纺织学报 ›› 2023, Vol. 44 ›› Issue (09): 213-222.doi: 10.13475/j.fzxb.20220500202

• 综合述评 • 上一篇    下一篇

丝素蛋白基药物载体的应用研究进展

罗元泽1, 戴梦男1, 李蒙1, 俞杨销1, 王建南1,2()   

  1. 1.苏州大学 纺织与服装工程学院, 江苏 苏州 215123
    2.苏州大学 纺织行业医疗健康用蚕丝制品重点实验室, 江苏 苏州 215123
  • 收稿日期:2022-05-05 修回日期:2022-10-12 出版日期:2023-09-15 发布日期:2023-10-30
  • 通讯作者: 王建南(1970—),女,教授,博士。主要研究方向为生物医用材料、生物医用纺织品和功能纺织品。E-mail:wangjn@suda.edu.cn
  • 作者简介:罗元泽(1998—),男,硕士。主要研究方向为生物医用材料。
  • 基金资助:
    国家自然科学基金项目(51873141)

Application of silk fibroin-based biomaterials for drug delivery

LUO Yuanze1, DAI Mengnan1, LI Meng1, YU Yangxiao1, WANG Jiannan1,2()   

  1. 1. College of Textile and Clothing Engineering, Soochow University, Suzhou, Jiangsu 215123, China
    2. Key Laboratory of Silk Products for Medical and Health Use in Textile Industry,;Soochow University, Suzhou, Jiangsu 215123, China
  • Received:2022-05-05 Revised:2022-10-12 Published:2023-09-15 Online:2023-10-30

摘要:

为拓展和推动丝素蛋白材料在医药领域的临床应用,综述了近年来丝素蛋白在药物递送系统中的最新研究进展,重点阐述了丝素蛋白药物递送载体的种类、制备方法、主要的药物负载类型及其应用性能。通过分析发现:丝素蛋白除具有出色的生物相容性和可控降解性外,其大分子独特的亲疏水链结构还赋予丝素蛋白聚集态结构自组装的调控基础;以丝素蛋白为基材所构建的微球、水凝胶和微针等多种形貌的药物载体表现出高效的药物负载能力和可控的释放速率,从而达到患者减少服药次数和提高治疗效率的目的。除负载小分子化学药物,丝素蛋白载体还可改善蛋白和基因类药物易降解和半衰期短的缺点。由于丝素蛋白的这些优异特性,将其用于研制靶向与特异性药物载体可实现对不同类型疾病的针对性治疗,从而提高药物利用度和降低副作用。最后指出使用丝素蛋白作为药物载体存在的问题和重要的发展方向。

关键词: 丝素蛋白, 药物载体, 生物医药制品, 可控释放, 应用性能

Abstract:

Significance With the increasing incidence of chronic noncommunicable diseases (e.g. cancer), inflammation and diabetes, drug delivery systems with controlled release are developed and applied in clinical practice. Silk fibroin was studied widely for applications in the fields of tissue engineering and medicine because of its good biocompatibility and biodegradability. The research on the application of silk fibroin as drug carriers attracted much global attention. In order to expand and promote the clinical applications of silk fibroin materials in pharmaceutical field, this paper reviews the latest research progress of silk fibroin in drug delivery systems, with highlights on carrier types, preparation methods, drug loading types and application properties of silk fibroin drug delivery systems.

Progress As a controlled drug carrier, silk fibroin is an ideal candidate because of its unique chemical structure and aggregated structure, and its excellent biocompatibility and controllable degradation. Silk fibroin materials can be prepared into various forms, among which silk fibroin materials in the form of microspheres, hydrogels and microneedles exhibit efficient drug loading capacity and controllable release rate when used as drug carriers, resulting in the decrease of dosing frequency and improvement of the therapeutic efficiency. Silk fibroin-based drug delivery system not only can stably encapsulate various small molecule compounds, but also deliver biological macromolecules such as proteins and nucleic acids. Composite drug carriers developed by combining other materials can reduce the degradation of silk fibroin drug carriers in vivo and improve the drug availability. Furthermore, the alternating arrangement of hydrophilic and hydrophobic chains of the silk fibroin macromolecule provides a structure basis for regulating the molecular conformation and aggregation structure of silk fibroin materials. The crystal form and crystallinity of silk fibroin can be regulated by physical or chemical modification to control drug release or endow targeting ability to diseased cells, which can effectively improve the therapeutic efficiency of serious diseases and reduce the damage to normal tissue cells.

Conclusion and Prospect With the continuous exploration of the biological function of silk fibroin, some progress in silk fibroin-based drug release materials has been made in association with drug delivery forms, targeted therapy and drug availability. Silk fibroin as a drug carrier has been found to prolong the circulation time of drugs in the blood, reduce the frequency of drug use and alleviate adverse drug reactions of patients. These advantages provide important guidance for in-depth and sustainable development of silk fibroin in drug delivery application. However, there are still problems in the stability of the preparation of silk fibroin drug carriers and their drug loading capacity. The quality of silk fibroin varies greatly due to the different sources, such as varieties, seasons and regions. It is necessary to standardize the characteristics and extraction method of silk fibroin. Although silk fibroin drug carriers can be endowed with the capability of targeted drug delivery by physical or chemical modification, it may be attacked by the immune system at the initial stage of entering the body, leading to failure of targeted release. The accuracy and effectiveness of silk fibroin used as drug carriers still need long-term exploration and clinical trials. With the development of material science, chemical science and pharmacy, the research and application of silk fibroin-based drug delivery systems will be further continued.

Key words: silk fibroin, drug carrier, biomedical product, controlled release, application performance

中图分类号: 

  • TS101.4

图1

由蚕丝开发的各种形态的药物递送系统"

表1

丝素蛋白药物载体的制备方法"

载体形式 制备方法
纳微球 自组装法[30]、喷雾干燥法[35]、溶剂蒸发法[36]、电场调控法[40]、乳化扩散法[41]、盐析法[42]
水凝胶 超声波法[37]、化学交联法[2]、冻融法[38]
微针 模具法[39]、打印法[43]
铸造法[44]、沉积法[45]、静电纺丝法[46]
多孔材料 冷冻干燥法[34]
涂层 涂覆法[32]

图2

丝素蛋白在药物递送载体中的应用"

图3

二氧化硅纳米粒子稳定的乳状液和丝素蛋白微胶囊的形成过程及其在输送系统中的应用示意图"

[1] LIAN X J, XU R, LIU S C, et al. The preparation and study on properties of calcium sulfate bone cement combined tuning silk fibroin nanofibers and vancomycin-loaded silk fibroin microspheres[J]. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 2022, 110(3): 564-572.
doi: 10.1002/jbm.b.v110.3
[2] WANG Y Y, NIU C Q, SHI J, et al. A carbodiimide cross-linked silk fibroin/sodium alginate composite hydrogel with tunable properties for sustained drug delivery[J]. Macromolecular Materials and Engineering, 2021. DOI: 10.1002/mame.202100470.
[3] LI H L, SONG G Z, TIAN W, et al. Motility and function of smooth muscle cells in a silk small-caliber tubular scaffold after replacement of rabbit common carotid artery[J]. Materials Science and Engineering: C, 2020. DOI: 10.1016/j.msec.2020.110977.
[4] WANG Y F, CHENG Q Q, LIU J, et al. Tuning microcapsule shell thickness and structure with silk fibroin and nanoparticles for sustained release[J]. ACS Biomaterials Science & Engineering, 2020, 6(8): 4583-4594.
[5] SHEN H D, WANG P, HAN X K, et al. GOx/Hb cascade oxidized crosslinking of silk fibroin for tissue-responsive wound repair[J]. Gels, 2022. DOI: 10.3390/gels8010056.
[6] WANG K L, WANG X Y, JIANG D, et al. Delivery of mRNA vaccines and anti-PDL1 siRNA through non-invasive transcutaneous route effectively inhibits tumor growth[J]. Composites Part B: Engineering, 2022. DOI: 10.1016/j.compositesb.2022.109648.
[7] GUPTA V, ASEH A, RIOS C N, et al. Fabrication and characterization of silk fibroin-derived curcumin nanoparticles for cancer therapy[J]. International Journal of Nanomedicine, 2009, 4(1): 115-122.
[8] 于丽. 阿司匹林丝素微球的制备及红花丝素微球的初步研究[D]. 济南: 山东中医药大学, 2012:19-42.
YU Li. Studies on preparation of aspirin silk fibroin microspheres and silk fibroin microspheres of saf-flower[D]. Ji'nan: Shandong University of Traditional Chinese Medicine, 2012:19-42.
[9] GHAURI Z H, ISLAM A, QADIR M A, et al. Novel pH-responsive chitosan/sodium alginate/PEG based hydrogels for release of sodium ceftriaxone[J]. Materials Chemistry and Physics, 2022. DOI: 10.1016/j.matchemphys.2021.125456.
[10] KUZUHARA A, ASAKURA T, TOMODA R, et al. Use of silk fibroin for enzyme membrane[J]. Journal of Biotechnology, 1987, 5(3): 199-207.
doi: 10.1016/0168-1656(87)90016-2
[11] GOU S Q, YANG J, MA Y, et al. Multi-responsive nanococktails with programmable targeting capacity for imaging-guided mitochondrial phototherapy combined with chemotherapy[J]. Journal of Controlled Release, 2020, 327: 371-383.
doi: S0168-3659(20)30454-5 pmid: 32810527
[12] MAMNOON B, FENG L, FROBERG J, et al. Targeting estrogen receptor-positive breast microtumors with endoxifen-conjugated, hypoxia-sensitive polymer-somes[J]. ACS Omega, 2021, 6(42): 27654-27667.
doi: 10.1021/acsomega.1c02250
[13] ZHANG R, JIANG Y Y, HAO L K, et al. CD44/folate dual targeting receptor reductive response PLGA-based micelles for cancer therapy[J]. Frontiers in Pharmacology, 2022. DOI: 10.3389/fphar.2022.829590.
[14] LIN M X, LIU Y, GAO J W, et al. Synergistic effect of co-delivering ciprofloxacin and tetracycline hydrochloride for promoted wound healing by utilizing coaxial PCL/gelatin nanofiber membrane[J]. International Journal of Molecular Sciences, 2022. DOI: 10.3390/ijms23031895.
[15] BHATTACHARJEE A, PURKAIT M K, SASTRI C V, et al. CeO2 nanoparticles incorporated MIL-100(Fe) composites for loading of an anticancer drug: effects of HF in composite synthesis and drug loading capacity[J]. Inorganica Chimica Acta, 2022. DOI: 10.1016/j.ica.2021.120784.
[16] RAILEANU M, LONETTI B, SERPENTINI C, et al. Encapsulation of a cationic antimicrobial peptide into self-assembled polyion complex nano-objects enhances its antitumor properties[J]. Journal of Molecular Structure, 2022. DOI: 10.1016/j.molstruc.2021.131482.
[17] THANGAVEL K, LAKSHMIKUTTYAMMA A, THANGAVEL C, et al. CD44-targeted, indocyanine green-paclitaxel-loaded human serum albumin nanoparticles for potential image-guided drug delivery[J]. Colloids and Surfaces B: Biointerfaces, 2022. DOI: 10.1016/j.colsurfb.2021.112162.
[18] FARIA J, DIONISIO B, SOARES I, et al. Cellulose acetate fibres loaded with daptomycin for metal implant coatings[J]. Carbohydrate Polymers, 2022. DOI: 10.1016/j.carbpol.2021.118733.
[19] WANG Y L, CHEN L X, ZHANG Z L, et al. Ratiometric co-delivery of doxorubicin and paclitaxel prodrug by remote-loading liposomes for the treatment of triple-negative breast cancer[J]. Drug Delivery and Translational Research, 2022, 12(10): 2537-2549.
doi: 10.1007/s13346-021-01105-2 pmid: 35043372
[20] MARIADOSS A V A, SARAVANAKUMAR K, SATHIYASEELAN A, et al. Smart drug delivery of p-coumaric acid loaded aptamer conjugated starch nanoparticles for effective triple-negative breast cancer therapy[J]. International Journal of Biological Macromolecules, 2022, 195: 22-29.
doi: 10.1016/j.ijbiomac.2021.11.170
[21] LONG L Y, LIU W Q, LI L, et al. Dissolving microneedle-encapsulated drug-loaded nanoparticles and recombinant humanized collagen type III for the treatment of chronic wound via anti-inflammation and enhanced cell proliferation and angiogenesis[J]. Nanoscale, 2022, 14(4): 1285-1295.
doi: 10.1039/D1NR07708B
[22] KARIMZADEH Z, NAMAZI H. Nontoxic double-network polymeric hybrid aerogel functionalized with reduced graphene oxide: preparation, characterization, and evaluation as drug delivery agent[J]. Journal of Polymer Research, 2022. DOI: 10.1007/s10965-022-02902-0.
[23] PRITCHARD E M, SZYBALA C, BOISON D, et al. Silk fibroin encapsulated powder reservoirs for sustained release of adenosine[J]. Journal of Controlled Release, 2010, 144(2): 159-167.
doi: 10.1016/j.jconrel.2010.01.035 pmid: 20138938
[24] PERVAIZ F, TANVEER W, SHOUKAT H, et al. Formulation and evaluation of polyethylene glycol/xanthan gum-co-poly (acrylic acid) interpenetrating network for controlled release of venlafaxine[J]. Polymer Bulletin, 2022. DOI: 10.1007/s00289-022-04098-1.
[25] PERERA W P T D, DISSANAYAKE D M R K, UNAGOLLA J M, et al. Albumin grafted coaxial electrosparyed polycaprolactone-zinc oxide nanoparticle for sustained release and activity enhanced antibacterial drug delivery[J]. RSC Advances, 2022, 12(3): 1718-1727.
doi: 10.1039/d1ra07847j pmid: 35425191
[26] SATHISHKUMAR P, LI Z F, GOVINDAN R, et al. Zinc oxide-quercetin nanocomposite as a smart nano-drug delivery system: molecular-level interaction studies[J]. Applied Surface Science, 2021. DOI: 10.1016/j.apsusc.2020.147741.
[27] XU J J, WANG Y N, DING M Y, et al. Sequence-structure characterization of recombinant polypeptides derived from silk fibroin heavy chain[J]. Materials Science and Engineering: C, 2020. DOI: 10.1016/j.msec.2020.110831.
[28] WU J B, XIE X S, ZHENG Z Z, et al. Effect of pH on polyethylene glycol (PEG)-induced silk microsphere formation for drug delivery[J]. Materials Science & Engineering C:Materials for Biological Applications, 2017. DOI: 10.1016/j.msec.2017.05.072.
[29] BARI E, SERRA M, PAOLILLO M, et al. Silk fibroin nanoparticle functionalization with Arg-Gly-Asp cyclopentapeptide promotes active targeting for tumor site-specific delivery[J]. Cancers, 2021. DOI: 10.3390/cancers13051185.
[30] YANG W H, YU S Y, CHEN S, et al. Doxorubicin-loaded silk fibroin nanospheres[J]. Acta Chimica Sinica, 2014, 72(11): 1164-1168.
doi: 10.6023/A14080596
[31] LU X Y, SUN Y Y, HAN M S, et al. Silk fibroin double-layer microneedles for the encapsulation and controlled release of triptorelin[J]. International Journal of Pharmaceutics, 2022. DOI: 10.1016/j.ijpharm.2021.121433.
[32] LI S T, SHI X W, XU B, et al. In vitro drug release and antibacterial activity evaluation of silk fibroin coated vancomycin hydrochloride loaded poly (lactic-co-glycolic acid) (PLGA) sustained release micro-spheres[J]. Journal of Biomaterials Applications, 2022, 36(9): 1676-1688.
doi: 10.1177/08853282211064098
[33] SHI Y F, HUANG L L, WANG X, et al. Intelligent drug delivery system based on silk fibroin/wool Skeratin[J]. Mathematical Problems in Engineering, 2022. DOI: 10.1155/2022/6748645.
[34] LI D, CHEN K, DUAN L, et al. Strontium ranelate incorporated enzyme-cross-linked gelatin nanoparticle/silk fibroin aerogel for osteogenesis in OVX-induced osteoporosis[J]. ACS Biomaterials Science & Engineering, 2019, 5(3): 1440-1451.
[35] KIM S Y, NASKAR D, KUNDU S C, et al. Formulation of biologically-inspired silk-based drug carriers for pulmonary delivery targeted for lung cancer[J]. Scientific Reports, 2015. DOI: 10.1038/srep11878.
[36] WANG X Q, WENK E, ZHANG X H, et al. Growth factor gradients via microsphere delivery in biopolymer scaffolds for osteochondral tissue engineering[J]. Journal of Controlled Release, 2009, 134(2): 81-90.
doi: 10.1016/j.jconrel.2008.10.021 pmid: 19071168
[37] AKRAMI-HASAN-KOHAL M, ESKANDARI M, SOLOUK A. Silk fibroin hydrogel/dexamethasone sodium phosphate loaded chitosan nanoparticles as a potential drug delivery system[J]. Colloids and Surfaces B: Biointerfaces, 2021. DOI: 10.1016/j.colsurfb.2021.111892.
[38] ZHAO D, NUNTANARANONT T, THUAKSUBUN N, et al. Osteo-conductive hydrogel scaffolds of poly(vinyl-alcohol) with silk fibroin particles for bone augmentation: structural formation and in vitro testing[J]. Journal of Bioactive and Compatible Polymers, 2021, 36(6): 481-496.
doi: 10.1177/08839115211055720
[39] TSIORIS K, RAJA W K, PRITCHARD E M, et al. Fabrication of silk microneedles for controlled-release drug delivery[J]. Advanced Functional Materials, 2012, 22(2): 330-335.
doi: 10.1002/adfm.v22.2
[40] QU J, WANG L, NIU L X, et al. Porous silk fibroin microspheres sustainably releasing bioactive basic fibroblast growth factor[J]. Materials, 2018. DOI: 10.3390/ma11081280.
[41] HAN Q, ZHENG T T, ZHANG L H, et al. Metformin loaded injectable silk fibroin microsphere for the treatment of spinal cord injury[J]. Journal of Biomaterials Science Polymer Edition, 2021, 33(6):747-768.
doi: 10.1080/09205063.2021.2014113
[42] ZHANG H Y, MA X L, CAO C B, et al. Multifunctional iron oxide/silk-fibroin (Fe3O4-SF) composite microspheres for the delivery of cancer therapeutics[J]. RSC Advances, 2014, 4(78): 41572-41577.
doi: 10.1039/C4RA05919K
[43] SHIN D, HYUN J. Silk fibroin microneedles fabricated by digital light processing 3D printing[J]. Journal of Industrial and Engineering Chemistry, 2021, 95: 126-133.
doi: 10.1016/j.jiec.2020.12.011
[44] KOPP A, SCHUNCK L, GOSAU M, et al. Influence of the casting concentration on the mechanical and optical properties of FA/CaCl2-derived silk fibroin mem-branes[J]. International Journal of Molecular Sciences, 2020. DOI: 10.3390/ijms21186704.
[45] MOE Y M, NUNTANARANONT T, KHANGKHAMANO M, et al. Layer-by-layer particle deposited membranes of silk fibroin and poly (vinyl alcohol) for guided bone regeneration: molecular structure, morphology, and properties[J]. Materials Technology, 2020, 37(5): 332-344.
doi: 10.1080/10667857.2020.1842149
[46] WANG Y Q, YAO D Y, LI L H, et al. Effect of electrospun silk fibroin-silk sericin films on macrophage polarization and vascularization[J]. ACS Biomaterials Science & Engineering, 2020, 6(6): 3502-3512.
[47] QIN J W, JIANG Y Y, FU J J, et al. Evaluation of drug release property and blood compatibility of aspirin-loaded electrospun PLA/RSF composite nanofibers[J]. Iranian Polymer Journal, 2013, 22(10): 729-737.
doi: 10.1007/s13726-013-0171-1
[48] SEIB F P, JONES G T, RNJAK-KOVACINA J, et al. PH-Dependent anticancer drug release from silk nanoparticles[J]. Advanced Healthcare Materials, 2013, 2(12): 1606-1611.
doi: 10.1002/adhm.201300034 pmid: 23625825
[49] MOTTAGHITALAB F, KIANI M, FAROKHI M, et al. Targeted delivery system based on gemcitabine-loaded silk fibroin nanoparticles for lung cancer therapy[J]. ACS Applied Materials & Interfaces, 2017, 9(37): 31600-31611.
[50] GHAREHNAZIFAM Z, DOLATABADI R, BANIASSADI M, et al. Computational analysis of vincristine loaded silk fibroin hydrogel for sustained drug delivery applications: multiphysics modeling and experiments[J]. International Journal of Pharmaceutics, 2021. DOI: 10.1016/j.ijpharm.2021.121184.
[51] XIE M B, LI Y, ZHAO Z, et al. Solubility enhancement of curcumin via supercritical CO2 based silk fibroin carrier[J]. Journal of Supercritical Fluids, 2015, 103: 1-9.
doi: 10.1016/j.supflu.2015.04.021
[52] SUBIA B, CHANDRA S, TALUKDAR S, et al. Folate conjugated silk fibroin nanocarriers for targeted drug delivery[J]. Integrative Biology, 2014, 6(2): 203-214.
doi: 10.1039/C3IB40184G
[53] PILCH J, KOWALIK P, KOWALCZYK A, et al. Foliate-targeting quantum dots-β-cyclodextrin nanocarrier for efficient delivery of unsymmetrical bisacridines to lung and prostate cancer cells[J]. International Journal of Molecular Sciences, 2022. DOI: 10.3390/ijms23031261.
[54] ZHANG X X, ZHANG J Y, SHI B. Mesoporous bioglass/silk fibroin scaffolds as a drug delivery system: fabrication, drug loading and release in vitro and repair calvarial defects in vivo[J]. Journal of Wuhan University of Technology-Materials Science Edition, 2014, 29(2): 401-406.
doi: 10.1007/s11595-014-0929-0
[55] FAROKHI M, MOTTAGHITALAB F, SHOKRGOZAR M A, et al. Bio-hybrid silk fibroin/calcium phosphate/PLGA nanocomposite scaffold to control the delivery of vascular endothelial growth factor[J]. Materials Science & Engineering C:Materials for Biological Applications, 2014, 35: 401-410.
[56] WANG X Q, WENK E, MATSUMOTO A, et al. Silk microspheres for encapsulation and controlled release[J]. Journal of Controlled Release, 2007, 117(3): 360-370.
doi: 10.1016/j.jconrel.2006.11.021 pmid: 17218036
[57] ZHANG Y Q, WANG Y J, WANG H Y, et al. Highly efficient processing of silk fibroin nanoparticle-L-asparaginase bioconjugates and their characterization as a drug delivery system[J]. Soft Matter, 2011, 7(20): 9728-9736.
[58] XIAO M L, LÜ S S. Self-assembled regenerated silk fibroin microsphere-embedded Fe3O4 magnetic nanoparticles for immobilization of zymolyase[J]. ACS Omega, 2019, 4(25): 21612-21619.
doi: 10.1021/acsomega.9b03491
[59] TEUSCHL A H, ZIPPERLE J, HUBER-GRIES C, et al. Silk fibroin based carrier system for delivery of fibrinogen and thrombin as coagulant supplements[J]. Journal of Biomedical Materials Research:Part A, 2017, 105(3): 687-696.
doi: 10.1002/jbm.v105.3
[60] NUMATA K, HAMASAKI J, SUBRAMANIAN B, et al. Gene delivery mediated by recombinant silk proteins containing cationic and cell binding motifs[J]. Journal of Controlled Release, 2010, 146(1): 136-143.
doi: 10.1016/j.jconrel.2010.05.006 pmid: 20457191
[61] MA C L, LÜ L L, LIU Y, et al. Antheraea pernyi silk fibroin for targeted gene delivery of VEGF165-Ang-1 with PEI[J]. Biomedical Materials, 2014. DOI: 10.1088/1748-6041/9/3/035015.
[62] LIU Y, YOU R C, LIU G Y, et al. Antheraea pernyi silk fibroin-coated PEI/DNA complexes for targeted gene delivery in HEK 293 and HCT 116 Cells[J]. International Journal of Molecular Sciences, 2014, 15(5): 7049-7063.
doi: 10.3390/ijms15057049 pmid: 24776757
[63] SONG W X, GREGORY D A, AL-JANABI H, et al. Magnetic-silk/polyethyleneimine core-shell nanoparticles for targeted gene delivery into human breast cancer cells[J]. International Journal of Pharmaceutics, 2019, 555: 322-336.
doi: S0378-5173(18)30848-2 pmid: 30448314
[64] 刘岩, 吕志强, 张存, 等. IBDV丝素蛋白/壳聚糖DNA微球疫苗的制备及免疫原性分析[J]. 生物工程学报, 2014, 30(3): 393-403.
pmid: 25007575
LIU Yan, LÜ Zhiqiang, ZHANG Cun, et al. Preparation and immunogenicity of silk fibroin/chitosan microspheres for DNA vaccine delivery against infectious bursal disease virus[J]. Chinese Journal of Biotechnology, 2014, 30(3): 393-403.
pmid: 25007575
[65] WANG W W, YU Y N, JIANG Y, et al. Silk fibroin scaffolds loaded with angiogenic genes in adenovirus vectors for tissue regeneration[J]. Journal of Tissue Engineering and Regenerative Medicine, 2019, 13(5): 715-728.
doi: 10.1002/term.2819 pmid: 30770653
[66] NYARUABA R, HONG W, LI X H, et al. Long-term preservation of SARS-CoV-2 RNA in silk for downstream RT-PCR tests[J]. Analytical Chemistry, 2022, 94(10): 4522-4530.
doi: 10.1021/acs.analchem.2c00169 pmid: 35235308
[1] 杨其亮, 杨海伟, 王邓峰, 李长龙, 张乐乐, 王宗乾. 超疏水弹性丝素蛋白纤维气凝胶的制备及其吸油性能[J]. 纺织学报, 2023, 44(09): 1-10.
[2] 姚双双, 付少举, 张佩华, 孙秀丽. 再生丝素蛋白/聚乙烯醇共混取向纳米纤维膜的制备与性能[J]. 纺织学报, 2023, 44(09): 11-19.
[3] 狄纯秋, 郭静, 管福成, 相玉龙, 单继成. 双金属离子交联海藻酸盐复合相变纤维的制备与性能[J]. 纺织学报, 2023, 44(05): 54-62.
[4] 俞杨销, 李枫, 王煜煜, 王善龙, 王建南, 许建梅. 聚吡咯/丝素导电纳米纤维膜的制备及其性能[J]. 纺织学报, 2022, 43(10): 16-23.
[5] 刘蛟, 陈韶娟, 吴韶华. 丝素蛋白/聚左旋乳酸纳米纤维纱线肌腱补片的制备及其性能[J]. 纺织学报, 2022, 43(08): 60-66.
[6] 李艾元, 施心雨, 岳万福, 游卫云. 丝素蛋白水凝胶支架的制备及其性能[J]. 纺织学报, 2022, 43(06): 44-48.
[7] 顾张弘, 姚响, 王锦思, 张耀鹏. 具有细胞黏附反差特性的单层平行丝素蛋白纤维图案的制备及其性能[J]. 纺织学报, 2022, 43(05): 1-6.
[8] 雷彩虹, 俞林双, 朱海霖, 郑涛, 陈建勇. 不同水解方式下蚕丝丝素蛋白材料的止血性能[J]. 纺织学报, 2022, 43(04): 15-19.
[9] 张涛, 王富平, 陈国宝, 吴基玉, 庞亚妮, 陈忠敏. 壳聚糖基抑菌凝胶剂的制备及其性能[J]. 纺织学报, 2022, 43(03): 71-77.
[10] 姚若彤, 赵婧媛, 闫一欣, 段立蓉, 王恬, 严佳, 张淑军, 李刚. 新型可降解编织结构神经再生导管的制备及其性能[J]. 纺织学报, 2022, 43(02): 125-131.
[11] 姜雨淋, 王卉, 张克勤. 生物3D打印用丝素蛋白基凝胶墨水的研究进展[J]. 纺织学报, 2021, 42(11): 1-8.
[12] 孙钰晟, 左保齐. 高分子聚合物硬骨缺损修复材料研究进展[J]. 纺织学报, 2021, 42(08): 175-184.
[13] 刘浩, 路明磊, 黄晓卫, 王娜, 王雪芳, 宁新, 明津法. 酸-醇体系丝素蛋白水凝胶制备与性能表征[J]. 纺织学报, 2021, 42(08): 41-48.
[14] 丁梦瑶, 戴梦男, 李蒙, 刘苹, 徐晶晶, 王建南. 不同分子质量丝素蛋白的分离与表征[J]. 纺织学报, 2021, 42(07): 46-53.
[15] 杨亚, 闫凤祎, 王卉, 张克勤. 丝素蛋白/磷酸八钙复合材料生物界面的蛋白质吸附和细胞响应[J]. 纺织学报, 2021, 42(02): 41-46.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed   
No Suggested Reading articles found!