manner after treatment with RA-SFNs, reaching an IC50 value of 1.568 and 1.377 mg/mL on HeLa and MCF-7, respectively. For both cell lines, the IC50 of free RA was higher. The cellular uptake of the nanoparticles studied was increased when RA was loaded on them. The cell cycle and apoptosis studies revealed that RA-SFNs inhibit cell proliferation and induce apoptosis on HeLa and MCF-7 cell lines. It is concluded, therefore, that the RA delivery platform based on SFNs improves the antitumor potential of RA in the case of the above cancers.
Collapse
|
15
|
Malekpour Z, Akbari V, Varshosaz J, Taheri A. Preparation and characterization of poly (lactic-co-glycolic acid) nanofibers containing simvastatin coated with hyaluronic acid for using in periodontal tissue engineering. Biotechnol Prog 2021; 37:e3195. [PMID: 34296538 DOI: 10.1002/btpr.3195] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 07/04/2021] [Accepted: 07/18/2021] [Indexed: 01/02/2023]
Abstract
Periodontal diseases can lead to soft tissue defects. Tissue engineering can provide functional replacements for damaged tissues. Recently, electrospun nanofibers have attracted great interest for tissue engineering and drug delivery applications. This has been revealed that statins exhibit positive impacts on the proliferation and regeneration of periodontal tissues. Electrospun simvastatin loaded poly (lactic-co-glycolic acid) (SIM-PLGA-NF) were prepared using electrospinning technique. Optimal conditions for preparation of SIM-PLGA-NF (PLGA concentration of 30 wt%, voltage of 15 kV, and flow rate of 1.5 ml h-1 ) were identified using a 23 factorial design. The optimized SIM-PLGA-NFs (diameter of 640.2 ± 32.5 nm and simvastatin entrapment efficacy of 99.6 ± 1.5%) were surface modified with 1% w/v hyaluronic acid solution (1%HA- SIM-PLGA-NF) to improve their compatibility with fibroblasts and potential application as a periodontal tissue engineering scaffold. HA-SIM-PLGA NFs were analyzed using SEM, FTIR, and XRD. 1%HA-SIM-PLGA-NF had uniform, bead-free and interwoven morphology, which is similar to the extracellular matrix. The mechanical performance of SIM-PLGA-NFs and release profile of simvastatin from these nanofibers have been also greatly improved after coating with HA. In vitro cellular tests showed that the proliferation, adhesion, and differentiation of fibroblast cells positively enhanced on the surface of 1%HA- SIM-PLGA-NF. These results demonstrate the potential application of 1%HA-SIM-PLGA-NFs as a scaffold for periodontal tissue engineering.
Collapse
Affiliation(s)
- Zahra Malekpour
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Vajihe Akbari
- Department of Pharmaceutical Biotechnology and Isfahan Pharmaceutical Research Center, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Jaleh Varshosaz
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Azade Taheri
- Novel Drug Delivery Systems Research Center, Department of Pharmaceutics, Faculty of Pharmacy, Isfahan University of Medical Sciences, Isfahan, Iran
| |
Collapse
|
16
|
Modulating surface charge of dexamethasone non-spherical microcrystals for improved inner ear delivery. Colloids Surf B Biointerfaces 2021; 204:111806. [PMID: 33957492 DOI: 10.1016/j.colsurfb.2021.111806] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 04/13/2021] [Accepted: 04/26/2021] [Indexed: 11/24/2022]
Abstract
It is important to achieve precise surface charge manipulation of non-spherical drug microcrystals using facile and time-efficient methods for local drug delivery. In this study, silk-coated dexamethasone (DEX) non-spherical microcrystals were synthesized by precipitation technique followed by alternate deposition of poly(allylamine hydrochloride) (PAH) (or PAH-coated Fe3O4) and silk fibroin (SF) via layer-by-layer assembly. EDC and glutaraldehyde were employed to manipulate positive or negative charge of particles by simple chemical cross-linking reactions, respectively. In vivo assessment was carried out by intratympanic (IT) injection of DEX non-spherical microcrystals in guinea pigs. In vivo pharmacokinetic results demonstrate that negatively charged DEX microcrystals appeared to improve outcomes of inner ear delivery in comparison to positively-charged counterparts. This is partly because of the adhesive features of the SF. The present study may provide new ideas to construct surface charge-tunable drug delivery vehicles that are capable of crossing biological barriers, especially for inner ear delivery due to the simple and practical strategy.
Collapse
|
17
|
Polymeric non-spherical coarse microparticles fabricated by double emulsion-solvent evaporation for simvastatin delivery. Colloids Surf B Biointerfaces 2021; 199:111560. [DOI: 10.1016/j.colsurfb.2021.111560] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 12/18/2020] [Accepted: 01/03/2021] [Indexed: 01/24/2023]
|
18
|
Poly(d,l lactic-co-glycolic) membranes seeded with human adipose stem cells for dermal substitute. Polym Bull (Berl) 2020. [DOI: 10.1007/s00289-020-03476-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
19
|
Woo SY, Lee SY, Yu SL, Park SJ, Kang D, Kim JS, Jeong IB, Kwon SJ, Hwang WJ, Park CR, Son JW. MicroRNA-7-5p's role in the O-GlcNAcylation and cancer metabolism. Noncoding RNA Res 2020; 5:201-207. [PMID: 33251387 PMCID: PMC7677666 DOI: 10.1016/j.ncrna.2020.11.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 11/03/2020] [Accepted: 11/05/2020] [Indexed: 01/27/2023] Open
Abstract
O-GlcNAc Transferase (OGT) is a complementary enzyme that regulates O-linked N-acetylglucosaminylation(O-GlcNAcylation) and plays a critical role in various cancer phenotypes, including invasion, migration, and metabolic reprogramming. In our previous study we found that miR-7-5p was downregulated at lung cancer cells with highly metastatic capacity. In the in-silico approach, OGT is the predicted target of miR-7-5p. To identify miR-7-5p′s role in cell growth and metabolism, we transfected various lung cancer cell lines with miR-7-5p. The expression level of miR-7-5p was confirmed by qRT-PCR in lung cancer cell lines. Western blot assays and qRT-PCR were performed to demonstrate miR-7-5p′s effect. Bioinformatic analysis indicated that OGT is a direct target of miR-7-5p. The binding sites of miR-7-5p in the OGT 3′ UTR were verified by luciferase reporter assay. To investigate the role of miR-7-5p in the cancer metabolism of non-small cell lung cancer (NSCLC) cells, mimic of miR-7-5p was transfected into NSCLC cells, and the effect of miR-7-5p on cancer metabolism was analyzed by LDH assays, glucose uptake, and mitochondrial ATP synthase inhibitor assay. O-GlcNAcylated protein level was determined by Western blot. The role of miR-7-5p in lung cancer growth was measured by MTS assays. To identify the delivery of miR-7-5p via PLGA, an in vitro release assay of PLGA-miR-7-5p was done. miR-7-5p was highly expressed whereas OGT showed low expression in H358, H827. However, miR-7-5p exhibited low expression while OGT had high expression in H522, H460, and H1299 cell lines. OGT were repressed by binding of miR-7a-5p to the 3′-UTR. Overexpression of miR-7-5p also diminished anaerobic glycolysis. miR-181a-5p transfection induced expression levels of OGT were diminished compared to those in the control group. O-GlcNAcylation was suppressed by miR-7-5p. Moreover, the overexpression of miR-7-5a suppressed lung cancer cell growth. miR-7-5p was released via PLGA for up to 10 days. In the present study, inhibition of OGT by miR-7-5p decreased the growth and cancer metabolism of lung cancer.
Collapse
Affiliation(s)
- Sin Yung Woo
- Department of Internal Medicine, Konyang University Hospital, South Korea
| | - Su Yel Lee
- Priority Research Center, Myunggok Research Institute, College of Medicine, Konyang University, South Korea
| | - Seong-Lan Yu
- Priority Research Center, Myunggok Research Institute, College of Medicine, Konyang University, South Korea
| | - Se Jin Park
- Department of Internal Medicine, Konyang University Hospital, South Korea
| | - Daeun Kang
- Department of Internal Medicine, Konyang University Hospital, South Korea
| | - Jin Suk Kim
- Department of Nuclear Medicine, Konyang University Hospital, South Korea
| | - In Beom Jeong
- Department of Internal Medicine, Konyang University Hospital, South Korea
| | - Sun Jung Kwon
- Department of Internal Medicine, Konyang University Hospital, South Korea
| | - Wan Jin Hwang
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, South Korea
| | - Chang Ryul Park
- Ulsan University Hospital, University of Ulsan College of Medicine, South Korea
| | - Ji Woong Son
- Department of Internal Medicine, Konyang University Hospital, South Korea
| |
Collapse
|
20
|
Sadeghi D, Solouk A, Samadikuchaksaraei A, Seifalian AM. Preparation of internally-crosslinked alginate microspheres: Optimization of process parameters and study of pH-responsive behaviors. Carbohydr Polym 2020; 255:117336. [PMID: 33436179 DOI: 10.1016/j.carbpol.2020.117336] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2020] [Revised: 10/25/2020] [Accepted: 10/27/2020] [Indexed: 02/07/2023]
Abstract
In this study, the effects of various parameters of the water-in-oil emulsification/internal gelation method on the properties of calcium-alginate microparticles were evaluated and optimized. Results showed that the spherical-shaped microparticles with the highest circularity and high production yield can be produced by alginate solution with a concentration of 2 wt.%, calcium carbonate/alginate ratio of 10/1 (w/w), water/oil volume ratio of 1/20, emulsifier concentration of 5 % (v/v), and emulsification speed of 1000 rpm. Two model drugs including simvastatin lactone and simvastatin β-hydroxyacid were loaded into the microspheres with promising encapsulation efficiencies of 73 % and 69 %, respectively. The microspheres showed a pH-responsive swelling behavior with a percentage of 10.60 %, 352.65 %, 690.03 %, and 1211.46 % at the pH values of 2.0, 4.5, 7.4, and 8.5, respectively. The microspheres showed an increasing trend of release rate in direct proportion to pH. These findings would be useful for therapeutic applications which need pH-responsive drug carriers.
Collapse
Affiliation(s)
- Davoud Sadeghi
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Atefeh Solouk
- Biomedical Engineering Department, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Ali Samadikuchaksaraei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran; Department of Tissue Engineering and Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran; Department of Medical Biotechnology, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Alexander M Seifalian
- Nanotechnology and Regenerative Medicine Centre (Ltd), London BioScience Innovation Centre, London, United Kingdom
| |
Collapse
|
21
|
Hierarchical porous silk fibroin/poly(L-lactic acid) fibrous membranes towards vascular scaffolds. Int J Biol Macromol 2020; 166:1111-1120. [PMID: 33159945 DOI: 10.1016/j.ijbiomac.2020.10.266] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Revised: 10/27/2020] [Accepted: 10/31/2020] [Indexed: 11/21/2022]
Abstract
Fibrous membranes played an important role to prepare tubular scaffolds for muscular artery regeneration. In this study, a strategy has been developed to combine silk fibroin (SF) with highly porous electrospun poly(L-lactic acid) (PLLA) fibrous membrane towards vascular scaffolds. After PLLA fibres were electrospun and collected, they were immersed into acetone to generate a porous structure with ultra-high surface area. While the pores on PLLA fibres were fulfilled with SF solution and dried, SF was coated uniformly and tightly on PLLA fibres. A multi-layer tubular structure of the tunica media was simulated by winding and stacking a strip of electrospun fibrous membrane. In vitro viability and morphology studies of A7r5 smooth muscle cells were undertaken for up to 14 days. Because the hydrophilicity of SF/PLLA composite fibres were improved dramatically, it had a positive effect on cell adhesion rate (97%) and proliferation (64.4%). Moreover, good cell morphology was observed via a multiphoton laser confocal microscope on SF/PLLA bioactive materials. These results demonstrated that the hierarchical porous SF/PLLA fibrous membranes are promising off-the-shelf scaffolds for muscular artery regeneration.
Collapse
|
22
|
Liu M, Feng D, Liang X, Li M, Yang J, Wang H, Pang L, Zhou Z, Yang Z, Kong D, Li C. Old Dog New Tricks: PLGA Microparticles as an Adjuvant for Insulin Peptide Fragment-Induced Immune Tolerance against Type 1 Diabetes. Mol Pharm 2020; 17:3513-3525. [PMID: 32787283 DOI: 10.1021/acs.molpharmaceut.0c00525] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Poly[lactic-co-(glycolic acid)] (PLGA) is arguably one of the most versatile synthetic copolymers used for biomedical applications. In vivo delivery of multiple substances including cells, pharmaceutical compounds, and antigens has been achieved by using PLGA-based micro-/nanoparticles although, presently, the exact biological impact of PLGA particles on the immune system remains controversial. Type 1 diabetes (T1D) is one subtype of diabetes characterized by the attack of immune cells against self-insulin-producing pancreatic islet cells. Considering the autoimmune etiology of T1D and the recent use of PLGA particles for eliciting desired immune responses in various aspects of immunotherapy, for the present study, a combination of Ins29-23 peptide (a known autoantigen of T1D) and PLGA microparticles was selected for T1D prevention assessment in nonobese diabetic (NOD) mice, a well-known animal model with spontaneous development of T1D. Thus, inoculation of PLGA microparticles + Ins29-23 completely prevented T1D development, significantly better than untreated controls and mice treated by either PLGA microparticles or Ins29-23 per se. Subsequent mechanistic investigation further revealed a facilitative role of PLGA microparticles in immune tolerance induction. In summary, our data demonstrate an adjuvant potential of PLGA microparticles in tolerance induction and immune remodulation for effective prevention of autoimmune diseases such as T1D.
Collapse
Affiliation(s)
- Mohan Liu
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Dandan Feng
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Xiaoyu Liang
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Min Li
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Jing Yang
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Hai Wang
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Liyun Pang
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Zhimin Zhou
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| | - Zhimou Yang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Centre of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Collaborative Innovation Centre of Chemical Science and Engineering, and National Institute of Functional Materials, Nankai University, Tianjin 300071, China
| | - Chen Li
- Tianjin Key Laboratory of Biomedical Materials, Biomedical Barriers Research Centre, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
23
|
Bi X, Li L, Mao Z, Liu B, Yang L, He W, Fan Y, Li X. The effects of silk layer-by-layer surface modification on the mechanical and structural retention of extracellular matrix scaffolds. Biomater Sci 2020; 8:4026-4038. [PMID: 32573617 DOI: 10.1039/d0bm00448k] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2025]
Abstract
Naturally derived extracellular matrix scaffolds can effectively promote tissue repair and regeneration due to their remarkable bioactivity. However, their rapid degradation leads to the decrease of mechanical retention and the failure of physical support in vivo which limit their applications. In this paper, we modified a classic extracellular matrix scaffold - small intestinal submucosa (SIS) - by a silk fibroin (SF) layer-by-layer (LbL) assembly to replace the existing chemical crosslinking methods for improving its mechanical and structural stability. Experimental results showed that the SF LbL surface functionalized SIS scaffold had tunable mechanical properties and degradation rate by adjusting the number of layers of the SF deposited on the surface. For biological responses, in vitro NIH3T3 fibroblast culture studies demonstrated that SF surface modification did not affect the excellent biocompatibility of the SIS. In vivo subcutaneous implantation results showed that the SF modification could effectively extend the residence time of the SIS in the body, and elicit a more moderate inflammatory response compared to the traditional glutaraldehyde chemical crosslinking. Furthermore, we found that SF modification could maintain the ability of bioactive components of the SIS to regulate the transformation of M1 into M2 in macrophages in vivo. This SF LbL modification strategy offers a green process for the development of high-performance extracellular matrix-based scaffolds with tunable biodegradability.
Collapse
Affiliation(s)
- Xuewei Bi
- Key Laboratory for Biomechanics and Mechanobiology of Ministry of Education, School of Biological Science and Medical Engineering, Beihang University, Beijing 100083, China.
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Zhang C, Zheng Y, Li M, Zhang Z, Chang L, Ai M, Wang J, Zhao S, Li C, Zhou Z. Carboxymethyl Cellulose-Coated Tacrolimus Nonspherical Microcrystals for Improved Therapeutic Efficacy of Dry Eye. Macromol Biosci 2020; 20:e2000079. [PMID: 32537876 DOI: 10.1002/mabi.202000079] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/28/2020] [Indexed: 01/08/2023]
Abstract
Dry eye (DE) is a highly prevalent ocular surface disease which affects the quality of life and results in low working efficiency. Frequent instillation is required due to low bioavailability of conventional eye drops. The aim of this study is to develop a novel formulation of tacrolimus (TAC), routinely prescribed for DE, by combination of the microcrystal technology and layer-by-layer assembly. First, nonspherical tacrolimus microcrystals (TAC MCs) are synthesized by antisolvent-induced precipitation. These TAC MCs are modified by alternate deposition of poly(allylamine hydrochloride) (PAH) and carboxymethyl cellulose (CMC) subsequently to obtain CMC-coated TAC MCs (TAC-(PAH/CMC)3 ). The resultant formulations are evaluated in vivo in a mouse DE model induced by an intelligently controlled environmental system. Compared with commercially available TAC eye drops and the TAC MCs counterpart, TAC-(PAH/CMC)3 exhibits superior therapeutic performance with reduced drug instillation frequency, which is attributed to the nonspherical geometry of MCs, the lubricant, mucoadhesive effect of CMC, and the anti-inflammatory function of TAC. Therefore, TAC-(PAH/CMC)3 represents a better option for the management of DE.
Collapse
Affiliation(s)
- Caijie Zhang
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | | | - Min Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| | - Zhongfang Zhang
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Lianqing Chang
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Mingyue Ai
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| | - Jingjie Wang
- State Key Laboratory of Ophthalmology, Optometry and Vision Science, School of Ophthalmology and Optometry, School of Biomedical Engineering, Wenzhou Medical University, Wenzhou, 325027, China
| | - Shaozhen Zhao
- Tianjin International Joint Research and Development Centre of Ophthalmology and Vision Science, Eye Institute and School of Optometry, Tianjin Medical University Eye Hospital, Tianjin, 300384, China
| | - Chen Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin, 300192, China
| |
Collapse
|
25
|
Improving Anticancer Therapy with Naringenin-Loaded Silk Fibroin Nanoparticles. NANOMATERIALS 2020; 10:nano10040718. [PMID: 32290154 PMCID: PMC7221656 DOI: 10.3390/nano10040718] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/07/2020] [Accepted: 04/09/2020] [Indexed: 12/12/2022]
Abstract
Naringenin (NAR), a flavonoid present in a variety of fruits, vegetables and herbs, exhibits a wide range of pharmacological effects, including anticancer activity. Nevertheless, its application in cancer therapy is limited due to its low bioavailability at the tumour site because of its poor solubility in water and slow dissolution rate. To improve the therapeutic efficacy of NAR, emergent research is looking into using nanocarriers. Silk fibroin (SF), from the Bombyx mori silkworm, is a biocompatible and biodegradable polymer with excellent mechanical properties and an amphiphilic chemistry that make it a promising candidate as a controlled release drug system. The aim of this work is to synthesize naringenin-loaded silk fibroin nanoparticles (NAR-SFNs) by dissolving the SF in the ionic liquid 1-ethyl-3-methylimidazolium acetate, using high-power ultrasounds and rapid desolvation in methanol followed by the adsorption of NAR. The NAR-SFNs were characterized by dynamic light scattering, Fourier transform infrared spectroscopy and thermogravimetric analysis. The drug loading content and encapsulation efficiency were calculated. The drug release profile best fitted a first order equation. The cytotoxicity effects of free NAR, bare silk fibroin nanoparticles (SFNs) and NAR-SFNs were assessed on HeLa and EA.hy926 cells via 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The results demonstrated the higher in vitro anticancer potential of synthesized NAR-SFNs than that of free NAR in HeLa cancer cells.
Collapse
|
26
|
Zhang L, Zhang Y, Du Y, Wang J, Chi L. RETRACTED: Development of curcumin-loaded silk fibroin nanoparticles as drug delivery vehicle for the treatment of ischemic stroke for patients in nursing care in hospitals. J Drug Deliv Sci Technol 2020. [DOI: 10.1016/j.jddst.2019.101360] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
27
|
Li Y, Zhang Q, Xie X, Xiao D, Lin Y. Review of craniofacial regeneration in China. J Oral Rehabil 2019; 47 Suppl 1:107-117. [PMID: 30868603 DOI: 10.1111/joor.12793] [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: 01/13/2019] [Revised: 02/28/2019] [Accepted: 03/09/2019] [Indexed: 02/05/2023]
Abstract
AIM Tissue engineering has been recognised as one of the most effective means to form a new viable tissue for medical purpose. Tissue engineering involves a combination of scaffolds, cells, suitable biochemical and physicochemical factors, and engineering and materials methods. This review covered some biomedicine, such as biomaterials, bioactive factors, and stem cells, and manufacturing technologies used in tissue engineering in the oral maxillofacial region, especially in China. MATERIALS AND METHODS Data for this review were identified by searches of Web of Science and PubMed, and references from relevant articles using the search terms "biomaterials", "oral tissue regeneration", "bioactive factors" and "stem cells". Only articles published in English between 2013 and 2018 were included. CONCLUSION The combination of stem cells, bioactive factors and 3D scaffolds could be of far-reaching significance for the future therapies in tissue repair or tissue regeneration. Furthermore, the review also mentions issues that need to be solved in the application of these biomedicines.
Collapse
Affiliation(s)
- Yanjing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Qi Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xueping Xie
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Dexuan Xiao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yunfeng Lin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
28
|
A novel synthesis of selenium nanoparticles encapsulated PLGA nanospheres with curcumin molecules for the inhibition of amyloid β aggregation in Alzheimer's disease. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2019; 190:98-102. [DOI: 10.1016/j.jphotobiol.2018.11.008] [Citation(s) in RCA: 125] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Revised: 11/12/2018] [Accepted: 11/13/2018] [Indexed: 12/19/2022]
|
29
|
Zhang ZZ, Zhang HZ, Zhang ZY. 3D printed poly(ε-caprolactone) scaffolds function with simvastatin-loaded poly(lactic-co-glycolic acid) microspheres to repair load-bearing segmental bone defects. Exp Ther Med 2018; 17:79-90. [PMID: 30651767 PMCID: PMC6307523 DOI: 10.3892/etm.2018.6947] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2018] [Accepted: 08/09/2018] [Indexed: 12/21/2022] Open
Abstract
Repairing critical-sized bone defects has been a major challenge for orthopedic surgeons in the clinic. The generation of functioning bone tissue scaffolds using osteogenic induction factors is a promising method to facilitate bone healing. In the present study, three-dimensional (3D) printing of a poly(lactic-co-glycolic acid) (PLGA) scaffold with simvastatin (SIM) release functioning was generated by rapid prototyping, which was incorporated with collagen for surface activation, and was finally mixed with SIM-loaded PLGA microspheres. In vitro assays with bone marrow-derived mesenchymal stem cells were conducted. For the in vivo study, scaffolds were implanted into segmental defects created on the femurs of Sprague-Dawley rats. At 4 and 12 weeks following surgery, X-ray, micro-computed tomography and histological analysis were performed in order to evaluate bone regeneration. The results demonstrated that collagen functionalization of PLGA produced better cell adhesion, while the sustained release of SIM promoted greater cell proliferation with no significant cytotoxicity, compared with the blank PCL scaffold. Furthermore, in vivo experiments also confirmed that SIM-loaded scaffolds played a significant role in promoting bone regeneration. In conclusion, the present study successfully manufactured a 3D printing PLGA scaffold with sustained SIM release, which may meet the requirements for bone healing, including good mechanical strength and efficient osteoinduction ability. Thus, the results are indicative of a promising bone substitute to be used in the clinic.
Collapse
Affiliation(s)
- Zhan-Zhao Zhang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Hui-Zhong Zhang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| | - Zhi-Yong Zhang
- Shanghai Key Laboratory of Tissue Engineering, Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, P.R. China
| |
Collapse
|
30
|
Bee SL, Hamid ZAA, Mariatti M, Yahaya BH, Lim K, Bee ST, Sin LT. Approaches to Improve Therapeutic Efficacy of Biodegradable PLA/PLGA Microspheres: A Review. POLYM REV 2018. [DOI: 10.1080/15583724.2018.1437547] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Affiliation(s)
- Soo-Ling Bee
- School of Materials and Mineral Resources Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Penang, Malaysia
| | - Z. A. Abdul Hamid
- School of Materials and Mineral Resources Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Penang, Malaysia
| | - M. Mariatti
- School of Materials and Mineral Resources Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Penang, Malaysia
| | - B. H. Yahaya
- Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Penang, Malaysia
| | - Keemi Lim
- School of Materials and Mineral Resources Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Penang, Malaysia
| | - Soo-Tueen Bee
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang, Selangor, Malaysia
| | - Lee Tin Sin
- Department of Chemical Engineering, Lee Kong Chian Faculty of Engineering and Science, Universiti Tunku Abdul Rahman, Jalan Sungai Long, Bandar Sungai Long, Cheras, Kajang, Selangor, Malaysia
| |
Collapse
|
31
|
Montalbán MG, Coburn JM, Lozano-Pérez AA, Cenis JL, Víllora G, Kaplan DL. Production of Curcumin-Loaded Silk Fibroin Nanoparticles for Cancer Therapy. NANOMATERIALS (BASEL, SWITZERLAND) 2018; 8:E126. [PMID: 29495296 PMCID: PMC5853757 DOI: 10.3390/nano8020126] [Citation(s) in RCA: 129] [Impact Index Per Article: 18.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/27/2018] [Revised: 02/16/2018] [Accepted: 02/22/2018] [Indexed: 12/19/2022]
Abstract
Curcumin, extracted from the rhizome of Curcuma longa, has been widely used in medicine for centuries due to its anti-inflammatory, anti-cancer, anti-oxidant and anti-microbial effects. However, its bioavailability during treatments is poor because of its low solubility in water, slow dissolution rate and rapid intestinal metabolism. For these reasons, improving the therapeutic efficiency of curcumin using nanocarriers (e.g., biopolymer nanoparticles) has been a research focus, to foster delivery of the curcumin inside cells due to their small size and large surface area. Silk fibroin from the Bombyx mori silkworm is a biopolymer characterized by its biocompatibility, biodegradability, amphiphilic chemistry, and excellent mechanical properties in various material formats. These features make silk fibroin nanoparticles useful vehicles for delivering therapeutic drugs, such as curcumin. Curcumin-loaded silk fibroin nanoparticles were synthesized using two procedures (physical adsorption and coprecipitation) more scalable than methods previously described using ionic liquids. The results showed that nanoparticle formulations were 155 to 170 nm in diameter with a zeta potential of approximately -45 mV. The curcumin-loaded silk fibroin nanoparticles obtained by both processing methods were cytotoxic to carcinogenic cells, while not decreasing viability of healthy cells. In the case of tumor cells, curcumin-loaded silk fibroin nanoparticles presented higher efficacy in cytotoxicity against neuroblastoma cells than hepatocarcinoma cells. In conclusion, curcumin-loaded silk fibroin nanoparticles constitute a biodegradable and biocompatible delivery system with the potential to treat tumors by local, long-term sustained drug delivery.
Collapse
Affiliation(s)
- Mercedes G. Montalbán
- Department of Chemical Engineering, Faculty of Chemistry, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, 30071 Murcia, Spain;
| | - Jeannine M. Coburn
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; (J.M.C.); (D.L.K.)
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA 01609, USA
| | - A. Abel Lozano-Pérez
- Department of Biotechnology, Instituto Murciano de Investigación y Desarrollo Agrario y Alimentario (IMIDA), La Alberca, 30150 Murcia, Spain; (A.A.L.-P.); (J.L.C.)
| | - José L. Cenis
- Department of Biotechnology, Instituto Murciano de Investigación y Desarrollo Agrario y Alimentario (IMIDA), La Alberca, 30150 Murcia, Spain; (A.A.L.-P.); (J.L.C.)
| | - Gloria Víllora
- Department of Chemical Engineering, Faculty of Chemistry, Regional Campus of International Excellence “Campus Mare Nostrum”, University of Murcia, 30071 Murcia, Spain;
| | - David L. Kaplan
- Department of Biomedical Engineering, Tufts University, Medford, MA 02155, USA; (J.M.C.); (D.L.K.)
| |
Collapse
|
32
|
Zhang T, Li M, Wang X, Zhou Z, Yuan W, Ma J. Facile synthesis of polylactide coarse microspheres as artificial antigen-presenting cells. Chem Commun (Camb) 2018; 54:11356-11359. [DOI: 10.1039/c8cc04958k] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Magnetic poly(l-lactide) coarse microspheres as artificial antigen-presenting cells were synthesized via simple chemical etching and antibody immobilization.
Collapse
Affiliation(s)
- Tong Zhang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| | - Min Li
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials
- Tianjin
- China
| | - Xiaotong Wang
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| | - Zhimin Zhou
- Biomedical Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin Key Laboratory of Biomedical Materials
- Tianjin
- China
| | - Wei Yuan
- State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| | - Jie Ma
- Department of Biotherapy, Beijing Hospital, National Center of Gerontology, Chinese Academy of Medical Sciences & Peking Union Medical College
- Beijing
- China
| |
Collapse
|