Soft Nanomaterials and Their Applications

  • Chapter
  • First Online:
Nanomaterials: The Building Blocks of Modern Technology

Part of the book series: Smart Nanomaterials Technology ((SNT))

  • 318 Accesses

Abstract

Soft nanomaterials have garnered significant attention in recent years due to their unique mechanical and chemical properties. This chapter provides an overview of the synthesis, characterization, and applications of soft nanomaterials, including hydrogels, liposomes, and self-assembled macro-molecules. We begin by discussing the properties and synthesis methods of these materials, such as solution mixing, emulsification, and self-assembly. The chapter also explores the various applications of soft nanomaterials, including in drug delivery, tissue engineering, biosensors, and personal care products. Finally, the chapter concludes with a summary of the current state of research and suggests possible directions for future work in this exciting field.

This is a preview of subscription content, log in via an institution to check access.

Access this chapter

Subscribe and save

Springer+ Basic
EUR 32.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or Ebook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

Chapter
USD 29.95
Price excludes VAT (USA)
  • Available as PDF
  • Read on any device
  • Instant download
  • Own it forever
eBook
USD 149.00
Price excludes VAT (USA)
  • Available as EPUB and PDF
  • Read on any device
  • Instant download
  • Own it forever
Hardcover Book
USD 199.99
Price excludes VAT (USA)
  • Durable hardcover edition
  • Dispatched in 3 to 5 business days
  • Free ship** worldwide - see info

Tax calculation will be finalised at checkout

Purchases are for personal use only

Institutional subscriptions

Similar content being viewed by others

References

  1. García-Betancourt ML, Jiménez SIR, González-Hodges A, Salazar ZEN, Escalante-García IL et al (2021) Low dimensional structures: measurement and remediation technologies applied to trace heavy metals in water. In: Murillo-Tovar MA, Saldarriaga-Noreña H, Saeid A (eds) Trace metals in the environment. IntechOpen, London. https://doi.org/10.5772/intechopen.93263

  2. Chellaram C, Murugaboopathi G, John AA, Sivakumar R, Ganesan S et al (2014) Significance of nanotechnology in food industry. APCBEE Proc 8:109–113. https://doi.org/10.1016/j.apcbee.2014.03.010

    Article  CAS  Google Scholar 

  3. Geoffrion LD, Guisbiers G (2020) Quantum confinement: size on the grill! J Phys Chem Solids 140:109320. https://doi.org/10.1016/j.jpcs.2019.109320

    Article  CAS  Google Scholar 

  4. Asha AB, Narain R (2020) Nanomaterials properties, In: Narain R (ed) Polymer science and nanotechnology: fundamentals and applications. Elsevier, pp 243–259. https://doi.org/10.1016/B978-0-12-816806-6.00015-7

  5. Bhagyaraj SM, Oluwafemi OS (2018) Nanotechnology: the science of the invisible. In: Bhagyaraj SM, Oluwafemi OS, Kalarikkal N, Thomas S (eds) Synthesis of inorganic nanomaterials: advances and key technologies. Woodhead Publishing, Elsevier, pp 1–18. https://doi.org/10.1016/B978-0-08-101975-7.00001-4

  6. Malhotra BD, Ali MA (2018) Nanomaterials in biosensors—fundamentals and applications. Nanomaterial Biosens 1–74. https://doi.org/10.1016/B978-0-323-44923-6.00001-7

  7. Barhoum A, García-Betancourt ML, Jeevanandam J, Hussein EA, Mekkawy SA et al (2022) Review on natural, incidental, bioinspired, and engineered nanomaterials: history, definitions, classifications, synthesis, properties, market, toxicities, risks, and regulations. Nanomaterials 12(2):177. https://doi.org/10.3390/nano12020177

    Article  CAS  Google Scholar 

  8. Hochella Jr MF, Mogk DW, Ranville J, Allen IC, Luther GW (2019) Natural, incidental, and engineered nanomaterials and their impacts on the earth system. Science 363(6434). https://doi.org/10.1126/science.aau8299

  9. Lespes G, Faucher S, Slaveykova VI (2020) Natural nanoparticles, anthropogenic nanoparticles, where is the frontier? Front Environ Sci 8. https://doi.org/10.3389/fenvs.2020.00071

  10. Gebeshuber IC, Gazsó A, Pavlicek A, Rose G (2020) Bio-inspired and biomimetic nanomaterials. Proj NanoTrust. https://doi.org/10.1553/ita-nt-054en

  11. Bhattacharya P, Du D, Lin Y (2014) Bioinspired nanoscale materials for biomedical and energy applications. J Roy Soc Interface 11(95). https://doi.org/10.1098/rsif.2013.1067

  12. Kaur N, Prakash C, Bhalla A, Chaudhary GR (2020) Nanodiamonds and other organic nanoparticles: synthesis and surface modifications. In: Kumar V, Guleria P, Dasgupta N, Ranjan S (eds) Functionalized nanomaterials i: fabrications. CRC Press

    Google Scholar 

  13. Xu M, Yim W, Zhou J, Zhou J, ** Z et al (2021) The application of organic nanomaterials for bioimaging, drug delivery, and therapy. IEEE Nanatechnol Mag 15(4):8–28. https://doi.org/10.1109/MNANO.2021.3081758

    Article  Google Scholar 

  14. Khalid K, Tan X, Zaid HFM, Tao Y, Chew CL et al (2020) Advanced in developmental organic and inorganic nanomaterials: a review. Bioengineered 11(1):328–355. https://doi.org/10.1080/21655979.2020.1736240

    Article  CAS  Google Scholar 

  15. Poon C, Patel AA (2020) Organic and inorganic nanoparticle vaccines for prevention of infectious diseases. Nano Express 1(1). https://doi.org/10.1088/2632-959X/ab8075

  16. Baig N, Kammakakam I, Falath W (2021) Nanomaterials: a review of synthesis methods, recent progress, and challenges. Mater Adv 2:1821–1871. https://doi.org/10.1039/D0MA00807A

    Article  Google Scholar 

  17. Hadef F (2018) An introduction to nanomaterials. In: Dasgupta N, Ranjan S, Lichtfouse E (eds) Environmental nanotechnology, vol 1. Springer Cham, pp 1–58. https://doi.org/10.1007/978-3-319-76090-2_1

  18. Augustine R, Hasan A (2020) Multimodal applications of phytonanoparticles. In: Thajuddin N, Mathew S (eds) Phytonanotechnology: challenges and prospects. Elsevier, pp 195–219. https://doi.org/10.1016/B978-012-822348-2.00011-5

  19. Ying S, Guan Z, Ofoegbu PC, Clubb P, Rico C et al (2022) Green synthesis of nanoparticles: current developments and limitations. Environ Technol Innovation 26. https://doi.org/10.1016/j.eti.2022.102336

  20. Roduner E (2006) Size matters: why are nanomaterials different. Chem Soc Rev 35(7):583–592. https://doi.org/10.1039/B502142C

    Article  CAS  Google Scholar 

  21. Hong NH (2019) Introduction to nanomaterials: Basic properties, synthesis, and characterization. In: Hong NH (eds) Nano-sized multifunctional materials: synthesis, properties and applications. Elsevier, pp 1–19. https://doi.org/10.1016/B978-0-12-813943-9.00001-3

  22. Bharmoria P, Ventura SPM (2019) Optical applications of nanomaterials. In: Bhatt AH et al (eds) Nanomaterials for healthcare, energy and environment, adavnaced structured materials, vol 118. Springer Nature Singapore, pp 1–29. https://doi.org/10.1007/978-981-13-9833-9 1

  23. Singh M, Goyal M, Devlal K (2018) Size and shape effects on the band gap of semiconductor compound nanomaterials. J Taibah Univ Sci 12(4):470–475. https://doi.org/10.1080/16583655.2018.1473946

    Article  Google Scholar 

  24. Yang CC, Mai YW (2014) Thermodynanomics at the nanoscale: a new approach to the investigation of unique physicochemical properties of nanomaterials. Mater Sci Eng R Rep 79:1–40. https://doi.org/10.1016/j.mser.2014.02.001

    Article  Google Scholar 

  25. Karak N (2019) Fundamentals of nanomaterials and polymer nanocomposites. In: Karak N (ed) Nanomaterials and polymer nanocomposites: raw materials to applications. Elsevier, pp 1–45. https://doi.org/10.1016/B978-0-12-814615-6.00001-1

  26. Koksharov YA (2009) Magnetism of nanoparticles: effects of size, shape, and interactions. In: Gubin SP (eds) Magnetic nanoparticles. Wiley-VCH Verlag GmbH & Co. KGaA, pp 197–254. https://doi.org/10.1002/9783527627561.ch6

  27. Wu Q, Miao WS, Zhang YD, Gao HJ, Hui D (2020) Mechanical properties of nanomaterials: a review. Nanotechnol Rev 9(1):259–273. https://doi.org/10.1515/ntrev-2020-0021

    Article  CAS  Google Scholar 

  28. Reghunadhan A, Kalarikkal N, Thomas S (2018) Mechanical property analysis of nanomaterials. In: Bhagyaraj SM, Oluwafemi OS, Kalarikkal N, Thomas S (eds) Characterization of nanomaterials: advances and key technologies. Woodhead Publishing, Elsevier, pp 191–212. https://doi.org/10.1016/B978-0-08-101973-3.00007-9

  29. Murty BS, Shankar P, Raj B, Rath BB, Murday J (2012) Unique properties of nanomaterials. In: Raj B (ed) Textbook of nanoscience and nanotechnology, vol 1. Springer, Berlin, pp 29–65. https://doi.org/10.1007/978-3-642-28030-6_2

  30. Romera G, Moya SE (2012) Synthesis of organic nanoparticles. In: de la Fuente JM, Grazu V (eds) Frontiers of nanoscience—nanobiotechnology: inorganic nanoparticles vs organic nanoparticles, vol 4. Elsevier, pp 115–141. https://doi.org/10.1016/B978-0-12-415769-9.00004-2

  31. Lombardo D, Calandra P, Pasqua L, Magazù L (2020) Self-assembly of organic nanomaterials and biomaterials: the bottom-up approach for functional nanostructures formation and advanced applications. Mater (Basel, Switz) 13(5):1048. https://doi.org/10.3390/ma13051048

    Article  CAS  Google Scholar 

  32. Hamley IW (2003) Nanotechnology with soft nanomaterials. Angew Chem Int Ed Engl 42(15):1692–1712. https://doi.org/10.1002/anie.200200546

    Article  CAS  Google Scholar 

  33. Soni KS, Desale SS, Bronich TK (2016) Nanogels: an overview of properties, biomedical applications, and obstacles to clinical translation. J Control Release 240:109–126. https://doi.org/10.1016/j.jconrel.2015.11.009

    Article  CAS  Google Scholar 

  34. Karg M, Pich A, Hellweg T, Hoare T, Lyon LA et al (2019) Nanogels and microgels: from model colloids to applications, recent developments, and future trends. Langmuir 35(19):6231–6255. https://doi.org/10.1021/acs.langmuir.8b04304

    Article  CAS  Google Scholar 

  35. Nayak S, Lyon LA (2005) Soft nanotechnology with soft nanoparticles. Angew Chem Int Ed 44(47):7686–7708. https://doi.org/10.1002/anie.200501321

    Article  CAS  Google Scholar 

  36. Sánchez A, Villalonga A, Martínez-García G, Parrado C, Villalonga R (2019) Dendrimers as soft nanomaterials for electrochemical immunosensors. Mater (Basel, Switz) 9(12):1745. https://doi.org/10.3390/nano9121745

    Article  CAS  Google Scholar 

  37. Villalong R, Díez P, Casado S, Eguílas M, Yáñez-Sedeño P et al (2012) Electropolymerized network of polyamidoamine dendron-coated gold nanoparticles as novel nanostructured electrode surface for biosensor construction. Analyst 137(2):342–348. https://doi.org/10.1039/C1AN15850C

    Article  Google Scholar 

  38. Aguilar ZP (2013) Types of nanomaterials and corresponding methods of synthesis—nanomaterials for medical applications. In: Nanomaterials for medical applications, Elsevier, pp 33–82. https://doi.org/10.1016/B978-0-12-385089-8.00002-9

  39. Zielińska A, Carreiró F, Oliveira AM, Neves A, Pires B et al (2020) Polymeric nanoparticles: production, characterization, toxicology and ecotoxicology. Molecules 25(16):3731. https://doi.org/10.3390/molecules25163731

    Article  CAS  Google Scholar 

  40. Uchegbu IF (2006) Pharmaceutical nanotechnology: polymeric vesicles for drug and gene delivery. Expert Opin Drug Deliv 3(5):629–640. https://doi.org/10.1517/17425247.3.5.629

    Article  CAS  Google Scholar 

  41. Kaur N, Kaur S, Kaur G, Bhalla A, Srinivasan S et al (2019) Metallovesicles as smart nanoreactors for green catalytic synthesis of benzimidazole derivatives in water. J Mater Chem A 7:17306–17314. https://doi.org/10.1039/C9TA05441C

    Article  CAS  Google Scholar 

  42. Shukla F, Das M, Thakore S (2021) Copper nanoparticles loaded polymer vesicles as environmentally amicable nanoreactors: a sustainable approach for cascading synthesis of benzimidazole. J Mol Liq 336:116217. https://doi.org/10.1016/j.molliq.2021.116217

    Article  CAS  Google Scholar 

  43. Joseph M, Trinh HM, Mitra AK (2017) Peptide- and protein-based therapeutic agents. In: Mitra AK, Cholkar K, Mandal A (eds) Emerging nanotechnologies for diagnostics, drug delivery, and medical devices. Elsevier, pp 145–167. https://doi.org/10.1016/B978-0-323-42978-8.00007-3

  44. Rana S, Bhattacherjee A, Barrick KC, Verma G, Hassan PA et al (2017) Interfacial engineering of nanoparticles for cancer therapeutics. In: Ficai A, Grumezescu AM (eds) Nanostructures for cancer therapy. Elsevier, pp 177–209. https://doi.org/10.1016/B978-0-323-46144-3.00007-6

  45. Suhail N, Alzahrani AK, Basha WJ, Kizilbash N, Zaidi A et al (2021) Microemulsions: unique properties, pharmacological applications, and targeted drug delivery. Front Nanotechnol 3:754889. https://doi.org/10.3389/fnano.2021.754889

    Article  Google Scholar 

  46. Rodriguez-Dafonte P (2019) Nitrosation of amines in AOT-based microemulsions. In: Mejuto JC (ed) Microemulsion—a chemical nanoreactor. IntechOpen. https://doi.org/10.5772/intechopen.80947

  47. Shimizu T, Ding W, Kameta N (2020) Soft-matter nanotubes: a platform for diverse functions and applications. Chem Rev 120(4):2347–2407. https://doi.org/10.1021/acs.chemrev.9b00509

    Article  CAS  Google Scholar 

  48. Kawauchi H, Takahashi R, Kameta N, Fujii S, Lee JH et al (2019) Encapsulation of albumin in organic nanotube channel: structural investigation by small angle X ray scattering. ACS Appl BioMater 2(4):1652–1659. https://doi.org/10.1021/acsabm.9b00047

    Article  CAS  Google Scholar 

  49. Das I, Padhi A, Mukherjee S, Dash DP, Kar S et al (2017) Biocompatible chitosan nanoparticles as an efficient delivery vehicle for mycobacterium tuberculosis lipids to induce cytokines and antibody response through activation of γδ t cells in mice. Nanotechnology 28(16):165101. https://doi.org/10.1088/1361-6528/aa60fd

    Article  CAS  Google Scholar 

  50. Zhou Q, Hou Y, Zhang L, Wang J, Qiao Z et al (2017) Dual pH-sensitive charge-reversal nanocomplex for tumor-targeted drug delivery with enhanced anticancer activity. Theranostics 7(7):1806–1819. https://doi.org/10.7150/thno.18607

    Article  CAS  Google Scholar 

  51. Li K, Zhang Y, Chen M, Hu Y, Jiang W et al. (2018) Enhanced antitumor efficacy of doxorubicin-encapsulated halloysite nanotubes. Int J Nanomed 13:19–30. https://doi.org/10.2147/IJN.S143928

  52. Newland B, Taplan C, Pette D, Friedrichs J, Steinhart M et al (2018) Soft and flexible poly(ethylene glycol) nanotubes for local drug delivery. Nanoscale 10:8413–8421. https://doi.org/10.1039/C8NR00603B

    Article  CAS  Google Scholar 

  53. Rao KM, Kumar A, Suneetha M, Han SS (2018) pH and near infrared active; chitosan-coated halloysite nanotubes loaded with curcumin-Au hybrid nanoparticles for cancer drug delivery. Int J Biol Macromol 112:119–125. https://doi.org/10.1016/j.ijbiomac.2018.01.163

    Article  CAS  Google Scholar 

  54. Koshy ST, Zhang DKY, Grolman JM, Stafford AG, Mooney DJ (2018) Injectable nanocomposite hydrogels for versatile protein drug delivery. Acta Biomater 65:36–43. https://doi.org/10.1016/j.actbio.2017.11.024

    Article  CAS  Google Scholar 

  55. Joseph J, Vedha Hari BN, Ramya DD (2018) Experimental optimization of lornoxicam liposomes for sustained topical delivery. Eur J Pharm Sci 112:38–51. https://doi.org/10.1016/j.ejps.2017.10.032

    Article  CAS  Google Scholar 

  56. Bera S, Chowdhary A, Sarkar K, Dastidar P (2019) Design and synthesis of Zn(II)-coordination polymers anchored with NSAIDs: metallovesicle formation and multi-drug delivery. Chem An Asian J 15(4):503–510. https://doi.org/10.1002/asia.201901664

  57. Fan Y, Stronsky SM, Xu Y, Steffens JT, van Tongeren SA et al (2019) Multilamellar vaccine particle elicits potent immune activation with protein antigens and protects mice against Ebola virus infection. ACS Nano 13(10):11087–11096. https://doi.org/10.1021/acsnano.9b03660

  58. Maiorova LA, Erokhina SI, Pisani M, Barucca G, Marcaccio M et al (2019) Encapsulation of vitamin B12 into nanoengineered capsules and soft matter nanosystems for targeted delivery. Colloids Surf B 182:110366. https://doi.org/10.1016/j.colsurfb.2019.110366

  59. Liu C, Wan T, Wang H, Zhang S, ** Y et al (2019) A boronic acid-rich dendrimer with robust and unprecedented efficiency for cytosolic protein delivery and CRISPR-Cas9 genome editing, Sci Adv 5(6). https://doi.org/10.1126/sciadv.aaw8922

  60. Zhao Y, Cai F, Shen X, Su H (2020) A high stable pH-temperature dual-sensitive liposome for tuning anticancer drug release. Synth Syst Biotechnol 5(2):103–110. https://doi.org/10.1016/j.synbio.2020.05.004

    Article  Google Scholar 

  61. Qu Y, Ju Y, Cortez-Jugo C, Lin Z, Li S et al (2020) Template-mediated assembly of DNA microcapsules for immunological modulation. Small 16(7):2022750. https://doi.org/10.1002/smll.202002750

    Article  CAS  Google Scholar 

  62. Feng S, Sun Y, Wang D, Sun P, Shao P (2020) Effect of adjusting pH and chondroitin sulfate on the formation of curcumin-zein nanoparticles: synthesis, characterization and morphology. Carbohydr Polym 250:116970. https://doi.org/10.1016/j.carbpol.2020.116970

    Article  CAS  Google Scholar 

  63. Meng R, Wu Z, **e QT, Cheng JS, Zhang B (2021) Preparation and characterization of zein/ carboxymethyl dextrin nanoparticles to encapsulate curcumin: physicochemical stability, antioxidant activity and controlled release properties. Food Chem 340:127893. https://doi.org/10.1016/j.foodchem.2020.127893

    Article  CAS  Google Scholar 

  64. Zhang H, van Os WL, Tian X, Zu G, Ribovski L et al (2021) Development of curcumin-loaded zein nanoparticles for transport across the blood-brain barrier and inhibition of glioblastoma cell growth. Biomater Sci 9:7092–7103. https://doi.org/10.1039/D0BM01536A

    Article  CAS  Google Scholar 

  65. Penalva R, González-Navarro CJ, Gamazo C, Esparza I, Irache JM (2017) Zein nanoparticles for oral delivery of quercetin: pharmacokinetic studies and preventive anti-inflammatory effects in a mouse model of endotoxemia. Nanomedicine 13(1):103–110. https://doi.org/10.1016/j.nano.2016.08.033

    Article  CAS  Google Scholar 

  66. Rodríguez-Félix F, Del-Toro-Sánchez CL, Cinco-Moroyoqui FJ, Juárez J, Ruiz-Cruz S et al (2019) Preparation and characterization of quercetin-loaded zein nanoparticles by electrospraying and study of in vitro bioavailability. J Food Sci 84(10):2883–2897. https://doi.org/10.1111/1750-3841.14803

    Article  CAS  Google Scholar 

  67. Zhou Y, Qian Y, Rong X, Cao K, McClements DJ et al (2021) Encapsulation of quercetin in biopolymer-coated zein nanoparticles: formation, stability, antioxidant capacity and bioaccessibility. Food Hydrocolloids 120:106980. https://doi.org/10.1016/j.foodhyd.2021.106980

    Article  CAS  Google Scholar 

  68. Vale EP, dos Santos Morais E, de Souza Tavares W, de Souza FFO (2022) Epigallocatechin-3 gallate loaded zein nanoparticles: characterization, stability and anti-oxidant, anti-tyorsinase and sun-protection properties. J Mol Liq 358:119107. https://doi.org/10.1016/j.molliq.2022.119107

    Article  CAS  Google Scholar 

  69. Chen X, Zhou B, Gao J, Wu D, Liang H (2022) EGCG-decorated zein complex nanoparticles: relationship to synergistic interfacial properties. Colloids Surf, A 652:129879. https://doi.org/10.1016/j.colsurfa.2022.129879

    Article  CAS  Google Scholar 

  70. Sun CC, Su H, Zheng GD, Wang WJ, Yuan E et al (2020) Fabrication and characterization of dihydromyricetin encapsulated zein-caseinate nanoparticles and its bioavailability in rat. Food Chem 330:127245. https://doi.org/10.1016/j.foodchem.2020.127245

    Article  CAS  Google Scholar 

  71. Huang X, Liu Y, Zhou Y, Liang X, Peng Y et al (2019) Encapsulation of resveratrol in zein/ pectin core-shell nanoparticles: stability, bioaccessibility, and antioxidant capacity after simulated gastrointestinal digestion. Food Hydrocolloids 93:261–269. https://doi.org/10.1016/j.foodhyd.2019.02.039

    Article  CAS  Google Scholar 

  72. Nunes R, Baião A, Monteiro D, das Neves J, Sarmento B (2020) Zein nanoparticles as low-cost, safe, and effective carriers to improve the oral bioavailaibilty of resveratrol. Drug Delivery Transl Res 10(3):826–837. https://doi.org/10.1007/s13346-020-00738-z

  73. Ferreira IVW, Focke WW, du Toit EL (2018) Spontaneous microencapsulation of geraniol by Zein. eXPRESS Polym Lett 12(11):986–995. https://doi.org/10.3144/expresspolymlett.2018.85

  74. Chen Y, Shu M, Yao X, Wu K, Zhang K et al (2018) Effect of zein-based microencapsules on the release and oxidation of loaded limonene. Food Hydrocolloids 84:330–336. https://doi.org/10.1016/j.foodhyd.2018.05.049

    Article  CAS  Google Scholar 

  75. Merino N, Berdejo D, Bento R, Salman H, Lanz M et al (2019) Anti-microbial efficacy of Thymbra capitata (L.) Cav. essential oil loaded in self-assembled zein nanoparticle in combination with heat. Ind Crops Prod 133:98–104. https://doi.org/10.1016/j.indcrop.2019.03.003

    Article  CAS  Google Scholar 

  76. Kim S, Peterson SC (2021) Optimal conditions for the encapsulation of menthol into zein nanoparticles. LWT 144:111213. https://doi.org/10.1016/j.lwt.2021.111213

    Article  CAS  Google Scholar 

  77. Li H, Xu Y, Sun X, Wang S, Wang J et al (2018) Stability, bioactivity, and bioaccessibility of fucoxanthin in zein-caseinate composite nanoparticles fabricated at neutral pH by antisolvent precipitation. Food Hydrocolloids 84:379–388. https://doi.org/10.1016/j.foodhyd.2018.06.032

    Article  CAS  Google Scholar 

  78. Cheng CJ, Ferruzzi M, Jones OG (2019) Fate of lutein-containing zein nanoparticles following simulated gastric and intestinal digestion. Food Hydrocolloids 87:229–236. https://doi.org/10.1016/j.foodhyd.2018.08.013

    Article  CAS  Google Scholar 

  79. Chen S, Li Q, McClements DJ, Han Y, Dai L et al (2020) Co-delivery of curcumin and piperine in zein-carrageenan core-shell nanoparticles: formation, structure, stability, and in vitro gastrointestinal digestion. Food Hydrocolloids 99:105334. https://doi.org/10.1016/j.foodhyd.2019.105334

    Article  CAS  Google Scholar 

  80. Chen S, Han Y, Jian L, Liao W, Zhang Y (2020) Fabrication, characterization, and physicochemical stability of zein-chitosan nanocomplex for co-encapsulating curcumin and resveratrol. Carbohyd Polym 236:116090. https://doi.org/10.1016/j.carbpol.2020.116090

    Article  CAS  Google Scholar 

  81. Kurczewska J, Pecyna P, Ratajczak M, Gajęcka M, Schroeder G (2017) Halloysite nanotubes as carriers of vancomycin in alginate-based wound dressing. Saudi Pharm J 25(6):911–920. https://doi.org/10.1016/j.jsps.2017.02.007

    Article  Google Scholar 

  82. Lu Z, Zhang J, Yu Z, Liu Q, Liu K et al (2017) Hydrogel degradation triggered by pH for the smart release of antibiotics to combat bacterial infection. New J Chem 41(2):432–436. https://doi.org/10.1039/C6NJ03260E

    Article  CAS  Google Scholar 

  83. Li L, Yan B, Yang J, Huang W, Chen L et al (2017) Injectable self-healing hydrogel with antimicrobial and antifouling properties. ACS Appl Mater Interfaces 9(11):9221–9225. https://doi.org/10.1021/acsami.6b16192

    Article  CAS  Google Scholar 

  84. Sandri G, Aguzzi C, Rossi S, Bonferoni MC, Bruni G et al (2017) Halloysite and chitosan oligosaccharide nanocomposite for wound healing. Acta Biomater 57:216–224. https://doi.org/10.1016/j.actbio.2017.05.032

    Article  CAS  Google Scholar 

  85. Jiang Y, Li J, Zhen X, **e C, Pu K (2018) Dual-peak absorbing semiconducting copolymer nanoparticles for first and second-near infrared window for photothermal therapy: a comparative study. Adv Mater 30(14):1705980. https://doi.org/10.1002/adma.201705980

    Article  CAS  Google Scholar 

  86. Qin J, Wei X, Chen H, Lv F, Nan W et al (2018) mPEG-g-CS-modified PLGA nanoparticle carrier for the codelivery of paclitaxel and epirubicin for breast cancer synergistic therapy. ACS Biomater Sci Eng 4(5):1651–1660. https://doi.org/10.1021/acsbiomaterials.7b01003

    Article  CAS  Google Scholar 

  87. Wang M, Cai X, Yang J, Wang C, Tong L et al (2018) A targeted and pH-responsive bortezomib nanomedicine in the treatment of metastatic bone tumors. ACS Appl Mater Interfaces 10(48):41003–41011. https://doi.org/10.1021/acsami.8b07527

    Article  CAS  Google Scholar 

  88. Yu W, Liu T, Zhang M, Wang Z, Ye J et al (2019) O2-economizer for inhibiting cell respiration to combat hypoxia obstacle in tumor treatments. ACS Nano 13(2):1784–1794. https://doi.org/10.1021/acsnano.8b07852

    Article  CAS  Google Scholar 

  89. Pandya AD, Øverbye A, Sahariah P, Gaware VS, Høgset H et al (2020) Drug-loaded photosensitizer-chitosan nanoparticles for combinatorial chemo- and photodynamic therapy of cancer. Biomacromol 21(4):1489–1498. https://doi.org/10.1021/acs.biomac.0c00061

    Article  CAS  Google Scholar 

  90. Shinde P, Agraval H, Srivastav AK, Yadav UCS, Kumar U (2020) Physico-chemical characterization of carvacrol loaded zein nanoparticles for enhanced anticancer activity and investigation of molecular interactions between them by molecular docking. Int J Pharm 588:119795. https://doi.org/10.1016/j.ijpharm.2020.119795

    Article  CAS  Google Scholar 

  91. Zhang S, Song W, Wu H, Wang J, Wang Y et al (2020) Lecithin-zein nanoparticles for antifungal treatment: enhancement and prolongation of drug retention in skin with reduced toxicity. Int J Pharm 590:119894. https://doi.org/10.1016/j.ijpharm.2020.119894

    Article  CAS  Google Scholar 

  92. Chen K, Qian Y, Wang C, Yang D, Qiu X et al (2021) Tumour microenvironment pH-responsive, high internal phase pickering emulsions stabilized by lignin/chitosan oligosaccharide particles for synergistic cancer therapy. J Colloid Interface Sci 591:352–362. https://doi.org/10.1016/j.jcis.2021.02.012

    Article  CAS  Google Scholar 

  93. Zhu JJ, Tang CH, Luo FC, Yin SW, Yang XQ (2022) Topical application of zein-silk sericin nanoparticles loaded with curcumin for improved therapy of dermatitis. Mater Today Chem 24:100802. https://doi.org/10.1016/j.mtchem.2022.100802

    Article  CAS  Google Scholar 

  94. Chen X, Wang B, Mao Y, Al MA, Wu M et al (2022) Zein nanoparticles loaded with chloroquine improve functional recovery and attenuate neuroinflammation after spinal cord injury. Chem Eng J 450(1):137882. https://doi.org/10.1016/j.cej.2022.137882

    Article  CAS  Google Scholar 

  95. Xu Y, Asghar S, Gao S, Chen Z, Huang L et al (2017) Polysaccharide-based nanoparticles for co-loading mitoxantrone and verapamil to overcome multidrug resistance in breast tumor. Int J Nanomedicinec 12:7337–7350. https://doi.org/10.2147/IJN.S145620

    Article  CAS  Google Scholar 

  96. Sun H, Li S, Qi W, **ng R, Zou Q et al (2018) Stimuli-responsive nanoparticles based on co-assembly of naturally-occurring biomacromolecules for in vitro photodynamic therapy. Colloidas Surf Physicochemical Eng Aspects 538:795–801. https://doi.org/10.1016/j.colsurfa.2017.11.072

    Article  CAS  Google Scholar 

  97. Ding J, Liang T, Min Q, Jiang L, Zhu JJ (2018) “Stealth and fully laden” drug carriers: self- assembled nanogels encapsulated with epigallocatechin gallate and sirna for drug-resistant breast cancer therapy. ACS Appl Mater Interfaces 10(12):9938–9948. https://doi.org/10.1021/acsami.7b19577

    Article  CAS  Google Scholar 

  98. Zhang Q, Wang J, Liu D, Zhu W, Guan S et al (2020) Targeted delivery of honokiol by zein/hyaluronic acid core-shell nanoparticles to suppress breast cancer growth and metastasis. Carbohydr Polym 240:116325. https://doi.org/10.1016/j.carbpol.2020.116325

    Article  CAS  Google Scholar 

  99. Zhang R, Ru Y, Gao Y, Li J, Mao S (2017) Layer-by-layer nanoparticles co-loading gemcitabine and platinum (IV) prodrugs for synergistic combination therapy of lung cancer. Drug Des Dev Ther 11:2631–2642. https://doi.org/10.2147/DDDT.S143047

    Article  CAS  Google Scholar 

  100. Simonson AW, Lawanprasert A, Goralski TDP, Keiler KC, Medina SH (2019) Bioresponsive peptide-polysaccharide nanogels—a versatile delivery system to augment the utility of bioactive cargo. Nanomedicine 17:391–400. https://doi.org/10.1016/j.nano.2018.10.008

    Article  CAS  Google Scholar 

  101. Guo M, Meng Y, Qin X, Zhou W (2021) Dopamine-grafted hyaluronic acid coated highly-branched poly (β-amino esters)/ DNA nano-complexes for enhanced gene delivery and biosafety. Crystals 11(4):347. https://doi.org/10.3390/cryst11040347

    Article  CAS  Google Scholar 

  102. Shariati M, Lollo G, Matha K, Descamps B, Vanhove C et al (2020) Synergy between intraperitoneal aerolization (pipac) and cancer nanomedicine: cisplatin-loaded polyarginine-hyaluronic acid nanocarriers efficiently eradicate peritoneal metastasis of advanced human ovarian cancer. ACS Appl Mater Interfaces 12(26):29024–29036. https://doi.org/10.1021/acsami.0c05554

    Article  CAS  Google Scholar 

  103. Seok HY, Re**old NS, Lekshmi KM, Cherukula K, Park IK et al (2018) CD44 targeting biocompatible and biodegradable hyaluronic acid cross-linked zein nanogels for curcumin delivery to cancer cells: in vitro and in vivo evaluation. J Contr Release 280:20–30. https://doi.org/10.1016/j.jconrel.2018.04.050

    Article  CAS  Google Scholar 

  104. Zhang H, Pei M, Liu P (2020) pH-activated surface charge-reversal double cross-linked hyaluronic acid nanogels with feather keratin as multifunctional crosslinker for tumor-targeting dox delivery. Int J Biol Macromol 150:1104–1112. https://doi.org/10.1016/j.ijbiomac.2019.10.116

    Article  CAS  Google Scholar 

  105. Zu Y, Asghar S, Yang L, Li H, Wang Z, ** Q et al (2017) Lactoferrin-coated polysaccharide nanoparticles based on chitosan hydrochloride/hyluronic acid/peg for brain glioma. Carbohyd Polym 157:419–428. https://doi.org/10.1016/j.carbpol.2016.09.085

    Article  CAS  Google Scholar 

  106. Sato T, Nakata M, Yang Z, Torizuka Y, Kishimoto S et al (2017) In vitro and in vivo gene delivery using chitosan/ hyaluronic acid nanoparticles: influences of molecular mass of hyaluronic acid and lyophilization on transfection efficiency. J Gene Med 19(8). https://doi.org/10.1002/jgm.2968

  107. Shi Q, Wang X, Tang X, Zhen N, Wang Y et al (2021) In vitro antioxidant and antitumor study of zein/SHA nanoparticles loaded with resveratrol. Food Sci Nutr 9(7):3530–3537. https://doi.org/10.1002/fsn3.2302

    Article  CAS  Google Scholar 

  108. Baglioni P, Giorgi R (2006) Soft and hard nanomaterials for the restoration and conservation of cultural heritage. Soft Matter 2:293–303. https://doi.org/10.1039/B516442G

    Article  CAS  Google Scholar 

  109. Syah R, Zahar M, Kianfar E (2021) Nanoreactors: properties, applications, and characterizations. Int J Chem Reactor Eng 19(10):1–27. https://doi.org/10.1515/ijcre-2021-0069

    Article  CAS  Google Scholar 

  110. Wang ZQ, Zhang GY, Zhong ZX, Lin Y, Su ZH (2018) In-situ synthesis of platinum nanoclusters in polyelectrolyte multilayer films. Thin Solid Filmsi 660:59–64. https://doi.org/10.1016/j.tsf.2018.05.051

    Article  CAS  Google Scholar 

  111. Jiang SD, Tang G, Chen J, Huang ZQ, Hu Y (2018) Biobased polyelectrolyte multilayer-coated hollow mesoporous silica as a green flame retardant for epoxy resin. J Hazard Mater 342:689–697. https://doi.org/10.1016/j.jhazmat.2017.09.001

    Article  CAS  Google Scholar 

  112. Chau NTT, Koh ES, Lee SJ, Rui Z, Yang SY (2021) Functional polyelectrolyte coatings on polymeric and magnetic colloidal particles for antifouling and non-toxic bioconjugate nanoparticles. Macromol Res 29(12):843–846. https://doi.org/10.1007/s13233-021-9102-8

    Article  CAS  Google Scholar 

  113. Klačić T, Peranić N, Radatović B, Kovačević D (2022) Biocompatible hydroxyapatite nanoparticles as templates for the preparation of thin film polyelectrolyte multilayer capsules. Colloids Surf, A 648:129385. https://doi.org/10.1016/j.colsurfa.2022.129385

    Article  CAS  Google Scholar 

  114. Shi X, Shen M, Möhwald H (2004) Polyelectrolyte multilayer nanoreactors toward the synthesis of diverse nanostructured materials. Prog Polym Sci 29(10):987–1019. https://doi.org/10.1016/j.progpolymsci.2004.07.001

    Article  CAS  Google Scholar 

  115. Kulshrestha A, Kumar G, Kumar A (2022) Cu(II)-amino acid ionic liquid surfactants: metallovesicles as nano-catalytic reactors for cross degenerative coupling reaction in water. Chem Sel 7(16). https://doi.org/10.1002/slct.202200159

  116. Tojo C, Buceta D, López-Quintela MA (2018) Microemulsion as nanoreactors to obtain bimetallic nanoparticles. In: Mejuto JC (ed) Microemulsion—a chemical nanoreactor. IntechOpen. https://doi.org/10.5772/intechopen.80549

  117. Chin SF, Jimmy FB, Pang SC (2018) Size-controlled fabrication of cellulose nanoparticles for drug delivery applications. J Drug Deliv Sci Technol 43:262–266. https://doi.org/10.1016/j.jddst.2017.10.021

    Article  CAS  Google Scholar 

  118. Ke W, Li J, Mohammed F, Wang Y, Tou K et al (2019) Therapeutic polymersome nanoreactors with tumor-specific activable cascade reactions for cooperative cancer therapy. ACS Nano 13(2):2357–2369. https://doi.org/10.1021/acsnano.8b09082

    Article  CAS  Google Scholar 

  119. Alqarni Y, Ishizuka F, Bell TDM, Tabor RF, Zetterlund PB et al (2020) Confined polymerization of bis-thyminyl monomers within nanoreactors: towards molecular weight control. Polym Chem 11(26):4326–4334. https://doi.org/10.1039/D0PY00523A

    Article  CAS  Google Scholar 

  120. Mandal RP, Sekh S, Mondal D, De S (2018) Multifunctional role of liposome-mimicking vesicles—potential nanoreactors and effective storehouses for haemoglobin. Colloids Surf, A 558:33–44. https://doi.org/10.1016/j.colsurfa.2018.08.048

    Article  CAS  Google Scholar 

  121. Bonifacio MA, Gentile P, Ferreira AM, Cometa S, de Giglio E (2017) Insight into halloysite nanotubes-loaded gellan gum hydrogels for soft tissue engineering applications. Carbohyd Polym 163:280–291. https://doi.org/10.1016/j.carbpol.2017.01.064

    Article  CAS  Google Scholar 

  122. Chen F, Ni Y, Liu B, Zhou T, Yu C et al (2017) Self-crosslinking and injectable hyaluronic acid/ RGD-grafted oxidized pectin hydrogel for cartilage tissue engineering. Carbohyd Polym 166:31–44. https://doi.org/10.1016/j.carbpol.2017.02.059

    Article  CAS  Google Scholar 

  123. Sionkowska A, Kaczmarek B (2017) Preparation and characterization of composites based on blends of chitosan, collagen, and hyaluronic acid with nano-hydroxyapatite. Int J Biol Macromol 102:658–666. https://doi.org/10.1016/j.ijbiomac.2017.03.196

    Article  CAS  Google Scholar 

  124. De Silva RT, Dissanayake RK, Mantilaka MMMGPG, Wijesinghe WPSL, Kaleel SS et al (2018) Drug-loaded halloysite nanotube reinforced-electrospun alginate-based nanofibrous scaffolds with sustained antimicrobial protection. ACS Appl Mater Interfaces 10(40):33913–33922. https://doi.org/10.1021/acsami.8b11013

    Article  CAS  Google Scholar 

  125. Caddeo C, Manca ML, Peris JE, Usach I, Diez-Sales O et al (2018) Tocopherol-loaded transfersomes: in vitro antioxidant activity and efficacy in skin regeneration. Int J Pharm 551(1–2):34–41. https://doi.org/10.1016/j.ijpharm.2018.09.009

    Article  CAS  Google Scholar 

  126. Balagangadharan K, Dhivya S, Selvamurugan N (2017) Chitosan-based nanofibers in bone tissue engineering. Int J Biol Macromol 104(B):1372–1382. https://doi.org/10.1016/j.ijbiomac.2016.12.046

  127. Tao F, Cheng Y, Shi X, Zheng H, Du Y et al (2019) Applications of chitin and chitosan nanofibers in bone regenerative engineering. Carbohyd Polym 230:115658. https://doi.org/10.1016/j.carbpol.2019.115658

    Article  CAS  Google Scholar 

  128. Duan B, Shou K, Su X, Niu Y, Zheng G et al (2017) Hierarchical microspheres constructed from chitin nanofibers penetrated hydroxyapatite crystals for bone regeneration. Biomacromol 18(7):2080–2089. https://doi.org/10.1021/acs.biomac.7b00408

    Article  CAS  Google Scholar 

  129. Hamidabadi HG, Rezvani Z, Bojnordi MN, Shirinzadeh H, Seifalian AM et al (2017) Chitosan-intercalated montmorillonite/poly(vinyl alcohol) nanofibers as a guided platform for neuronlike differentiation of human dental pulp stem cells. Appl Mater Interfaces 9(13):11392–11304. https://doi.org/10.1021/acsami.6b14283

    Article  CAS  Google Scholar 

  130. Cao L, Kong X, Lin S, Zhang S, Wang J et al (2018) Synergistic effects of dual growth factor delivery from composite hydrogels incorporating 2-n,6-o-sulphated chitosan on bone regeneration. Artifi Cells Nanomed Biotechnol 46(sup3):S1–S7. https://doi.org/10.1080/21691401.2018.1488721

    Article  CAS  Google Scholar 

  131. Seethalakshmi K, Venkatachalapathy B, Kaviya M, Mubeena S, Punnoose AM et al (2019) 6-o-trityl chitosan reinforced polycaprolactone nano scaffolds for bone replacement applications: a physicochemical study. Mater Res Express 6(6):065308. https://doi.org/10.1088/2053-1591/ab0bd0

    Article  CAS  Google Scholar 

  132. Luo J, Zhang H, Zhu J, Cui X, Gao J et al (2018) 3-D mineralized silk fibroin/polycaprolactone composite scaffold modified with polyglutamate conjugated with bmp-2 peptide for bone-tissue engineering. Colloids Surf B 163:369–378. https://doi.org/10.1016/j.colsurfb.2017.12.043

    Article  CAS  Google Scholar 

  133. Lee JU, Kim GH (2018) Three-dimensional hierarchical nanofibrous collagen scaffold fabricated using fibrillated collagen and Pluronic F-127 for regenerating bone tissue. ACS Appl Mater Interfaces 10(42):35801–35811. https://doi.org/10.1021/acsami.8b14088

    Article  CAS  Google Scholar 

  134. Abazari MF, Soleimanifar F, Faskhodi MA, Mansour RN, Mahabdi JA et al (2019) Improved osteogenic differentiation of human induced pluripotent stem cells cultured on polyvinylidene fluoride/collagen/platelet-rich plasma composite nanofibers. J Cell Physiol 235(2):1155–1164. https://doi.org/10.1002/jcp.29029

  135. Khan S, Kumar V, Roy P, Kundu PP (2019) TiO2 doped chitosan/hydroxyapatite/halloysite nanotube membranes with enhanced mechanical properties and osteoblast-like cell response for application in bone tissue engineering. RSC Adv 9(68):39768–39779. https://doi.org/10.1039/C9RA08366A

    Article  CAS  Google Scholar 

  136. Avani F, Damoogh S, Mottaghitalab F, Karkhaneh A, Farokhi M (2019) Vancomycin loaded halloysite nanotubes embedded in silk fibroin hydrogel applicable for bone tissue engineering. Int J Polym Mater Polym Biomater 69(1):32–43. https://doi.org/10.1080/00914037.2019.1616201

    Article  CAS  Google Scholar 

  137. Tang Q, Hu Z, ** H, Zheng G, Yu XF et al (2019) Microporous polysaccharide multilayer coated BCP composite scaffolds with immobilized calcitriol promote osteoporotic bone regeneration both in vitro and in vivo. Theranostics 9(4):1125–1143. https://doi.org/10.7150/thno.29566

    Article  CAS  Google Scholar 

  138. Danti S, Trombi L, Fusco A, Azimi B, Lazzeri A et al (2019) Chitin nanofibrils and nanolignin as functional agents in skin regeneration. Int J Mol Sci 20(11):2669. https://doi.org/10.3390/ijms20112669

    Article  CAS  Google Scholar 

  139. Wei J, Wang B, Li Z, Wu Z, Zhang M et al (2020) A 3D-printable tempo/oxidized bacterial cellulose/alginate hydrogel with enhanced stability via nanoclay incorporation. Carbohyd Polym 238:116207. https://doi.org/10.1016/j.carbpol.2020.116207

    Article  CAS  Google Scholar 

  140. Peng W, Ren S, Zhang Y, Fan R, Zhou Y et al (2021) MgO nanoparticles-incorporated PCL/gelatin-derived coaxial electrospinning nanocellulose membranes for periodontal tissue regeneration. Front Bioeng Biotechnol 9. https://doi.org/10.3389/fbioe.2021.668428

  141. Shao L, Wang X, Yang B, Wang Q, Tian Q et al (2017) A highly sensitive ascorbic acid sensor based on hierarchical polyaniline coated halloysite nanotubes prepared by electrophoretic deposition. Electrochim Acta 225:286–297. https://doi.org/10.1016/j.electacta.2017.09.178

    Article  CAS  Google Scholar 

  142. Wan H, Yue J, Zhu S, Uno T, Zhang X et al (2018) A bright organic NIR-II nanofluorophore for three-dimensional imaging into biological tissues. Nat Commun 9. https://doi.org/10.1038/s41467-018-03505-4

  143. Rajendrakumar SK, Venu A, Revuri V, Thomas RJ, Thirunavukkarasu GK et al (2019) Hyaluronan-stabilized redox-sensitive nanoassembly for chemogene therapy and dual T1/T2 MR imaging in drug-resistant breast cancer cells. Mol Pharm 16(5):2226–2234. https://doi.org/10.1021/acs.molpharmaceut.9b00189

    Article  CAS  Google Scholar 

  144. Jeevarathinam AS, Lemaster JE, Chen F, Zhao E, Jokerst JV (2020) Photoacoustic imaging quantifies drug release from nanocarriers via redox chemistry of dye-labelled cargo. Angew Chem Int Ed 59(12):4678–4683. https://doi.org/10.1002/anie.201914120

    Article  CAS  Google Scholar 

  145. Xu C, He XY, Ren XH, Cheng SX (2021) Direct detection of intracellular miRNA in living circulating tumor cells by tumor targeting nanoprobe in peripheral blood. Biosens Bioelectron 190:113401. https://doi.org/10.1016/j.bios.2021.113401

    Article  CAS  Google Scholar 

  146. Sim T, Choi B, Kwon SW, Kim KS, Choi H et al (2021) Magneto-activation and magnetic resonance imaging of natural killer cells labelled with magnetic nanocomplexes for the treatment of solid tumors. ACS Nano 15(8):12780–12793. https://doi.org/10.1021/acsnano.1c01889

    Article  CAS  Google Scholar 

  147. Wang YM, Xu Y, Yang ZR, Zhang X, Hu Y et al (2021) Multi-functional lanthanide coordination polymers for multi-modal detection of nitroaromatics and trace water in organic solvents. J Colloid Interface Sci 598:474–482. https://doi.org/10.1016/j.jcis.2021.04.045

    Article  CAS  Google Scholar 

  148. Shen Y, Shen G, Zhang Y (2018) Voltametric immunoassay for α-fetoprotein by using a gold nanoparticle/dendrimer conjugate and a ferrocene derived ionic liquid. Microchim Acta 185. https://doi.org/10.1007/s00604-018-2886-3

  149. Pirzada M, Altintas Z (2022) Nanomaterials for virus sensing and tracking. Chem Soc Rev 51:5805–5841. https://doi.org/10.1039/D1CS01150B

    Article  CAS  Google Scholar 

  150. Hu WP, Zhang B, Zhang J, Luo WL, Guo Y et al (2017) Ag/ alginate nanofiber membrane for flexible electronic skin. Nanotechnology 28(44):445502. https://doi.org/10.1088/1361-6528/aa8746

    Article  CAS  Google Scholar 

  151. Doğaç YI, Deveci I, Mercimek B, Teke M (2017) A comparative study for lipase immobilization onto alginate based composite electrospun nanofibers with effective and enhanced stability. Int J Biol Macromol 96:302–311. https://doi.org/10.1016/j.ijbiomac.2016.11.120

    Article  CAS  Google Scholar 

  152. Liu C, Wang T, Ji J, Wang C, Wang H et al (2019) The effect of pore size and layer numbers of metal-porphyrin coordination nanosheets on sensing DNA. J Mater Chem C 7(33):10240–12446. https://doi.org/10.1039/C9TC02778E

    Article  CAS  Google Scholar 

  153. Sehit E, Drzazgowska J, Buchenau D, Yesildag C, Lensen M et al (2020) Ultrasensitive nonenzymatic electrochemical glucose sensor based on gold nanoparticles and molecularly imprinted polymers. Biosens Bioelectron 165:112432. https://doi.org/10.1016/j.bios.2020.112432

    Article  CAS  Google Scholar 

  154. Pirzada M, Sehit E, Altintas Z (2020) Cancer biomarker detection in human serum samples using nanoparticle decorated epitope-mediated hybrid MIP. Biosens Bioelectron 166:112464. https://doi.org/10.1016/j.bios.2020.112464

    Article  CAS  Google Scholar 

  155. Sun AL (2018) Potentiometric immunosensor for enterovirus 71 based on bis-MHA-COOH-dendrimer doped AgCl nanospheres with silver ion-selective electrode. Analyst 143(2):487–492. https://doi.org/10.1039/C7AN01305A

    Article  CAS  Google Scholar 

  156. Barrios-Gumiel A, Sepúlveda-Crespo D, Jiménez JL, Gómez R, Muños-Fernández MA et al (2019) Dendronized magnetic nanoparticles for HIV-1 capture and rapid diagnostic. Colloids Surf B 181:360–368. https://doi.org/10.1016/j.colsurfb.2019.05.050

    Article  CAS  Google Scholar 

  157. Nguyen HC, Park H, Shin HJ, Nguyen NM, Nguyen ATV et al (2019) Fluorescent immunosorbent assay for detection of chikungunya virus. Intervirology 62(3–4):145–155. https://doi.org/10.1159/000502823

    Article  CAS  Google Scholar 

  158. Zhang YW, Liu WS, Chen JS, Niu HL, Mao CJ et al (2020) Metal-organic gel and metal-organic framework based switchable electroluminiscence RNA sensing platform for Zika virus. Sens Actuators B Chem 321:128456. https://doi.org/10.1016/j.snb.2020.128456

    Article  CAS  Google Scholar 

  159. Luo L, Zhang F, Chen C, Cai C (2020) Molecularly imprinting resonance light scattering nanoprobes based on pH-responsive metal-organic framework for determination of hepatitis a virus. Microchimca Acta 187. https://doi.org/10.1007/s00604-020-4122-1

  160. Kim HO, Na W, Yeom M, Lim JW, Bae EH et al (2020) Dengue virus-polymersome hybrid nanovesicles for advanced drug screening using real-time single nanoparticle-virus tracking. ACS Appl Mater Interfaces 12(6):6876–6884. https://doi.org/10.1021/acsami.9b20492

    Article  CAS  Google Scholar 

  161. Mokhena TC, Jacobs NV, Luyt AS (2017) Electrospun alginate nanofibers as potential bio-sorption agents of heavy metals in water treatment. eXPRESS Polym Lett 11(8):652–663. https://doi.org/10.3144/expresspolymlett.2017.63

  162. Geng B, Xu Z, Liang P, Zhang J, Christie P et al (2019) Three-dimensional macroscopic aminosilylated nanocellulose aerogels as sustainable bio-adsorbbents for the effective removal of heavy metal ions. Int J Biol Macromol 190:170–177. https://doi.org/10.1016/j.ijbiomac.2021.08.186

    Article  CAS  Google Scholar 

  163. Zhijiang C, Cong Z, ** X, Jie G, Kongyin Z (2018) Calcium alginate-coated electrospun polyhydroxybutyrate/carbon nanotubes composite nanofibers as nanofiltration membrane for dye removal. J Mater Sci 53:14801–14820. https://doi.org/10.1007/s10853-018-2607-7

    Article  CAS  Google Scholar 

  164. Soon CY, Rahman NA, Bond TY, Talib RA, Tan CH et al (2019) Electrospun biocomposite: nanocellulose and chitosan entrapped within a poly(hydroxyalkanoate) matrix for congo red removal. J Mater Res Technol 8(6). https://doi.org/10.1016/j.jmrt.2019.08.030

  165. Derami HG, Gupta P, Gupta R, Rathi P, Morrissey JJ et al (2020) Palladium nanoparticle-decorated mesoporous polydopamine/bacterial nanocellulose as a catalytically active universal dye removal ultrafiltration membrane. ACS Appl Nano Mater 3(6):5437–5448. https://doi.org/10.1021/acsanm.0c00787

    Article  CAS  Google Scholar 

  166. Zhang W, Wang X, Zhang Y, van Bochove B, Mäkilä E et al (2020) Robust shape-retaining nanocellulose-based aerogels decorated with silver nanoparticles for fast continuous catalytic discoloration of organic dyes. Sep Purif Technol 242:116523. https://doi.org/10.1016/j.seppur.2020.116523

    Article  CAS  Google Scholar 

  167. Sun H, Jiang J, **ao Y, Du J (2018) Efficient removal of polyacyclic aromatic hydrocarbons, dyes, and heavy metal ions by a homopolymer vesicle. ACS Appl Mater Interfaces 10(1):713–720. https://doi.org/10.1021/acsami.7b15242

    Article  CAS  Google Scholar 

  168. Sijabat EK, Nurrudin A, Aditiawati P, Purwasasmita BS (2019) Synthesis and characterization of bacterial nanocellulose from banana peel for water filtration membrane application. J Phys Conf Ser 1230:012085. https://doi.org/10.1088/1742-6596/1230/1/012085

    Article  CAS  Google Scholar 

  169. Yang M, Hadi P, Yin X, Yu J, Huang X et al (2020) Antifouling nanocellulose membranes: how subtle adjustment of surface charge lead to self-cleaning property. J Membr Sci 618:118739. https://doi.org/10.1016/j.memsci.2020.118739

    Article  CAS  Google Scholar 

  170. Hassan ML, Fadel SM, Abouzeid RE, Elseoud WSA, Hassan EA et al (2020) Water purification ultrafiltration membranes using nanofibers from unbleached and bleached rice straw. Sci Rep 10. https://doi.org/10.1038/s41598-020-67909-3

  171. Wang M, Sun T, Wan D, Dai M, Ling S et al (2021) Solar-powered nanostructured biopolymer hygroscopic aerogels for atmospheric water harvesting. Nano Energy 80:105569. https://doi.org/10.1016/j.nanoen.2020.105569

    Article  CAS  Google Scholar 

  172. Jahan Z, Niazi MBK, Hägg MB, Gregerson WØ (2018) Decoupling the effect of membrane thickness and CMC concentration in PVA based nanocomposite membranes for CO2/CH4 separation. Sep Purif Technol 204:220–225. https://doi.org/10.1016/j.seppur.2018.04.076

    Article  CAS  Google Scholar 

  173. Torstensen JØ, Helberg RML, Deng L, Gregerson ØW, Syverud K (2019) PVA/nanocellulose nanocomposite membranes for CO2 separation from fuel gas. Int J Greenhouse Gas Control 81:93–102. https://doi.org/10.1016/j.ijggc.2018.10.007

    Article  CAS  Google Scholar 

  174. Helberg RML, Torstensen JØ, Dai Z, Janakiram S, Chinga-Carrasco G et al (2021) Nanocomposite membranes with high-charge and size-screened phosphorylated nanocellulose fibrils for CO2 separation. Green Energy Environ 6(4):585–596. https://doi.org/10.1016/j.gee.2020.08.004

    Article  CAS  Google Scholar 

  175. Wang X, Yao C, Wang F, Li Z (2017) Cellulose-based nanomaterials for energy applications. Small 13(42):1702240. https://doi.org/10.1002/smll.201702240

    Article  CAS  Google Scholar 

  176. He X, Zou H, Geng Z, Wang X, Ding W et al (2018) A hierarchically nanostructured cellulose fibre-based triboelectric nanogenerator for self-powered healthcare products. Adv Func Mater 28–45:1805540. https://doi.org/10.1002/adfm.201805540

    Article  CAS  Google Scholar 

  177. Kim K, Ha M, Choi B, Joo SH, Kang HS et al (2018) Bio-degradable, electro-active chitin nanofiber films for flexible piezoelectric transducers. Nano Energy 48:275–283. https://doi.org/10.1016/j.nanoen.2018.03.056

    Article  CAS  Google Scholar 

  178. Liu C, Li J, Che L, Chen S, Wang Z et al (2017) Toward large-scale fabrication of triboelectric nanogenerator (TENG) with silk-fibroin patches film via spray-coating process. Nano Energy 41:359–366. https://doi.org/10.1016/j.nanoen.2017.09.038

    Article  CAS  Google Scholar 

  179. Karan SK, Maiti S, Kwon O, Paria S, Maitra A et al (2018) Nature driven spider silk as high energy conversion efficient bio-piezoelectric nanogenerator. Nano Energy 49:655–666. https://doi.org/10.1016/j.nanoen.2018.05.014

    Article  CAS  Google Scholar 

  180. Niu Q, Huang L, Lv S, Shao H, Fan S et al (2020) Pulse-driver biotriboelectric nanogenerators based on silk nanoribbons. Nano Energy 74:104837. https://doi.org/10.1016/j.nanoen.2020.104837

    Article  CAS  Google Scholar 

  181. Nguyen V, Kelly S, Yang R (2017) Piezoelectric peptide-based nanogenerator enhanced by single-electrode triboelectric nanogenerator. APL Mater 5:074108. https://doi.org/10.1063/1.4983701

    Article  CAS  Google Scholar 

  182. Jenkins K, Kelly S, Nguyen V, Wu Y, Yang R (2018) Piezoelectric diphenylalanine peptide for greatly improved flexible nanogenerators. Nano Energy 51:317–323. https://doi.org/10.1016/j.nanoen.2018.06.061

    Article  CAS  Google Scholar 

  183. Ji W, Xue B, Arnon ZA, Yuan H, Bera S et al (2019) Rigid tightly packed amino acid crystals as functional supramolecular materials. ACS Nano 13(12):14477–14485. https://doi.org/10.1021/acsnano.9b08217

    Article  CAS  Google Scholar 

  184. Wang R, Gao S, Yang Z, Li Y, Chen W et al (2018) Engineered and laser-processed chitosan biopolymers for sustainable and biodegradable triboelectric power generation. Adv Mater 30(11):17062–17067. https://doi.org/10.1002/adma.201706267

    Article  CAS  Google Scholar 

  185. Khalil AM, Hassan ML, Ward AA (2017) Novel nanofibrillated cellulose/ polyvinylpyrrolidone/silver nanoparticles films with electrical conductivity properties. Carbohyd Polym 157:503–511. https://doi.org/10.1016/j.carbpol.2016.10.008

    Article  CAS  Google Scholar 

  186. Sun JG, Yang TN, Kuo IS, Wu JM, Wang CY et al (2017) A leaf-molded transparent triboelectric nanogenerator for smart multifunctional applications. Nano Energy 32:180–186. https://doi.org/10.1016/j.nanoen.2016.12.032

    Article  CAS  Google Scholar 

  187. Lee JH, Heo K, Schulz-Schönhagen K, Lee JH, Desai MS et al (2018) Diphenylalanine peptide nanotube energy harvesters. ACS Nano 12(8):8138–8144. https://doi.org/10.1021/acsnano.8b03118

    Article  CAS  Google Scholar 

  188. Bhakat D, Barik P, Bhattacharjee A (2018) Electrical conductivity behavior of gum arabic biopolymer-Fe3O4 nanocomposites. J Phys Chem Solids 112:73–79. https://doi.org/10.1016/j.jpcs.2017.09.002

    Article  CAS  Google Scholar 

  189. Wang M, Zhang J, Tang Y, Li J, Zhang B et al (2018) Air-flow-driven triboelectric nanogenerators for self-powered real-time respiratory monitoring. ACS Nano 12(6):6156–6162. https://doi.org/10.1021/acsnano.8b02562

    Article  CAS  Google Scholar 

  190. Šutka A, Ruža J, Järvekülg M, Linarts A, Mālnieks K et al (2018) Triboelectric nanogenerator based on immersion precipitation derived from highly porous ethyl cellulose. J Electrostat 92:1–5. https://doi.org/10.1016/j.elstat.2018.01.003

    Article  CAS  Google Scholar 

  191. Gao L, Chao L, Hou M, Liang J, Chen Y et al (2019) Flexible, transparent nanocellulose paper based-perovskite solar cells. npj Flex Electron 3. https://doi.org/10.1038/s41528-019-0048-2

  192. Liu Z, Ma Y, Ouyang H, Shi B, Li N et al (2018) Transcatheter self-powered ultrasensitive endocardial pressure sensor. Adv Func Mater 29(3):1807560. https://doi.org/10.1002/adfm.201807560

    Article  CAS  Google Scholar 

  193. Ccorahua R, Huaroto J, Luyo C, Quintana M, Vela EA (2019) Enhanced-performance bio-triboelectric nanogenerator based on starch polymer electrolyte obtained by a cleanroom-free processing method. Nano Energy 59:610–618. https://doi.org/10.1016/j.nanoen.2019.03.018

    Article  CAS  Google Scholar 

  194. Liu J, Yang B, Lu L, Wang X, Li X et al (2019) Flexible and lead-free piezoelectric nanogenerator based on electrospinning BZT-BCT-P(VDF-TRFE) nanofibers. Sens Actuators Phys 303:11796. https://doi.org/10.1016/j.sna.2019.111796

  195. Salvioni L, Morreli L, Ochoa E, Labra M, Fiandra L et al (2021) The emerging role of nanotechnology in skincare. Adv Coll Interface Sci 293:102437. https://doi.org/10.1016/j.cis.2021.102437

    Article  CAS  Google Scholar 

  196. Castleberry SA, Quadir MA, Sharkh MA, Shopsowitz KE, Hammond PT (2017) Polymer conjugated retinoids for controlled transdermal delivery. J Control Release 262:1–9. https://doi.org/10.1016/j.jconrel.2017.07.003

    Article  CAS  Google Scholar 

  197. Pegoraro NS, Barbieri AV, Camponogara C, Mattiazzi J, Brum ES et al (2017) Nanoencapsulation of coenzyme Q10 and vitamin e acetate protects against UVB-radiation induced skin injury in mice. Colloids Surf B Biointerfaces 150:32–40. https://doi.org/10.1016/j.colsurfb.2016.11.013

    Article  CAS  Google Scholar 

  198. El-Leithy ES, Makky AM, Khattab AM, Hussein DG (2018) Optimization of nutraceutical coenzyme Q10 nanoemulsion with improved skin permeability and anti- wrinkle activity. Drug Dev Ind Pharm 44(2):316–328. https://doi.org/10.1080/03639045.2017.1391836

    Article  CAS  Google Scholar 

  199. Su R, Yang L, Wang Y, Yu WS, Guo Y et al (2017) Formulation, development, and optimization of a novel octyldodecanol-based nanoemulsion for transdermal delivery of ceramide IIIB. Int J Nanomed 12:5203–5221. https://doi.org/10.2147/IJN.S139975

    Article  CAS  Google Scholar 

  200. Chen J, Wei N, Lopez-Garcia M, Ambrose D, Lee J et al (2017) Development and evaluation of resveratrol, vitamin E, and epigallocatechin gallate loaded lipid nanoparticles for skin care applications. Eur J Pharm Biopharm 117:286–291. https://doi.org/10.1016/j.ejpb.2017.04.008

    Article  CAS  Google Scholar 

  201. Avadhani KS, Manikkath J, Tiwari M, Chandrasekhar M, Godavarthi A et al (2017) Skin delivery of epigallocatechin-3-gallate (EGCG) and hyaluronic acid loaded nano-transferosomes for antioxidant and anti-ageing effect in UV radiation-induced skin damage. Drug Deliv 24(1):61–74. https://doi.org/10.1080/10717544.2016.1228718

    Article  CAS  Google Scholar 

  202. Muzzalupo R, Pérez L, Pinazo A, Tavano L (2017) Pharmaceutical versatility of cationic niosomes derived from amino acid-based surfactants: skin penetration behaviour and controlled drug release. Int J Pharm 529(1–2):245–252. https://doi.org/10.1016/j.ijpharm.2017.06.083

    Article  CAS  Google Scholar 

  203. Pentek T, Newenhouse E, O’Brien B, Chauhan AS (2017) Development of a topical resveratrol formulation for commercial applications using dendrimer nanotechnology. Molecules 22(1):137. https://doi.org/10.3390/molecules22010137

    Article  Google Scholar 

  204. Holmes AM, Scurr DJ, Heylings JR, Wan KW, Moss GP (2017) Dendrimer pre-treatment enhances the skin permeation of chlorhexidine digluconate: characterization by in vitro percutaneous absorption studies and time-of-flight secondary ion mass spectrometry. Eur J Pharm Sci 104:90–101. https://doi.org/10.1016/j.ejps.2017.03.034

    Article  CAS  Google Scholar 

  205. Ki P, ** P, II K, Hyun K (2017) Fermented extract promoting hair growth-hydrogel nanoparticle hybrid and method for preparing the same. CARI LTD, Editor, KR20170121454A

    Google Scholar 

  206. Ayumi NS, Sahudin S, Hussain Z, Hussain M, Samah NHA (2018) Polymeric nanoparticles for topical delivery of alpha and beta arbutin: preparation and characterization. Drug Deliv Transl Res 9(2):482–496. https://doi.org/10.1007/s13346-018-0508-6

    Article  CAS  Google Scholar 

  207. Panchal A, Fakhrullina G, Fakhrullin R, Lvov Y (2018) Self-assembly of clay nanotubes on hair surface for medical and cosmetic formulations. Nanoscale 10(38):18205–18216. https://doi.org/10.1039/C8NR05949G

    Article  CAS  Google Scholar 

  208. Ntohogian S, Gavriliadou V, Christodoulou E, Nanaki S, Lykidou S et al (2018) Chitosan nanoparticles encapsulated with natural and UF-purified annatto and saffron for the preparation of UV-protective cosmetic emulsions. Molecules 23(9):2107. https://doi.org/10.3390/molecules23092107

    Article  CAS  Google Scholar 

  209. Hwa L, Sun S, ** B, MIN H, GYU K (2018) Stably-suspended polymeric composite nanoparticles containing organic sunscreen agents and preparing method thereof. LHH CARE, Editor, KR20200132092A

    Google Scholar 

  210. Leon-Méndez G, Osorio M, Ortego-Toto R, Pajaro-Castro N, Torrenegra ME et al (2018) Design of an emulgel-type cosmetic with antioxidant activity using essential oil microcapsules of thyme (Thymus vulgaris L.), cinnamon (Cinnamomum verum J.) and clove (Eugenia caryophyllata T.). Int J Polym Sci 2018:1–16. https://doi.org/10.1155/2018/2874391

    Article  CAS  Google Scholar 

  211. Hatahet T, Morille M, Hommoss A, Devoiselle JM, Müller RH (2018) Liposomes, lipid nanoparticles and SmartCrystal®: a comparative study for an effective quercetin delivery to the skin. Int J Pharm 542(1–2):176–185. https://doi.org/10.1016/j.ijpharm.2018.03.019

    Article  CAS  Google Scholar 

  212. Tokudome Y, Komi T, Omata A, Sekita M (2018) A new strategy for the passive skin delivery of nanoparticulate, high molecular weight hyaluronic acid prepared by a polyion complex method. Sci Rep 8. https://doi.org/10.1038/s41598-018-20805-3

  213. Alhakamy NA, Hosny KM (2019) Nano-vesicular delivery system loaded by bifonazole: preparation, optimization, and assessment of pharmacokinetic and antifungal activity. J Drug Deliv Sci Technol 49:316–322. https://doi.org/10.1016/j.jddst.2018.11.020

    Article  CAS  Google Scholar 

  214. Gupta S, Wairkar S, Bhatt LK (2020) Isotretenoin and α-tocopherol acetate loaded solid lipid nanoparticle topical gel for the treatment of acne. J Microencapsul 37(8):557–565. https://doi.org/10.1080/02652048.2020.1823499

    Article  CAS  Google Scholar 

  215. Sliem MA, Karas RA, Harith MA (2017) A promising protected ascorbic acid-hydroxyapatite nanocomposite as a skin anti-ager: a detailed photo- and thermal stability study. J Photochem Photobiol B Biol 173:661–671. https://doi.org/10.1016/j.jphotobiol.2017.07.004

    Article  CAS  Google Scholar 

  216. Maione-Silva L, de Castro EG, Nascimento TL, Cintra ER, Moreira LC et al (2019) Ascorbic acid encapsulated into negatively charged liposomes exhibits increased skin permeation, retention and enhances collagen synthesis by fibroblasts. Sci Rep 9. https://doi.org/10.1038/s41598-018-36682-9

  217. Aboul-Einien MH, Kandil SM, Abdou EM, Diab HM, Zaki MSE (2020) Ascorbic acid-derived modified aspasomes: formulation, in vivo, ex vivo and clinical evaluation for melasma treatment. J Liposome Res 30(1):54–67. https://doi.org/10.1080/08982104.2019.1585448

    Article  CAS  Google Scholar 

  218. Zhu JJ, Huang XN, Yang T, Tang CH, Yin SW et al (2022) An eco-friendly zein nanoparticle as robust cosmetic ingredient ameliorates skin photoaging. Ind Crops Prod 177:114521. https://doi.org/10.1016/j.indcrop.2022.114521

    Article  CAS  Google Scholar 

  219. Vale EP, Morais ES, Tavares WS, de Souza FFO (2022) Epigallocatechin-3-gallate loaded zein nanoparticles: characterization, stability and associated antioxidant, anti-tyrosinase and sun protection properties. J Mol Liq 358:119107. https://doi.org/10.1016/j.molliq.2022.119107

    Article  CAS  Google Scholar 

  220. Katiyar V, Ghosh T (2021) Edible food packaging: an introduction. In: Katiyar V, Ghosh T (eds) Nanotechnology in edible food packaging. Springer Singapore, pp 1–23. 978-981-33-6169-0

    Google Scholar 

  221. Corral ML, Cerrutti P, Vázquez A, Califano A (2017) Bacterial cellulose as a potential additive for wheat bread. Food Hydrocolloids 67:189–196. https://doi.org/10.1016/j.foodhyd.2016.11.037

    Article  CAS  Google Scholar 

  222. Zbikowska A, Kowalska M, Pieniowska J (2018) Assessment of shortcrust biscuits with reduced fat content of microcyrstalline cellulose and psyllium as fat replacements. J Food Proc Preserv 42(8). https://doi.org/10.1111/jfpp.13675

  223. Collar C (2019) Gluten-free dough-based foods and technologies. In: Taylor JRN, Duodu KG (eds) Sorghum and millets (Second Edition). AACC International Press, pp 331–354. https://doi.org/10.1016/B978-0-12-811527-5.00011-3

  224. Marchetti L, Muzzio B, Cerrutti P, Andrés SC, Califano AN (2017) Bacterial nanocellulose as novel additive in low-lipid low-sodium meat sausages. Food Struct 14:52–59. https://doi.org/10.1016/j.foostr.2017.06.004

    Article  Google Scholar 

  225. Tesfay SZ, Magwaza LS (2017) Evaluating the efficacy of moringa leaf extract, chitosan and carboxymethyl cellulose as edible coatings for enhancing quality and extending postharvest life of avocado (Persea americana Mill.) fruit. Food Packag Shelf Life 11:40–48. https://doi.org/10.1016/j.fpsl.2016.12.001

    Article  Google Scholar 

  226. Deng Z, Jung J, Zhao Y (2017) Development, characterization, and validation of chitosan adsorbed cellulose nanofiber (CNF) films as water resistant and antibacterial food contact packaging. LWT—Food Sci Technol 83:132–140. https://doi.org/10.1016/j.lwt.2017.05.013

  227. Yu Z, Alsammarraie FK, Nayigiziki FX, Wang W, Vardhanabhuti B et al (2017) Effect and mechanism of cellulose nanofibrils on the active functions of biopolymer-based nanocomposite films. Food Res Int 99(1):166–172. https://doi.org/10.1016/j.foodres.2017.05.009

    Article  CAS  Google Scholar 

  228. Cazón P, Vázquez M, Velazquez G (2018) Cellulose-glycerol-polyvinyl alcohol composite films for food packaging: evaluation of water adsorption, mechanical properties, light-barrier properties, and transparency. Carbohyd Polym 195:432–443. https://doi.org/10.1016/j.carbpol.2018.04.120

    Article  CAS  Google Scholar 

  229. Costa ALR, Gomes A, Tibolla H, Menegalli FC, Cunha RL (2018) Cellulose nanofibers from banana peels as a pickering emulsifier: high-energy emulsification processes. Carbohyd Polym 194:122–131. https://doi.org/10.1016/j.carbpol.2018.04.001

    Article  CAS  Google Scholar 

  230. 2019—Pickering emulsions stabilized by hydrophobically modified nanocellulose containing various structural characteristics

    Google Scholar 

  231. Winuprasith T, Khomein P, Mitbumrung W, Suphantharika M, Nitithamyong A et al (2018) Encapsulation of vitamin D3 in Pickering emulsions stabilized by nanofibrillated mangosteen cellulose: impact on in vitro digestion and bioaccessibility. Food Hydrocolloids 83:153–164. https://doi.org/10.1016/j.foodhyd.2018.04.047

    Article  CAS  Google Scholar 

  232. Luan Q, Zhou W, Zhang H, Bao Y, Zheng M et al (2018) Cellulose-based composite macrogels from cellulose fibre and cellulose nanofiber as intestinal delivery vehicle for probiotics. J Agric Food Chem 66(1):339–345. https://doi.org/10.1021/acs.jafc.7b04754

    Article  CAS  Google Scholar 

  233. Velásquez-Cock J, Serpa A, Vélez L, Gañán P, Hoyos CG et al (2019) Influence of cellulose nanofibrils on the structural elements of ice cream. Food Hydrocolloids 87:2014–2213. https://doi.org/10.1016/j.foodhyd.2018.07.035

    Article  CAS  Google Scholar 

  234. Liu L, Kong F (2019) Influence of nanocellulose on in vitro digestion of whey protein isolate. Carbohyd Polym 210:399–411. https://doi.org/10.1016/j.carbpol.2019.01.071

    Article  CAS  Google Scholar 

  235. Ngo TMP, Nyugen TH, Dang TMQ, Tran TX, Rachtanapun P (2020) Characteristics and antimicrobial properties of active edible films based on pectin and nanochitosan. Int J Mol Sci 21(6):2224. https://doi.org/10.3390/ijms21062224

    Article  CAS  Google Scholar 

  236. Kumar S, Mukherjee A, Dutta J (2020) Chitosan-based nanocomposite films and coatings: emerging antimicrobial food packaging alternatives. Trends Food Sci Technol 97:196–209. https://doi.org/10.1016/j.tifs.2020.01.002

    Article  CAS  Google Scholar 

  237. Divya K, Smitha V, Jisha MS (2018) Antifungal, antioxidant and cytotoxic activities of chitosan nanoparticles and its use as an edible coating on vegetables. Int J Biol Macromol 114:572–577. https://doi.org/10.1016/j.ijbiomac.2018.03.130

    Article  CAS  Google Scholar 

  238. Lustriane C, Dwivany FM, Suendo V, Reza M (2018) Effect of chitosan and chitosan nanoparticles on post-harvest quality of banana fruits. J Plant Biotechnol 45(1):36–44. https://doi.org/10.5010/JPB.2018.45.1.036

    Article  Google Scholar 

  239. Cao Y, Warner RD, Fang Z (2019) Effect of chitosan/ nisin/ gallic acid coating on preservation of pork loin in high oxygen modified atmosphere packaging. Food Control 101:9–16. https://doi.org/10.1016/j.foodcont.2019.02.013

    Article  CAS  Google Scholar 

  240. Javaherzadeh R, Bafroee AST, Kanjari A (2020) Preservation effect of Polylophium involucratum essential oil incorporated poly lactic acid/ nanochitosan composite film on shelf life and sensory properties of chicken fillets at refrigeration temperature. LWT 118:108783. https://doi.org/10.1016/j.lwt.2019.108783

    Article  CAS  Google Scholar 

  241. Luchese CL, Garrido T, Spada JC, Tessaro IC (2018) Development and characterization of cassava starch films incorporated with blueberry pomace. Int J Biol Macromol 106:834–839. https://doi.org/10.1016/j.ijbiomac.2017.08.083

    Article  CAS  Google Scholar 

  242. Dai L, Zhang J, Cheng F (2020) Cross-linked starch-based edible coating reinforced by starch nanocrystals and its preservation effects on huangguan pear. Food Chem 311:125891. https://doi.org/10.1016/j.foodchem.2019.125891

    Article  CAS  Google Scholar 

  243. Roy K, Theory R, Sinhmar A, Pathera AK, Nain V (2020) Development and characterization of nano-starch based composite films from mung beans (Vigna radiata). Int J Biol Macromol 144:242–251. https://doi.org/10.1016/j.ijbiomac.2019.12.113

    Article  CAS  Google Scholar 

  244. Sanyang ML, Ilyas RA, Sapuan SM, Jumaidin R (2018) Sugar palm starch-based composites for packaging applications. In: Jawaid M, Swain S (eds) Bionanocomposites for packaging applications. Springer, Cham, pp 125–147. https://doi.org/10.1007/978-3-319-67319-6_7

  245. Podshivalov A, Zakharova M, Glazacheva E, Uspenskaya M (2017) Gelatin/potato starch edible biocomposite films: correlation between morphology and physical properties. Carbohyd Polym 157:1162–1172. https://doi.org/10.1016/j.carbpol.2016.10.079

    Article  CAS  Google Scholar 

  246. Zakaria NH, Muhammad N, Sandu AV, Abdullah MMAB (2018) Effect of mixing temperature on characteristics of thermoplastic potato starch film. IOP Conf Ser Mater Sci Eng 374:012083. https://doi.org/10.1088/1757-899X/374/1/012083

    Article  Google Scholar 

  247. Yang J, Li F, Li M, Zhang S, Liu J et al (2017) Fabrication and characterization of hollow starch nanoparticles by gelation process for drug delivery application. Carbohyd Polym 173:223–232. https://doi.org/10.1016/j.carbpol.2017.06.006

    Article  CAS  Google Scholar 

  248. Farrag Y, Ide W, Montero B, Rico M, Rodríguez-Llamazares S et al (2018) Preparation of starch nanoparticles loaded with quercetin using nanoprecipitation technique. Int J Biol Macromol 114:426–433. https://doi.org/10.1016/j.ijbiomac.2018.03.134

    Article  CAS  Google Scholar 

  249. Ahmad M, Mudgil P, Gani A, Hamed F, Masoodi FA et al (2019) Nano-encapsulation of catechin in starch nanoparticles: characterization, release behavior and bioactivity retention during simulated in vitro digestion. Food Chem 270:95–104. https://doi.org/10.1016/j.foodchem.2018.07.024

    Article  CAS  Google Scholar 

  250. Suaza-Nieto L, Acevedo-Guevara L, Sánchez LT, Pinzón MI, Villa CC (2019) Characterization of aloe vera-banana starch composite films reinforced with curcumin loaded starch nanoparticles. Food Struct 22:100131. https://doi.org/10.1016/j.foostr.2019.100131

    Article  Google Scholar 

  251. Escobar-Puentes AA, García-Gurrola A, Rincón S, Zepeda A, Martínez-Bustos F (2020) Effect of amylose/amylopectin content and succinylation on properties of corn starch nanoparticles as encapsulants of anthocyanins. Carbohyd Polym 250:116972. https://doi.org/10.1016/j.carbpol.2020.116972

    Article  CAS  Google Scholar 

  252. Ahmad M, Gani A (2021) Ultrasonicated resveratrol loaded starch nanocapsules: characterization, bioactivity and release behaviour under in-vitro digestion. Carbohyd Polym 251:117111. https://doi.org/10.1016/j.carbpol.2020.117111

    Article  CAS  Google Scholar 

  253. Pei X, Zhai K, Liang X, Deng Y, Tan Y et al (2017) Interfacial activity of starch-based nanoparticles at the oil–water interface. Langmuir 33(15):3787–3793. https://doi.org/10.1021/acs.langmuir.7b00035

    Article  CAS  Google Scholar 

  254. Javidi F, Razavi SMA, Amini AM (2019) Cornstarch nanocrystals as a potential fat replacer in reduced fat o/w emulsions: a rheological and physical study. Food Hydrocolloids 90:172–181. https://doi.org/10.1016/j.foodhyd.2018.12.003

    Article  CAS  Google Scholar 

  255. Chang S, Chen X, Liu S, Wang C (2020) Novel gel-like Pickering emulsions stabilized solely by hydrophobic starch nanocrystals. Int J Biol Macromol 152:703–708. https://doi.org/10.1016/j.ijbiomac.2020.02.175

    Article  CAS  Google Scholar 

  256. Dai L, Sun C, Li R, Mao L, Liu F et al (2017) Structural characterization, formation mechanism and stability of curcumin in zein-lecithin composite nanoparticles fabricated by antisolvent co-precipitation. Food Chem 237:1163–1171. https://doi.org/10.1016/j.foodchem.2017.05.134

    Article  CAS  Google Scholar 

  257. Hasankhan S, Tabibiazar M, Hosseini SM, Ehsani A, Ghorbani M (2020) Fabrication of curcumin-zein-ethyl cellulose composite nanoparticles using antisolvent co-precipitation method. Int J Biol Macromol 163:1538–1545. https://doi.org/10.1016/j.ijbiomac.2020.08.045

    Article  CAS  Google Scholar 

  258. Cheng CJ, Ferruzzi M, Jones OG (2019) Fate of lutein-containing zein nanoparticles following simulated gastric and intestinal digestion. 87:229–236. https://doi.org/10.1016/j.foodhyd.2018.08.013

  259. Ma JJ, Yu YG, Yin SW, Tang CH, Yang XQ (2018) Cellular uptake and intracellular antioxidant activity of zein/chitosan nanoparticles incorporated with quercetin. J Agric Food Chem 66(48):12783–12793. https://doi.org/10.1021/acs.jafc.8b04571

    Article  CAS  Google Scholar 

  260. Zhou JF, Zheng GD, Wang WJ, Yin ZP, Chen JG et al (2021) Physicochemical properties and bioavailability comparison of two quercetin loading zein nanoparticles with outer shell of caseinate and chitosan. Food Hydrocolloids 120:106959. https://doi.org/10.1016/j.foodhyd.2021.106959

    Article  CAS  Google Scholar 

  261. Zhang F, Khan MA, Cheng H, Liang L (2019) Co-encapsulation of α-tocopherol and resveratrol within zein nanoparticles: impact on antioxidant activity and stability. J Food Eng 247:9–18. https://doi.org/10.1016/j.jfoodeng.2018.11.021

    Article  CAS  Google Scholar 

  262. Li H, Xu Y, Sun X, Wang S, Zhu J et al (2018) Stability, bioactivity, and bioaccessibility of fucoxanthin in zein-caseinate composite nanoparticles fabricated at neutral pH by antisolvent precipitation. Food Hydrocolloids 84:379–388. https://doi.org/10.1016/j.foodhyd.2018.06.032

    Article  CAS  Google Scholar 

  263. Ju M, Zhu G, Huang G, Shen X, Zhang Y (2019) A novel pickering emulsion produced using soy protein-anthocyanin complex nanoparticles. Food Hydrocolloids 99:105329. https://doi.org/10.1016/j.foodhyd.2019.105329

    Article  CAS  Google Scholar 

  264. Feng Z, Li L, Wang Q, Wu G, Liu C et al (2019) Effect of antioxidant and antimicrobial coating based on whey protein nanofibrils with TiO2 nanotubes on the quality and shelf-life of chilled meat. Int J Mol Sci 20(5):1184. https://doi.org/10.3390/ijms20051184

    Article  CAS  Google Scholar 

  265. Wang Q, Liu W, Tian B, Li D, Liu C et al (2020) Preparation and characterization of coating based on protein nanofibers and polyphenol and application for salted duck egg yolks. Foods 9(4):449. https://doi.org/10.3390/foods9040449

    Article  CAS  Google Scholar 

  266. Wang Q, Yu H, Tian B, Jiang B, Xu J et al (2019) Novel edible coating with antioxidant and antimicrobial activities based on whey protein isolate nanofibrils and carvacrol and its application on fresh-cut cheese. Coatings 9(9):583. https://doi.org/10.3390/coatings9090583

    Article  CAS  Google Scholar 

  267. Bora A, Mishra P (2019) Casein and Ag nanoparticles synthesis, characterization, and their application in biopolymer-based bilayer film. J Food Proc Preserv 43(9). https://doi.org/10.1111/jfpp.14062

  268. Leena MM, Yoha KS, Moses JA, Anandharamakrishnan C (2020) Edible coating with resveratrol loaded electrospun zein nanofibers with enhanced bioaccessibility. Food Biosci 36:100669. https://doi.org/10.1016/j.fbio.2020.100669

    Article  CAS  Google Scholar 

  269. Yi F, Wu K, Yu G, Su C (2021) Preparation of pickering emulsion based on soy protein isolate—gallic acid with outstanding antioxidation and antimicrobial. Colloids Surf B 206:111954. https://doi.org/10.1016/j.colsurfb.2021.111954

    Article  CAS  Google Scholar 

  270. Cui H, Yuan L, Lin L (2017) Edible film incorporated with chitosan and Artemisia annua oil nanoliposomes for inactivation of Escherichia coli O157:H7 on cherry tomato. Int J Food Sci Technol 52(3):687–698. https://doi.org/10.1111/ijfs.13322

    Article  CAS  Google Scholar 

  271. García-Betanzos CI, Hernandéz-Sánchez H, Bernal-Couoh TF, Quintanar-Guerrero D, Zambrano-Zaragoza ML (2017) Physicochemical, total phenols and pectin methylesterase changes on quality maintenance on guava fruit (Psidium guajava L.) coated with candeuba wax solid lipid nanoparticles-xanthan gum. Food Res Int 101:218–227. https://doi.org/10.1016/j.foodres.2017.08.065

    Article  CAS  Google Scholar 

  272. Robledo N, López L, Bunger A, Tapia C, Abugoch L (2018) Effects of antimicrobial edible coating of thymol nanoemulsion/quinoa protein/chitosan on the safety, sensorial properties, and quality of refrigerated strawberries (Fragaria ananassa) under commercial storage environment. Food Bioprocess Technol 11:1566–1574. https://doi.org/10.1007/s11947-018-2124-3

    Article  CAS  Google Scholar 

  273. Miranda-Linares V, Escamilla-Rendón P, Real AD, González-Reza RM, Zambrano-Zaragoza ML (2018) Solid lipid nanoparticles based edible coating for saladette tomato preservation. Acta Hort 1194:305–312. https://doi.org/10.17660/ActaHortic.2018.1194.44

    Article  Google Scholar 

  274. Zambrano-Zaragoza ML, Quintanar-Guerrero D, Real AD, González-Reza RM, Cornejo-Villegas MA et al (2020) Effect of nano-edible coating based on beeswax solid lipid nanoparticles on strawberry’s preservation. Coatings 10(3):253. https://doi.org/10.3390/coatings10030253

    Article  CAS  Google Scholar 

  275. Pabast M, Shariatifar N, Beikzadeh S, Jahed G (2018) Effects of chitosan coatings incorporating with free or nano-encapsulated Satureja plant essential oil on quality characteristics of lamb meat. Food Control 91:185–192. https://doi.org/10.1016/j.foodcont.2018.03.047

    Article  CAS  Google Scholar 

  276. Amjadi S, Nazari M, Alizadeh SA, Hamishekhar H (2020) Multifunctional betanin nanoliposomes-incorporated gelatin/chitosan nanofiber/ZnO nanoparticles nanocomposite film for fresh beef preservation. Meat Sci 167:108161. https://doi.org/10.1016/j.meatsci.2020.108161

    Article  CAS  Google Scholar 

  277. **ong Y, Li S, Warner RD, Fang Z (2020) Effect of oregano essential oil and resveratrol nanoemulsion loaded pectin edible coating on the preservation of pork loin in modified atmosphere packaging. Food Control 114:107226. https://doi.org/10.1016/j.foodcont.2020.107226

    Article  CAS  Google Scholar 

  278. Pascual-Villalobos MJ, Guirao P, Díaz-Baños FG, Cantó-Tejero M, Villora G (2019) Oil in water nanoemulsion formulations of botanical active substances, In: Koul O (ed) Nano-biopesticides today and future perspective. Academic Press, pp 223–247. https://doi.org/10.1016/B978-0-12-815829-6.00009-7

  279. Pascual-Villalobos MJ, Cantó-Tejero M, Vallejo R, Guirao P, Rodríguez-Rojo S et al (2017) Use of nanoemulsions of plant essential oils as aphid repellents. Ind Crops Prod 110:45–57. https://doi.org/10.1016/j.indcrop.2017.05.019

    Article  CAS  Google Scholar 

  280. Sandhya KS, Kumar D, Dilbaghi N (2017) Preparation, characterization, and bio-efficacy evaluation of controlled release carbendazim-loaded polymeric nanoparticles. Environ Sci Pollut Res Int 24(1):926–937. https://doi.org/10.1007/s11356-016-7774-y

    Article  CAS  Google Scholar 

  281. Choudhary RC, Kumaraswamy RV, Kumari S, Sharma SS, Pal A et al (2017) Cu-chitosan nanoparticle boost defense responses and plant growth in maize (Zea mays L.). Sci Rep 7. https://doi.org/10.1038/s41598-017-08571-0

  282. Kottegoda N, Sandaruwan C, Priyadarshana G, Siriwardhana A, Rathnayake UA et al (2017) Urea-hydroxyapatite nanohybrids for the slow release of nitrogen. ACS Nano 11(2):1214–1221. https://doi.org/10.1021/acsnano.6b07781

    Article  CAS  Google Scholar 

  283. Liang J, Yu M, Guo L, Cui B, Zhao X et al (2017) Bio-inspired P(St-MAA)-avermectin nanoparticles with high affinity for foliage to enhance folia retention. J Agric Food Chem 66(26):6578–6584. https://doi.org/10.1021/acs.jafc.7b01998

    Article  CAS  Google Scholar 

  284. Chauhan N, Dilbaghi N, Gopal M, Kumar R, Kim KH et al (2017) Development of chitosan nanocapsule for the controlled release of hexaconazole. Int J Biol Macromol 97:616–624. https://doi.org/10.1016/j.ijbiomac.2016.12.059

    Article  CAS  Google Scholar 

  285. Thombare N, Mishra S, Siddiqui MZ, Jha U, Singh D et al (2018) Design and development of guar gum-based novel, superabsorbent and moisture-retaining hydrogels for agricultural applications. Carbohyd Polym 185:169–178. https://doi.org/10.1016/j.carbpol.2018.01.018

    Article  CAS  Google Scholar 

  286. Behboudi F, Sarvestani ZT, Kassaee MZ, Sanavi SAMM, Sorooshzadeh A et al (2018) Evaluation of chitosan nanoparticles effects on yield and yield components of barley (Hordeum vulgare L.) under late season drought stress. J Water Environ Nanotechnol 3(1):22–39. https://doi.org/10.22090/jwent.2018.01.003

  287. Akalin GO, Pulat M (2019) Controlled release behavior of zinc-loaded carboxymethyl cellulose and carrageenan hydrogels and their effects on wheatgrass growth. J Polym Res 27. https://doi.org/10.1007/s10965-019-1950-y

  288. Khan MZH, Islam MR, Nahar N, Al-Mamun MR, Khan MAS et al (2020) Synthesis and characterization of nanozeolite-based composite fertilizer for sustainable release and use efficiency of nutrients. Heliyon 7(1). https://doi.org/10.1016/j.heliyon.2021.e06091

  289. Hao L, Lin G, Wang H, Wei C, Chen L et al (2020) Preparation and characterization of zein-based nanoparticles via ring-opening reaction and self-assembly as aqueous nanocarriers for pesticides. J Agric Food Chem 68(36):9624–9635. https://doi.org/10.1021/acs.jafc.0c01592

    Article  CAS  Google Scholar 

  290. da Cruz Silva G, de Oliveira Filho JG, de Mori Morselli Ribeiro M, de Souza CWO, Ferreira MD (2022) Antibacterial activity of nanoemulsions based on essential oils compounds against species of xanthomonas that cause citrus canker. Biointerface Res Appl Chem 12(2):1835–1846. 10.33263/ BRIAC122.18351846

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Najmul Arfin .

Editor information

Editors and Affiliations

Rights and permissions

Reprints and permissions

Copyright information

© 2023 The Author(s), under exclusive license to Springer Nature Singapore Pte Ltd.

About this chapter

Check for updates. Verify currency and authenticity via CrossMark

Cite this chapter

Dua, S., Ahmed, H., Arfin, N. (2023). Soft Nanomaterials and Their Applications. In: Khan, T., Jawaid, M., Ahmad, K.A., Singh, B. (eds) Nanomaterials: The Building Blocks of Modern Technology. Smart Nanomaterials Technology. Springer, Singapore. https://doi.org/10.1007/978-981-99-4149-0_3

Download citation

Publish with us

Policies and ethics

Navigation