ferritin nanocages: a novel platform for biomedical ... · of suitable non-viral carrier for...

28
See discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/264557153 Ferritin Nanocages: A Novel Platform for Biomedical Applications Article in Journal of Biomedical Nanotechnology · October 2014 DOI: 10.1166/jbn.2014.1980 CITATIONS 17 READS 496 6 authors, including: Some of the authors of this publication are also working on these related projects: Friction and durability of virgin and damaged skin with and without skin cream treatment using atomic force microscopy View project Nanomedicine View project Bharat Bhushan The Ohio State University 1,135 PUBLICATIONS 40,575 CITATIONS SEE PROFILE Uday Kumar Indian Institute of Technology Roorkee 23 PUBLICATIONS 318 CITATIONS SEE PROFILE Ishita Matai Indian Institute of Technology Roorkee 20 PUBLICATIONS 393 CITATIONS SEE PROFILE Abhay Sachdev Indian Institute of Technology Roorkee 23 PUBLICATIONS 428 CITATIONS SEE PROFILE All content following this page was uploaded by Gopinath Packirisamy on 27 January 2016. The user has requested enhancement of the downloaded file.

Upload: others

Post on 23-Oct-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

  • Seediscussions,stats,andauthorprofilesforthispublicationat:https://www.researchgate.net/publication/264557153

    FerritinNanocages:ANovelPlatformforBiomedicalApplications

    ArticleinJournalofBiomedicalNanotechnology·October2014

    DOI:10.1166/jbn.2014.1980

    CITATIONS

    17

    READS

    496

    6authors,including:

    Someoftheauthorsofthispublicationarealsoworkingontheserelatedprojects:

    Frictionanddurabilityofvirginanddamagedskinwithandwithoutskincreamtreatmentusing

    atomicforcemicroscopyViewproject

    NanomedicineViewproject

    BharatBhushan

    TheOhioStateUniversity

    1,135PUBLICATIONS40,575CITATIONS

    SEEPROFILE

    UdayKumar

    IndianInstituteofTechnologyRoorkee

    23PUBLICATIONS318CITATIONS

    SEEPROFILE

    IshitaMatai

    IndianInstituteofTechnologyRoorkee

    20PUBLICATIONS393CITATIONS

    SEEPROFILE

    AbhaySachdev

    IndianInstituteofTechnologyRoorkee

    23PUBLICATIONS428CITATIONS

    SEEPROFILE

    AllcontentfollowingthispagewasuploadedbyGopinathPackirisamyon27January2016.

    Theuserhasrequestedenhancementofthedownloadedfile.

    https://www.researchgate.net/publication/264557153_Ferritin_Nanocages_A_Novel_Platform_for_Biomedical_Applications?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_2&_esc=publicationCoverPdfhttps://www.researchgate.net/publication/264557153_Ferritin_Nanocages_A_Novel_Platform_for_Biomedical_Applications?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_3&_esc=publicationCoverPdfhttps://www.researchgate.net/project/Friction-and-durability-of-virgin-and-damaged-skin-with-and-without-skin-cream-treatment-using-atomic-force-microscopy?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_9&_esc=publicationCoverPdfhttps://www.researchgate.net/project/Nanomedicine-32?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_9&_esc=publicationCoverPdfhttps://www.researchgate.net/?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_1&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Bharat_Bhushan11?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_4&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Bharat_Bhushan11?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_5&_esc=publicationCoverPdfhttps://www.researchgate.net/institution/The_Ohio_State_University?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_6&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Bharat_Bhushan11?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_7&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Uday_Kumar72?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_4&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Uday_Kumar72?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_5&_esc=publicationCoverPdfhttps://www.researchgate.net/institution/Indian_Institute_of_Technology_Roorkee?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_6&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Uday_Kumar72?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_7&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Ishita_Matai?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_4&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Ishita_Matai?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_5&_esc=publicationCoverPdfhttps://www.researchgate.net/institution/Indian_Institute_of_Technology_Roorkee?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_6&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Ishita_Matai?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_7&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Abhay_Sachdev?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_4&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Abhay_Sachdev?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_5&_esc=publicationCoverPdfhttps://www.researchgate.net/institution/Indian_Institute_of_Technology_Roorkee?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_6&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Abhay_Sachdev?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_7&_esc=publicationCoverPdfhttps://www.researchgate.net/profile/Gopinath_Packirisamy2?enrichId=rgreq-9cc35a51e06257bdc088662ac301496a-XXX&enrichSource=Y292ZXJQYWdlOzI2NDU1NzE1MztBUzozMjI0NjM2ODUxMjAwMDJAMTQ1Mzg5Mjc0MTc1NA%3D%3D&el=1_x_10&_esc=publicationCoverPdf

  • Copyright © 2014 American Scientific PublishersAll rights reservedPrinted in the United States of America

    ReviewJournal of

    Biomedical NanotechnologyVol. 10, 2950–2976, 2014

    www.aspbs.com/jbn

    Ferritin Nanocages: A Novel Platform forBiomedical Applications

    Bharat Bhushan1, S. Uday Kumar1, Ishita Matai1, Abhay Sachdev1,Poornima Dubey1, and P. Gopinath1�2�∗1Nanobiotechnology Laboratory, Centre for Nanotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India2Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand 247667, India

    Ferritin is a ubiquitous iron storage protein responsible for maintaining the iron homeostasis in living organism and therebyprotects the cell from oxidative damage. The ferritin protein cages have been used as a reaction vessel for the synthesisof various non-native metallic nanoparticles inside its core and also used as a nanocarrier for various applications. Lackof suitable non-viral carrier for targeted delivery of anticancer drugs and imaging agents is the major problem in cancertherapy and diagnosis. The pH dependent reversible assembling and disassembling property of ferritin renders it asa suitable candidate for encapsulating a variety of anticancer drugs and imaging probes. Ferritins external surface ischemically and genetically modifiable which can serve as attachment site for tumor specific targeting peptides or moieties.Recent studies, further establishes ferritin as a multifunctional nanocarrier for targeted cancer diagnosis and therapy.Moreover, the biological origin of these protein cages makes it a biocompatible nanocarrier that stabilizes and protectsthe enclosed particles from the external environment without provoking any toxic or immunogenic responses. This reviewmainly focuses on the application of ferritin nanocages as a novel non-viral nanocarrier for cancer therapy and it alsohighlights various biomedical applications of ferritin nanocages.

    KEYWORDS: Apoferritin, Protein Cages, Nanoparticles Synthesis, Biomedical Applications, Cancer Therapy, Cancer Imaging.

    CONTENTSIntroduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2950Ferritin and Its Biological Role . . . . . . . . . . . . . . . . . . . . . . 2952Structure of Ferritin . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2953Routes of Loading . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2955Biomineralization . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2955Ferritin as a Template for Nanoparticles (NPs) Synthesis . . . . . . 2956Applications of Ferritin Protein Cages . . . . . . . . . . . . . . . . . . 2960

    Tumor Therapy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2960Tumor Imaging . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2961Tumor Targeting . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2963Cellular Uptake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2964Bioassays . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2964Biosensors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2965Biocatalyst . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2966Enzyme Immobilization . . . . . . . . . . . . . . . . . . . . . . . . . 2967Artificial Antioxidant . . . . . . . . . . . . . . . . . . . . . . . . . . . 2967Magnetic Resonance Imaging (MRI)Contrasting Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2967Biocompatibility . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2969

    ∗Author to whom correspondence should be addressed.Emails: [email protected], [email protected]: 20 January 2014Accepted: 28 March 2014

    Other Applications . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2969Conclusion and Future Perspectives . . . . . . . . . . . . . . . . . . . . 2970

    Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2970References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2970

    INTRODUCTIONNanoscale materials play a vital role during the course ofevolution of life in the form of nanosized biomoleculessuch as nucleic acids, lipids, carbohydrates and peptides.In the 20th century, nanotechnology and nanoscience hasemerged as a fascinating area of research where manynanosized structures have proven their role in the specificfield particularly in their biomedical aspects.1 Physical andchemical properties of nanoparticles such as size, shape,composition and surface chemistry determine the suitabil-ity of these particles for such applications.2�3

    A variety of nanoscale materials, such as metalbased nanoparticles,4–7 polymeric nanoparticles,8�9 mag-netic nanoparticles,10�11 fluorescent nanoparticles,12–14 andnanocomposites,15�16 has been extensively synthesized andstudied for their diagnosis and therapeutic roles. With

    2950 J. Biomed. Nanotechnol. 2014, Vol. 10, No. 10 1550-7033/2014/10/2950/027 doi:10.1166/jbn.2014.1980

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    increase in knowledge in this field, effective techniquesare emerging against dreadful human diseases, particularlycancer in which conventional methods are not efficient.17�18

    These nanostructures have comes out as blessing with dis-guise for human being as certain nanoparticles itself gen-erate toxicity and become a major concern for humanhealth.19 This provokes the researchers to search for morebiocompatible nanostructured materials for therapeutic anddiagnostic procedures.20�21

    Bharat Bhushan received his B.Sc. degree in Industrial Chemistry and M.Sc. degreein Biotechnology from Aligarh Muslim University, India. Currently he is pursuing hisPh.D. degree from Centre for Nanotechnology, Indian Institute of Technology Roorkee,India. His research work focus on development of protein based nanocarriers for variousbiomedical applications.

    S. Uday Kumar is pursuing his doctoral degree from Centre for Nanotechnology, IITRoorkee. At present he is involved in developing a multifunctional nanoscale carriersystem for lung cancer theranostics. Apart from this, his work also includes fabricationof cell specific-tissue engineering scaffold systems wherein the theranostic systems canbe evaluated. His research interest also includes tissue engineering and nanomedicine.

    Ishita Matai received her M.Tech. in Nanotechnology from IIT Roorkee, India. Sheis currently a Ph.D. student in the Centre for Nanotechnology at IIT Roorkee, India.Her current research interests include developing multifunctional nanocomposites fortargeted delivery of anticancer agents.

    Abhay Sachdev received his M.Tech. in Nanotechnology from IIT Roorkee, India.Presently he is pursuing his Ph.D. in the Centre for Nanotechnology at IIT Roorkee,India. His research work focus on development of biocompatible imaging agents forbioimaging applications.

    In this regard, biologically derived protein cage nano-structures emerge as potential nanoplatform in thedevelopment of theranostic (therapeutic and diagnostic)nanocarrier for the simultaneous delivery of anticancer andimaging agents. Protein cages get self-assembled from lim-ited number of subunits to form a spherical nanocage hav-ing an interior cavity that is utilized for the storage ofvarious therapeutic materials while exterior surface canbe functionalized with tumor specific targeting moieties.

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2951

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    Poornima Dubey received her M.Sc. degree in Biotechnology from University ofMysore, India. Currently she is a Ph.D. student in the Centre for Nanotechnology atIIT Roorkee, India. She is investigating the molecular mechanism of toxicity of variousnanoparticles and nanocomposites. Her research interests include nanotoxicology andcancer biology.

    P. Gopinath is an Assistant Professor in the Department of Biotechnology at IndianInstitute of Technology (IIT) Roorkee, India. He received his B.Sc. degree in Micro-biology and M.Sc. degree in Biotechnology from Bharathidasan University, India. Heearned his Ph.D. in Biotechnology at Indian Institute of Technology Guwahati, India.He did his postdoctoral research at University of Rochester Medical Center, New York,USA. Currently his research group in nanobiotechnology laboratory is working on thedevelopment of various protein and polymer based nanocarriers for the delivery of var-ious anticancer agents including anticancer drugs, siRNA, genes etc. This group is alsoexploring the possibilities of various biocompatible imaging agents for cancer diagno-sis. In order to realize the efficacy of such therapeutic and imaging agents, they arevalidating these systems in an artificial scaffold which mimics the in vivo condition to

    the closest extent.

    These nanoparticles overcome various limitations of con-ventional therapy such as non-specific distribution and tar-geting of drug, poor solubility of drug, poor bioavailabilityand therapeutic efficacy of drug.The most commonly used protein nanocarrier includes

    ferritin, heat shock protein (Hsp), and viral nanoparticlessuch as cowpea chlorotic mottle virus (CCMV) and cow-pea mosaic virus (CPMV), as discussed in the Table I.These protein nanocarriers have more advantages overother micrometer and sub-micrometer size delivery sys-tems, such as liposome because the protein nanocarriershave high surface area to volume ratio which increasestheir drug holding capacity, enhance the solubility of drug,increases their bioavailability by controlled release of drug,biocompatible and do not produce any toxic effect due toits biological origin.41

    Among the protein nanocarriers, viral nanoparticles arethe most extensively studied protein cages. Several draw-backs have been coupled with the use of viral deliveryvectors, which includes evoking immune response, prob-ability of integrating with the host chromosome to pro-duce a replication-competent infectious virus, rapid renalclearance from the body, difficulties in the modificationof viral capsids for tumor specific targeted delivery andhigh cost of production.42 Thus more attention is givento non-viral protein cages as they offer advantages, suchas less immunogenicity, larger drug/DNA holding capac-ity, not removed by the complement system, repeatedlyadministered without generating adverse effects, cheap,easily modifiable for targeted delivery and have negligible

    safety issues due to the non-viral nature of the deliverysystem. However, one disadvantage coupled with thesenanocarriers is low transfection efficiency. So, most ofthe recent research has been focused on the developmentof novel non-viral nanoscale delivery system by utilizingthe biologically originated protein cages having geneticallycontrolled ordered structural symmetries and modifiablesurface chemistries. Thus, by inducing genetic alterationsvarious novel functionalities, such as multiple ligands, pep-tides and small chemical entities can be anchored to thesenanocaged structures to make them competent for cancertheranostics and other biomedical applications.In last few decades, the uses of biological nanopar-

    ticles, as nanocarriers become an emerging approachfor the development of theranostic nanoparticles. Amongthese supramolecular assemblies of protein subunits, fer-ritins form a synthetic biomimetic platform for the size-constrained synthesis of nanomaterials. Thus this reviewsummarizes the role of ferritin nanocages in the nanopar-ticles synthesis and also highlights their potential biomed-ical applications.

    FERRITIN AND ITS BIOLOGICAL ROLEIn 1937, ferritin was first isolated from horse spleen43

    and later its crystal structure was elucidated in 1991.44

    The ferritin superfamily has been divided into two maingroups depending on their size namely: maxi-ferritin andmini-ferritin as described in Table II. Ferritin performstwo major functions in the body. Firstly, they act as an

    2952 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    Table I. Various types of protein cages, their structure and applications.

    Protein Interior Exteriorcages diameter diameter Structure Properties and function Applications Refs.

    HSA 8 12 Spherical shell composed of24-subunits, giving them octahedral(4:3:2) symmetry.

    Disassemble at low pHand reassemble athigh pH.

    Easily modifiable structureused in cell targetingand MRI imaging.

    [22]

    Dps 4.5 9 Spherical shell composed of 12 subunitswith 23 point group symmetry, alongwith two type of 3 fold symmetrychannel having size 0.7–0.9 nm.

    Protect cell from oxidativestress

    Template for synthesis ofvariety of NPs.

    [23, 24]

    CCMV 24 28 Capsid is composed of 180 copies of 20kDa coat protein, which assemble intoa T = 3 capsid with three positivesense RNA molecules packagedinside making a 28 nm virus and 2 nmpores exist at the quasi 3-fold axis.

    Capsids swell at pHgreater than 6.5.

    Easily modified by geneticand chemicalmodification and used inMRI imaging, celltargeting and imaging.

    [25–27]

    Mj sHsp 6.5 12 Composed of 24 subunits, which forms acage with cubic (4:3:2) symmetry andwith eight 3 nm pores located at the 3fold axes. Six smaller (1.7 nm) poresalso exist at the 4-fold axis.

    Extremely stable protein,function as molecularchaperones and overexpressed during stress.

    Easily modified, used todeliver variety ofmolecules such as MRIcontrasting agents etc.

    [28–30]

    LS 8 15 Hollow icosahedral shell with negativelycharged protein cavity, composed of60 beta subunit and 3 alpha subunit.

    Enzyme involved in thesynthesis of lumazine,a precursor of riboflavin

    Biomimetic packing ofGFP and HIV protease.

    [31–33]

    TMV 4 18 Contain ssRNA surrounded by 300nm×18 nm hollow protein tube,composed of 2130 capsomer subunitshaving both positively and negativelycharged amino acids on both surfacesthat act as the nucleation centres.

    Rod shaped and havingdistinct amino acidcomposition in interiorand exterior.

    pH dependent synthesis ofNPs. Used in synthesisof nanotubes and othernanoelectronic devices.

    [34, 35]

    P22 54 60 The mature phage form composed of415 copies of 46.6 kDa coat proteinassemble into a spherical T = 7structure with as many as 300 of 33kDa scaffold protein.10 nm pores arepresent in the P22 capsid.

    P22 naturally infectsSalmonella typhimurium

    Easily modified byattaching functionalmoieties such as biotinto encapsulate variety ofparticles.

    [36, 37]

    MS2 23 27 Self assembled structure composed of180 subunit having 32 pores ofdiameter 1.8 nm.

    Infect E.coli Easily modifiable, used todeliver variety ofmolecules, such asimaging agent for PETand MRI.

    [38–40]

    Notes: HSA-Horse spleen apoferritin; Dps-DNA-binding protein from starved cells; CCMV-Cowpea chlorotic mottle virus; Mj sHsp-Heat shock protein fromMethanococcus jannaschii; LS-Lumazine synthase from Bacillus subtilis; TMV-Tobacco mosaic virus; P22-P22-Bacteriophage; MS2–MS2 Bacteriophage.

    iron storage component and thereby maintain the availabil-ity of iron during biological synthesis of various proteins,which comprise iron as co-factor (such as heme protein,iron sulfur protein (Fe–porphyrin, Fe–S, and Fe)). Theseiron-containing proteins constitute a crucial component invarious biological processes, such as respiration, photosyn-thesis and play an important role in hydroxylation reac-tions and oxygen sensing.57�58 Secondly, ferritins play avital role in the iron metabolism and protect the cells fromoxidative damage.22�59

    STRUCTURE OF FERRITINThe primary amino acid sequences of the ferritins doesnot have any homological similarities however a clearstructural homology were found at the 2�, 3�, and 4� levels,

    indicating that the structure of ferritins remain conservedduring the evolution. The structure of ferritin is shown inFigure 1 having 24 identical subunits with octahedral sym-metry. These subunits possess a four-helix bundle alongwith a fifth E helix which is found at 60� to the four-helix bundle axis.44�61�62 Ferritin is a spherical hollow pro-tein cage with internal and external diameter of about120 Å and 75 Å, respectively.63 It can accumulate andstore approximately 4500 iron atoms.The apoferritin protein cage is composed of 80–90%

    of L-chain (light chain) and 10–20% of H-chain (heavychain) subunits with 55% sequence homology. The differ-ence between these two subunits lies in their outer surface,cavity, and hydrophobic channel sequences whereas thehydrophilic channel sequence found to be identical.64�65

    The negatively charged L chain subunit found inside the

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2953

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    Table II. Difference between maxi-ferritin and mini-ferritin.

    Characteristics Maxi-ferritin Mini-ferritin References

    Size 8–12 nm 4.5–9 nm [23]

    Structure • 24 subunits (∼ 20 kDa), four-helix bundle fold,with octahedral symmetry (432 point groupsymmetry) forming a larger spherical cavity thataccumulate 4500 Fe atoms.

    • Monomer is made up of a four-helix bundle(A, B, C and D helices) with a short fifth helix(E helix) at the C-terminus.

    • Each subunit interacts with six adjacentmonomers through three types ofsymmetry-related interfaces.

    • There are twelve dimerization interactioninterfaces at the two-fold axes, eighttrimerization interaction interfaces at thethree-fold axes and six tetramerizationinterfaces at the four-fold axes.

    • 12 subunits, four-helix bundle fold with 32(tetrahedral) point group symmetry forming asmaller cavity that accumulate 500 Fe atoms and isa structural analogue of the maxi-ferritins.

    • Monomer folds into a four-helix bundle (A, B, C andD helices), with no E helix

    • Each subunit interacts with five surroundingmonomers through two types of symmetry-relatedprotein-protein interfaces.

    • Six dimer interactions are at two-fold symmetryaxes, and four trimerization interactions arecentered at the three-fold axes. Two types ofnonequivalent three-fold interfaces exist in themini-ferritin tetrahedral dodecamer.

    [23, 44–46]

    Occurrence Bacteria, archaea, and eukaryotes Bacteria and archaea

    Examples Human ferritin, HSA, bacterioferritins Dps

    Function Store excess iron and protect from oxidativestress by removing iron and oxygen,predominantly dioxygen.

    Protecting bacteria from oxidative damage byremoving iron and oxygen, predominantly hydrogenperoxide.

    [47]

    Ferrioxidasesite/active site

    • Located in the middle of the monomericfour-helix bundle.

    • 24 active sites are saturated with 48 Fe(II).

    • Situated at the interface between two-foldaxis-related monomers.

    • 12 active sites are saturated with 12 Fe(II) atoms.Except in proteins that can use dioxygen as thesubstrate, where 24 Fe(II) bind/cage.

    [23, 48, 49]

    Stability Highly resistant to chemical denaturation, pHchanges and heat. Stable in dimer form insolution and assembly proceed from dimers totetramers and octamers.

    The protein was found to be extremely pH stable,Dps dissociated reversibly into dimers at conditionsabove pH 7.5 and below 6.0. Furthermore, dimersdissociate to monomers at pH 4.0.

    [50–53]

    Self assembly 6 amino acids at the end of the C-terminal tip ofthe D helix are essential for self-assembly.

    26 residues of the C-terminus are essential forself-assembly.

    [54]

    Iron entry The channel carboxylates in 24 subunit ferritinsselectively control Fe2+ entry.

    The channel carboxylate groups control both Fe2+entry and Fe2+ exit.

    [55, 56]

    inner cavity of assembled protein cage has clusters ofacidic residues (Glu and Asp) which form the mineralnucleation site. This site mainly performs the function ofdelivery of iron and help in the nucleation of ferrihydritecore.61�66 So, these chains were found in the extra cellularferritin as they act as an iron carrier for different cells.67

    The heavy chain which catalyzed the oxidation of Fe+2 is

    Figure 1. (a)–(c) Ribbon diagrams of L-ferritin taken fromPDB entry 1DAT: (a) the 24-subunit assembled cage; (b) theinner cavity; (c) the 3-fold axis channel. Reprinted with per-mission from [60], S. Abe, et al., Polymerization of pheny-lacetylene by rhodium complexes within a discrete space ofapo-ferritin. J. Am. Chem. Soc. 131, 6958 (2009). © 2009,American Chemical Society.

    responsible for the iron mineralization and the formationof iron crystal.65 The nucleation site of H chain subunitfound in close proximity with the ferroxidase site shar-ing one glutamate residue between them.61 Recent studieson different ferritin further strengthened that iron storingcapability of ferritin is related to the number of L sub-units. Moreover, presence of small number of H subunitin ferritin obtained from iron storage organ reveal theimportance of oxidative process in iron storage.68 The fer-roxidase activity of the apoferritin gets affected in thepresence of metal nanoparticles as it has been found toget increased in the presence of platinum, gold and silvernanoparticles.69�70

    There are 14 channels having a diameter of 0.3–0.4 nmeach, which are present at the junction of these sub-units. Out of these 14 channels, eight channels arehydrophilic in nature and posses four-fold symmetry, whilethe remaining six are hydrophobic and possess three-foldsymmetry.71 The aperture size of these hydrophilic chan-nels are adjusted according to the particles as demonstratedthat in presence of urea, these eight hydrophilic channels

    2954 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    attain sufficient flexibility and allow larger size moleculesto penetrate inside the apoferritin cavity.72

    The molecular species enter into the protein cavitythrough these channels by charge selective process. More-over, flow of ion through the pore is regulated by thelocal folding and unfolding of the ion channel pore.The four highly conserved residues, such as arginine 72,aspartate 122, leucine110 and leucine 134 are respon-sible for the stability of pore and form the pore gate.These pores are less stable compared to the overall sta-bility of ferritin nanocages, even at low temperature andlow concentration of denaturants, such as urea and guani-dine, pores show instability. It has been suggested thatbiological regulators are present in vivo, which recog-nize the pore gates and hold it in either open or closeconformation to maintain the iron homeostasis.73 Ferritincage without the ferrihydrite mineral core is called asapoferritin.

    ROUTES OF LOADINGThere are two major ways of loading materials inside apo-ferritin as shown in Scheme 1: First, by directly incubatingthe materials with the apoferritin in which the smaller par-ticles comparable to the size of channels move directly andget accumulated inside the inner cavity. This process mim-ics the natural biomineralization process. Second way isapplicable for the larger particles which cannot efficientlypass through the channels. In this route, the apoferritinprotein cage undergoes pH dependent assembly at higherpH and disassembly at lower pH.

    BIOMINERALIZATIONFerritin protein cages have been used as nanosized con-tainers for the controlled synthesis of a variety of nanopar-ticles by biomimetic process. So, in order to synthesizenanoparticles inside its cavity it becomes important tounderstand, how the process of biomineralization of ironnaturally occurs in ferritin (Scheme 2).Iron biomineralization in ferritin is a multistep process

    that includes:1. Entry and binding of iron ions inside the ferritin cagecavity.

    Scheme 1. Schematic representation of different routes ofloading in ferritin nanocage.

    Scheme 2. Schematic representation of naturally occurringbiomineralization process inside ferritin nanocage.

    2. Oxidation of iron ions followed by nucleation andgrowth of ferrihydrite core.3. Release of iron ions from ferritin.

    Step 1. Entry and binding of iron ions inside the ferritincage cavity. The iron enters into the ferritin through the15 Å long channels which are gated by extensions of thefour-helix bundle subunits. These metal ions were guidedinside the cavity by charged gradient of the channel cre-ated by the presence of conserved carboxylate residues:Glu130, Asp127, Ala26, Val42, Thr149.47�56 Moreoverthere are two basic types of functional channels present inferritin:

    (1) Iron ion entry channels formed by three subunitsaround the 3-fold cage axes which allows the passage ofFe2+ substrate to oxidoreductase sites (Asp127, Glu130).(2) Iron ion nucleation channels, which are present at

    the other side of the 4-helix bundles subunit around the4-fold cage axes (Ala26, Val42, Thr 149).

    The Fe(II) ions reacts with O2 after binding to the activesite and produce diferric oxo products in eukaryotes. Thediferric peroxo intermediate (DFP) is first detectable inter-mediate which forms and decays in seconds or less intothe di-Fe(III)O product, a mineral precursor which is laterreleased into protein nucleation channels.48

    Step 2. Oxidation of iron ions followed by nucleation andgrowth of ferrihydrite core. Two Fe2+ ions get oxidizedto Fe3+ in the presence of oxygen after binding to theferroxidase center. The Fe3+ ions then migrate to the fer-ritin inner cavity and finally a mineral core formation takeplaces at the nucleation sites of the L-chain ferritin. Thisis the initial process when no iron is present in ferritin,but as soon as the iron mineral core is formed, the irongets oxidized directly on the mineral core surface after

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2955

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    passing through the 3-fold channels.74 This oxidation pro-cess on mineral core is found to be more rapid than thatat the ferroxidase center, which remains functional aftera core is formed and with no significant contribution inFe2+ oxidation.75

    It has been shown by various in vitro studies using vari-ous mutant ferritin cage, which lack nucleation site or fer-roxidase activity or both, directly affect the encapsulationor mineralization process or both.76 This indicates the roleof nucleation site in aggregating the ions at the highly nega-tively charged protein interface and in facilitating oxidativemineralization and ferroxidase centre in converting the sol-uble Fe2+ to insoluble Fe3+, absence of which leads to theuncontrolled growth and precipitation.76�77 Thus suggeststhat the ferritin biomineralization is highly specific for iron.Step 3. Release of iron from ferritin. In vitro removal ofiron from ferritin is a two-step process, which includesreduction of Fe3+ mineral followed by the chelation ofFe2+ from the mineral core. Four iron release reductionand chelation model namely subunit displacement, diffu-sion of molecules through the 3-fold channels, gated poresand electron transfer through the protein shell, has beenbriefly discussed in a review by Watt et al.78 Consideringall the possible ways of iron release mechanism and theirtransportation through the protein cage, the 3-fold chan-nels are currently accepted route for the passage of iron toenter and exit the protein cage.78 Moreover, the redox reac-tions occur during the iron mineralization and release areaccompanied by the simultaneous release of ion in order tobalance the charge on both sides of the cage. For example,the entries of electron during the reduction of iron in themineral core are accompanied by concomitant release ofnegative charge from the core. Some of the important ionsinvolved during this process are chloride and hydroxideions, moving throughout the protein cage and phosphateion release during the reduction process.78

    Iron releasing occurs on exposure of ferritin to UV lightor ionizing radiation, the iron mineral core acts as photore-ceptor and result in the reduction of Fe (III). In the absenceof oxygen, redox reaction results in the iron mobilizationfrom ferritin catalyzed by the hydrated electron, which actsas a reducing agent. In the presence of oxygen superox-ide radical anion (O•−2 ) is responsible for the iron releaseprocess. This suggests the requirement of an iron chela-tor for Fe(II) mobilization from ferritin, in the absence ofwhich ferritin act as a electron-storage molecule.79 More-over, the reversal of process of biomineralization is veryslow, as shown in the in vitro study by removing the excessiron in sickle cell disease and thalassemia with the help ofchelators.80�81

    FERRITIN AS A TEMPLATE FORNANOPARTICLES (NPs) SYNTHESISMetal nanoparticles can be fabricated inside the apoferritincavity, which act as a reaction vessel. Protein cage like

    structure of apoferritin can be used for the size dependentencapsulation of various materials by serving as templateto restrain the NPs growth and prevent aggregation. Theseself assembled protein shell form a reaction chamber forthe synthesis of non-native materials of controlled dimen-sions, while exterior surface can be easily modified withvarious functional moieties through genetic and chemicalmodification. A variety of different precursor ions havebeen formed by nucleation and subsequent mineral growthsuggesting that other non-native metals could also be min-eralized within the ferritin core. Due to the sharper densityof these NPs as compared to higher-dimensional struc-tures, these NPs offer superior quality that can be used inbiosensors, nanoelectronic devices, bioimaging and vari-ous other biomedical applications.Artificial synthesis of ferromagnetic iron oxide nanopar-

    ticles inside the apoferritin cavity has been reported byMann and co workers.82�83 They mimicked the naturalbiomineralization process and opened the way to utilizeapoferritin for the synthesis of various inorganic nanoparti-cles. Similarly, mini-ferritin (Dps) used for the synthesis ofNPs includes Co(O)OH and Co3O4 (dia 4�34±0�55 nm),84�-Fe2O3,

    85�86 CdS87 and Pt.88

    After loading, these NPs were undergoing various inter-mediate stages before leading to the final mineralizednanoparticles. Metal ions were reduced inside the cavity byusing a reducing agent for example H2, NaBH4, or light,then the encapsulated NPs were separated from the unen-capsulated ones by the implications of additional purifi-cation steps.89 Moreover the ferrihydrite core undergoesin situ reactions and gets modified to other iron productssuch as FeO,90 iron sulfide91 and semiconducting hematite(�-Fe2O3).

    92 Similarly, high temperature synthesis wascarried out using ferritin from thermophilic archaeon Pyro-cocus furious, which retains its cage-like structure evenat 120 �C.93 This can be further used as a template forsynthesis of magnetite94 and other metallic NPs such asgold, silver, lead, copper, nickel and semiconductors NPssuch as CdS. The outer surface of ferritin modified withPEG prevents the bulk precipitation and improves the yieldof NPs in ferritin cavity.95 The noble metals ions (Au3+,Ag+) bind to the exterior surface of the protein. In orderto facilitate internalization of these metal ions, reactivecysteine and histidine residues are removed from the exte-rior surface of ferritin and soft cysteine ligands are intro-duced in the interior surface.96 The metals ions bind tothe specific binding site present on the protein shell, bothinterior and exterior surfaces of the protein cage that pro-mote the growth of NPs both inside and outside the cage.Moreover, modification on the surface of ferritin leads tochange in their properties. For instance, the alkylation ofthe ferritin protein using a monoamine-terminated alkaneoligomer (dodecylamine) changes the charge of the proteinand type of interactions by converting the primary car-boxylic acid groups on the ferritin surface into hydropho-bic groups.97 Recently, reported recombinant apoferritin

    2956 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    Table

    III.

    Listofva

    riousmaterials

    synthes

    ized

    usingferritin

    cages

    andtheirap

    plic

    ations.

    Type

    ofType

    sof

    Load

    ing

    Prope

    rtiesof

    ferritin

    material

    Sou

    rceof

    material

    effic

    ienc

    ypa

    rticles

    App

    lications

    Referen

    ces

    HSA,RA,CA,

    PfFt

    Pdan

    dits

    orga

    nometallic

    complexes

    K2PdC

    l 4,[PdII(allyl)C

    l]296

    –500

    Catalytic

    and

    non-mag

    netic

    .•Use

    din

    high

    lysp

    ecificae

    robicox

    idationof

    alco

    hols

    inwater

    •Use

    din

    size

    -selec

    tiveolefi

    nhy

    drog

    enation.

    [99–

    102]

    CF,

    CA,HSA

    Cuan

    dits

    radioa

    ctive

    isotop

    e

    64CuC

    l 2,CuS

    O4·5H

    2O

    225–

    2000

    Catalytic.

    •Tu

    mor

    spec

    ifictargeting.

    •Can

    beus

    edin

    PETim

    agingan

    dna

    noelec

    tron

    icde

    vice

    s.•Ferritin

    actin

    gas

    photoc

    atalys

    tin

    pres

    ence

    ofvisiblelig

    htredu

    cing

    copp

    er.

    •Electrontran

    sportatio

    nstud

    ies.

    [103

    –106

    ]

    HSA

    CuS

    NS

    NS

    Sem

    icon

    ductor

    •Can

    beutilize

    das

    aco

    mpo

    nentsforna

    no-electric

    device

    s,su

    chas

    solarba

    tterie

    san

    dliq

    uidcrys

    tals

    [107

    ]

    HSA

    CuF

    e[Fe(CN) 6]3

    −22

    5Mag

    netic

    •Bas

    icload

    ingstud

    ies.

    [108

    ]

    HSA,CA,

    reco

    mbina

    ntFtLi,AvB

    F

    Coan

    dits

    oxides

    /ox

    yhyd

    roxide

    Co

    2+,CoS

    O4,

    Co(NO

    3) 2·6H

    2O,

    Co(OAc)

    2·4H

    2O

    200–

    2000

    Mag

    netic

    andca

    talytic

    •Pos

    siblyutiliza

    blein

    nano

    elec

    tron

    ics.

    •Electroca

    talyst

    inelec

    troc

    hemical

    reac

    tionforthe

    detectionof

    gluc

    ose.

    •Nov

    elbios

    enso

    rca

    nbe

    used

    inmed

    ical

    and

    indu

    stria

    lfields

    tode

    tect

    diffe

    rent

    analytes

    •Electrontran

    sportatio

    nstud

    ies.

    [84,

    105,

    106,

    109–

    115]

    RA,CA

    CoP

    t(N

    H4) 2PtC

    l 4,

    (CH

    3COO) 2Co·4H

    2O

    1000

    Mag

    netic

    •Can

    beutilize

    din

    theprep

    arationof

    high

    -den

    sity

    reco

    rdingmed

    ia.

    [116

    ,11

    7]

    HSA

    CoN

    iCoS

    O4,NiSO

    4NS

    Mag

    netic

    prop

    ertie

    s•Can

    beus

    edin

    MRI

    [118

    ]

    RA,HSA

    Nia

    ndits

    hydrox

    ide

    Ni2+

    8000

    Mag

    netic

    .•Pos

    siblyus

    able

    inna

    noelec

    tron

    ics.

    •Prepa

    ratio

    nof

    zero

    valent

    NPs.

    [105

    ,10

    9,11

    9]

    Rec

    ombina

    ntPfFt,HuH

    Ft,

    FtLi,RA,

    HSA,HuH

    Ft,

    CA,AfFtn

    Iron

    andits

    compo

    unds

    (oxide

    ,su

    lphide

    and

    radioa

    ctiveisotop

    e)

    FeC

    l 2,FeS

    O4,

    (NH

    4)Fe(SO

    4� 2

    100–

    7200

    Mag

    netic,ca

    talytic,an

    dpo

    tentialM

    RI

    contrast

    agen

    t

    •Use

    din

    tumor

    spec

    ifictargetingan

    dim

    aging

    •Fa

    cilitatece

    llularup

    take

    •Act

    asph

    otoca

    talyst

    inpres

    ence

    ofUV/Visible.

    •Use

    din

    prod

    uctio

    nof

    SWCNTan

    dco

    ntrolle

    dits

    size

    •Use

    din

    fluores

    cent

    andMR

    imag

    ing(in

    vivo

    /invitro).

    •Use

    din

    ironab

    sorptio

    nan

    delec

    tron

    tran

    sfer

    stud

    ies.

    •Can

    beutilize

    din

    nano

    device

    .

    [82,

    83,85

    ,91

    ,10

    4,10

    6,11

    4,12

    0–13

    8],

    CA

    FeC

    o(N

    H4) 2Fe(SO

    4)·6

    H2O,

    (Co(NO

    3) 2·6H

    2O

    1000

    Mag

    netic

    .•Mag

    netis

    mfoun

    dwith

    ferritin.

    [139

    ]

    HSA

    Iron

    arse

    nate,

    phos

    phate,

    vana

    date,molyb

    date

    particles.

    NS

    2000

    Catalytic.

    •Iron

    load

    ingstud

    ies

    [140

    ]

    PfFt

    FeP

    t(N

    H4) 2Fe(SO

    4) 2,

    K2PtC

    l 4

    500

    Mag

    netic

    •Bas

    icbiom

    imetic

    synthe

    sis.

    [141

    ]

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2957

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    Table

    III.

    Continued

    .

    Type

    ofType

    sof

    Load

    ing

    Prope

    rtiesof

    ferritin

    material

    Sou

    rceof

    material

    effic

    ienc

    ypa

    rticles

    App

    lications

    Referen

    ces

    RA

    Inox

    ide

    In2(S

    O4� 3

    NS

    Sem

    icon

    ductor

    •Pos

    siblyutiliza

    blein

    nano

    elec

    tron

    ics

    [136

    ]

    HSA

    Des

    ferrioxa

    mineB

    Des

    ferrioxa

    mineB

    3Catalytic

    •Poten

    tialtobe

    utilize

    din

    ironch

    elationtherap

    y.[142

    ]

    NS

    AuP

    tK

    2PtC

    l 4,HAuC

    l 4NS

    Antioxida

    nt•To

    stud

    ytherece

    ptor-m

    ediatedce

    llularup

    take

    ofNPs.

    [143

    ]

    RA,HSA,

    HuH

    Ft,

    Auan

    dits

    compo

    unds

    AuC

    l 3,HAuC

    l 4,KAuC

    l 425

    0–40

    00/Au,

    clus

    ter

    16–5

    0,/

    Au 2S-300

    0

    Catalytic,

    photolum

    ines

    cenc

    e,se

    micon

    ducting

    particles

    •In

    vivo

    kidn

    eytargetingan

    dbiom

    edical

    Imag

    ing

    •Produ

    ctionof

    SWCNTan

    dgo

    ldna

    nosh

    ell.

    •Enh

    ance

    catalytic

    activ

    ity.

    •Pos

    siblyutiliza

    blein

    nano

    elec

    tron

    ics

    [69,

    70,96

    ,98

    ,14

    4–15

    0],

    RA

    AuP

    dKAuC

    l 4,K

    2PdC

    l 428

    9–44

    1Catalytic.

    •Enh

    ance

    catalytic

    activ

    itydu

    ringolefi

    nhy

    drog

    enation

    [151

    ]

    HSA

    Au–

    AgNPsalloy

    AgN

    O3,HAuC

    l 4NS

    Catalytic

    •Catalyzed

    theredu

    ctionof

    4-nitrop

    heno

    linthe

    pres

    ence

    ofNaB

    H4.

    [152

    ]

    Apo

    PPF

    Au-5F

    UNH

    4AuC

    l 4,5F

    U1/45

    Antican

    cerdrug

    •Can

    beus

    edin

    chem

    othe

    rapy

    ofca

    ncerou

    sce

    lls[153

    ]

    HSA

    Pho

    spha

    tepa

    rticles

    from

    Cd,

    Zn,

    Pb,

    Cu

    Cdc

    l 2,Zn(NO

    3) 2,

    Pb(NO

    3) 2,Cu(NO

    3) 2,

    NS

    Red

    oxmarke

    rs•Partic

    lesus

    edas

    labe

    lsin

    elec

    troc

    hemical

    immun

    oass

    ayan

    dus

    edforde

    tectionof

    individu

    alsing

    lenu

    cleo

    tidepo

    lymorph

    isms(S

    NPs)

    [154

    –156

    ]

    CA,HuH

    Ft

    HuL

    Ft,PfFt,

    RA,HSA

    Ag

    AgN

    O3

    250–

    5000

    Antim

    icrobial

    activ

    ityan

    dca

    talytic

    activ

    ity.

    •Antibac

    teria

    laga

    inst

    S.aureu

    s.•Increa

    sein

    ferrox

    idas

    eac

    tivity

    offerritin.

    [69,

    70,96

    ,14

    8,15

    7–15

    9],

    NS

    pHindica

    tormolec

    ules

    NS

    NS

    pHindica

    tor

    •Use

    dforstud

    ying

    ferritin.

    [160

    ]

    CA,HSA

    Gdan

    dits

    complexes

    Gd-Me 2DO2A

    /Gd-

    DOTA

    ,GdH

    PDO3A

    ,Gd(NO

    3) 3·6H

    2O,

    LnCl 3.

    Com

    plexe8

    -36

    /oxide

    -17

    00

    Mag

    netic

    particlesan

    dMRIco

    ntrast

    agen

    t•Use

    din

    MRIex

    perim

    ents

    (invitroan

    din

    vivo

    ).•Visua

    lizationof

    tumor

    angiog

    enes

    isby

    mag

    netic

    reso

    nanc

    e

    [161

    –166

    ]

    HSA

    Curcu

    min

    C21H

    20O

    69�5±2

    The

    rape

    utic

    agen

    t•Use

    dforev

    alua

    tingdrug

    deliveryeffic

    ienc

    yin

    mice

    [161

    ]

    CA,HSA

    Pban

    dits

    compo

    unds

    (pho

    spha

    te,su

    lhide)

    Pb(NO

    3) 2,Pb(AcO

    ) 213

    00Signa

    lamplifica

    tion

    andqu

    antum

    dot

    •Electroch

    emical

    immun

    oass

    ayforqu

    antifi

    catio

    nof

    phos

    phorylated

    acetylch

    olines

    terase

    .•Can

    beus

    edas

    bioc

    ompa

    tible

    agen

    tin

    bioimag

    ing.

    •Act

    asan

    tican

    cerag

    entan

    dindu

    ceap

    optosis.

    [167

    –171

    ]

    HSA

    YPO

    4Y

    (NO

    3) 3

    500

    Rad

    ionu

    clideNPs

    •Poten

    tialtobe

    utilize

    din

    radioimmun

    othe

    rapy

    ofca

    ncer

    [172

    ]

    FtLi,HSA,RA

    Cdan

    dits

    compo

    unds

    (sulph

    ide,

    selenide

    ,ph

    osph

    ate)

    CdC

    l 2,Cd(CH

    3CO

    2) 2

    55–1

    350

    Sem

    icon

    ductor

    quan

    tum

    dots

    and

    marke

    rmolec

    ule

    •Pos

    siblyutiliza

    blein

    nano

    elec

    tron

    ics.

    •Use

    das

    fluores

    cent

    biom

    arke

    rin

    bioa

    ssay

    s.•Use

    din

    sequ

    ence

    -spe

    cific

    DNA

    detection.

    [24,

    87,

    173–

    177]

    HSA

    Methy

    lene

    blue

    .C

    16H

    18N

    3SCl

    NS

    Pho

    tose

    nsitize

    r•Cou

    ldbe

    useful

    inph

    otod

    ynam

    ictherap

    yof

    canc

    er.

    [178

    ,17

    9]

    HSA

    Fluores

    cein

    C20H

    10Na 2O

    5NS

    Fluores

    cent

    particles.

    •Partic

    lesus

    edas

    labe

    lsin

    bioa

    ssay.

    [180

    ]

    HSA

    Hex

    acya

    noferrate

    K3Fe(CN) 6

    65–1

    50Marke

    rmolec

    ule.

    •Partic

    lesus

    edas

    labe

    lsin

    elec

    troc

    hemical

    immun

    oass

    ay.

    [180

    ,18

    1]

    2958 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    Table

    III.

    Continued

    .

    Type

    ofType

    sof

    Load

    ing

    Prope

    rtiesof

    ferritin

    material

    Sou

    rceof

    material

    effic

    ienc

    ypa

    rticles

    App

    lications

    Referen

    ces

    Apo

    PPF,

    CA,

    HSA

    Cisplatin,ca

    rbop

    latin

    andox

    aliplatin

    PtC

    l 2(N

    H3) 2,

    500

    Antican

    cerdrug

    .•Deliver

    drug

    andindu

    ceap

    optosis

    •Sho

    wsincrea

    sedce

    llularup

    take

    [182

    –184

    ]

    HAS,RA

    Znan

    dits

    compo

    unds

    (selen

    ide,

    oxide)

    (Zn(NO

    3) 2·6H

    2O)

    1500

    Sem

    icon

    ductor

    •Cou

    ldbe

    used

    inna

    noelec

    tron

    ics

    [185

    ,18

    6]

    HSA,RA,AfFtn

    Mnan

    dits

    oxide,

    oxyh

    ydroxide

    MnC

    l 212

    00–4

    000

    Mag

    netic

    •MRIse

    nsor

    formelan

    inform

    ationin

    meloa

    nomace

    ll•Use

    das

    MRIco

    ntrast

    agen

    t.[106

    ,11

    3,11

    5,13

    3,18

    7–18

    9]

    HSA,CA

    Pt

    K2PtC

    l 4,K

    2PtC

    l 6,

    (NH

    4) 2PtC

    l 4

    250–

    4000

    Catalytic

    activ

    ity.

    •Increa

    seferrox

    idas

    eac

    tivity

    offerritin.

    •Use

    das

    artifi

    cial

    antio

    xida

    nt.

    •Morebioc

    ompa

    tible

    andinternalizevia

    rece

    ptor-m

    ediateden

    docy

    tosis

    [70,

    190–

    192]

    RA

    Cerium

    oxide(C

    eO2,

    Ce 2O

    3)

    CeC

    l 3·7H

    2O

    400

    Catalys

    t•Activeartifi

    cial

    redo

    xen

    zymewith

    mim

    etic

    SOD

    activ

    ity[193

    ,19

    4]

    CA

    Eva

    nblue

    ordirect

    yellow

    dye

    NS

    NS

    Dye

    •Use

    dto

    prep

    arefree

    stan

    ding

    mes

    oporou

    sprotein

    thin

    filmsan

    dstud

    yof

    theirpH

    depe

    nden

    treleas

    e[195

    ]

    CA

    Dau

    nomyc

    inC

    27H

    29NO

    10·H

    Cl

    NS

    Antican

    cerdrug

    •Binding

    stud

    ieswith

    DNA

    [196

    ]

    CA,RA

    Dox

    orub

    icin

    C27H

    29NO

    11·H

    Cl

    5–50

    Antican

    cerdrug

    •Prelim

    inarycy

    totoxicity

    hasbe

    ende

    mon

    strated

    [195

    –199

    ]

    CA

    Gluco

    seox

    idas

    e(G

    Ox)

    GOx

    7.8

    Catalytic

    activ

    ity•Catalyzed

    oxidationof

    D-gluco

    seto

    D-gluco

    no-1,5-la

    cton

    ean

    dhy

    drog

    enpe

    roxide

    .[200

    ]

    HSA

    PEI

    NS

    NS

    Coreac

    tant

    •Biose

    nsingan

    dbioa

    ssay

    applications

    [201

    ]

    RA,CA

    Euox

    idean

    dits

    complexes

    EuC

    l 314

    –100

    0La

    belm

    oiety

    •Bioaffin

    ityas

    sayan

    dbio-im

    aging

    [134

    ,20

    2,20

    3]

    CA

    Tio

    xide

    Ti(IV)

    1000

    NS

    •Bas

    icload

    ingstud

    ies.

    [134

    ]

    RA

    RhCom

    plexes

    [Rh(nb

    d)Cl] 2

    58Catalytic

    •Catalyzethepo

    lymerizationof

    phen

    ylac

    etylen

    e.[60]

    RA

    Ruco

    mplexes

    [Ru(pcy

    men

    e)Cl 2] 2

    88±1

    1Catalys

    ts•Bas

    icload

    ingstud

    ies.

    [204

    ]

    HSA

    LuPO

    4Lu

    Cl 3

    NS

    NS

    •Cou

    ldbe

    used

    inca

    ncer

    therap

    y.[205

    ]

    RA

    Caan

    dits

    compo

    und

    (carbo

    nate)

    Ca(HCO

    3) 2,CaC

    O3

    NS

    Che

    mically

    stab

    le•New

    mec

    hanism

    ofmineralizationus

    ingco

    ntrolle

    delec

    tros

    tatic

    potential

    [206

    ]

    HSA

    CaC

    O3,SrC

    O3,BaC

    O3

    andCa 3(P

    O4) 2

    CaC

    l 2,SrC

    l 2or

    BaC

    l 214

    60–1

    600

    NS

    •Bas

    icload

    ingstud

    ies.

    [207

    ]

    RA,HSA

    Crhy

    drox

    ide

    Cr3

    +48

    00NS

    •Pos

    siblyus

    able

    inna

    noelec

    tron

    ics.

    [119

    ]

    CA,HSA

    Uan

    dits

    oxide/ox

    yhyd

    roxide

    UO

    2+80

    0–40

    00Rad

    ioac

    tivepa

    rticles

    •Can

    beus

    edin

    uran

    ium

    neutron-ca

    pturetherap

    y[133

    ,20

    8]

    HSA

    Pruss

    ianblue

    K4Fe(CN) 6

    NS

    Catalytic

    •Enz

    ymemim

    etic

    activ

    ityca

    nbe

    utilize

    dfor

    biolog

    ical

    detection.

    [209

    ]

    Notes

    :NS—Not

    spec

    ify;HSA—Horse

    spleen

    ferritin;

    Apo

    PPF-Apo

    -pig

    panc

    reas

    ferritin;

    CA—Com

    mercial

    apoferritin;RA—Rec

    ombina

    ntap

    oferritin;CF—Chimeric

    ferritin

    (mixture

    of2

    heav

    ych

    ain

    ferritins

    );HuH

    Ftan

    dHuL

    Ft—

    Hea

    vyan

    dlight

    human

    ferritinch

    ains

    ;PfFt—

    Ferritin

    from

    hype

    rthe

    mop

    hilic

    bacterium

    Pyroc

    occu

    sfurio

    sus;

    AfFtn—Mutan

    tArcha

    eoglob

    usfulgidus

    ferritin;

    FtLi—

    Ferritin

    from

    Listeria

    inno

    cua;

    AaL

    S—Lu

    maz

    inesyntha

    sefrom

    Aqu

    ifexae

    olicus

    ;AvB

    F—Azo

    toba

    cter

    Vinelan

    diib

    acterio

    ferritin.

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2959

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    having gold-binding peptide and titanium-binding peptideat the C- and N-terminus, respectively that specificallycatch gold NPs and deliver them to the silicon dioxide sur-face under specific conditions.98 List of various moietiesencapsulated inside the ferritin protein cage is discussedin Table III.

    APPLICATIONS OF FERRITINPROTEIN CAGESFerritin nanocages have been widely used in various bio-logical and biomedical applications as discussed below(Scheme 3).

    Tumor TherapyApoferritin can encapsulate a variety of therapeutic agents,which can be utilized in different strategies for tumor treat-ment. Some of the strategies include:

    Neutron Capture TherapyNeutron capture therapy is a promising methodology forthe treatment of cancer. Boron and uranium are the basicelements used in this technique. They are localized to thetargeted tumor cell and irradiated with slow neutron, whichleads to the disintegration of nuclei into smaller fragments

    Scheme 3. Schematic representation of various applications of ferritin nanocages.

    along with ionizing particles that kill the cell. In early90’s, Hainfeld first described a method to deliver 235U byencapsulating it in apoferritin cage which minimize theimmune response and heavy metal toxicity. Antibody Fab-fragments were chemically coupled to the protein cage fortumor specific targeting. The isotope was then fissioned byneutron beam that produced the required localized lethalradiation for the tumor therapy.208

    Radioimmunoimaging and RadioimmunotherapyLutetium-177 is a radionuclide having a physical half-life of 6.7 days and other radiological properties suchas emission of low energy beta particles and gammaradiation have been utilized for targeting small tumorsfor radioimmunoimaging and radioimmunotherapy ofcancer.210 A radionuclide nanoparticles (NPs) have beensynthesized by conjugating apoferritin with lutetiumphosphate (LuPO4) or yttrium phosphate (YPO4) andfunctionalized them with biotin. The pretargeting capa-bilities of these nanoparticle conjugates were studiedusing biotin-modified LuPO4 or YPO4-apoferritin withstreptavidin-modified magnetic beads and in addition withthe aid of streptavidin-modified fluorescein isothiocyanate(FITC) tracer. This method can be further exploitedfor the preparation of radioactive LuPO4 or YPO4

    2960 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    conjugates that can be utilized in radioimmunotherapy ofcancer.172�205

    Photodynamic TherapyPhotodynamic therapy (PDT) has emerged as an importanttool in the field of tumor therapy and has been utilizedfor the treatment of both oncological (e.g., tumors anddysplasias) and non-oncological (e.g., age-related maculardegeneration, localized infection and non-malignant skinconditions) applications.211 In this strategy, photosensitiz-ing agents, light, and oxygen take part in photochemi-cal reaction. A photosensitizing agent, methylene blue hasbeen successfully encapsulated inside apoferritin cage, thatcan be internalized by the tumor cells and on irradiationwith a light of suitable wavelength (i.e., 633 nm) generatesa cytotoxic agent, a singlet oxygen intracellularly for pho-todynamic therapy (PDT) which induced cytotoxic effectson the human breast adenocarcinoma cells (MCF-7).178�179

    Recently, a RGD4C-modified ferritin encapsulated withZinc hexadecafluorophthalocyanine (ZnF16Pc), an effec-tive photosensitizer showed a high tumor accumulationrate, less toxicity toward major organs and effective tumorinhibition on light irradiation by inducing phototoxicity inU87MG subcutaneous tumor models.212

    Anticancer Drug CarrierToxicity and drug resistance of platinum based anticancerdrug limited their use for cancer therapy. Apoferritin canbe exploited as a drug delivery system to these plat-inum based drugs (cisplatin, carboplatin and oxaliplatin)to overcome these drawbacks and to enhance the cellu-lar uptake of anticancer drugs.182 Cisplatin and carboplatinloaded apoferritin showed a primary toxicity against ratpheochromocytoma PC12 cells.183 Recently, a novel nano-sized construct of cisplatin core-apo pig pancreas ferritin(NCC-PPF) has been developed and its anticancer activityon gastric cancer cells BGC823 (GCC) were studied.184

    Daunomycin (anticancer drug) used for the treatment ofacute myeloid leukemia and lymphocytic leukemia havebeen successfully encapsulated into apoferritin which ismodified by incorporating a negatively charged polypep-tide poly-L aspartic acid (PLAA) to improve their drugholding capacity.196 Similarly, apoferritin has been utilizedfor the encapsulation of anticancer drug doxorubicin.197�198

    A simple and easy method for preparation of thin meso-porous protein films has been developed for efficient load-ing and releasing of dye or doxorubicin by controlling thepH. It loaded at lower pH and released the drug at higherpH than the isoelectric point of protein.195

    The differential effect of near-infrared apoferritin-PbS(AFt-PbS) nanocomposites on cell cycle progression innormal and cancerous human cells has been recentlyreported. The nanocomposite did not alter the cell cycle innormal cell at concentration up to 1 mg mL−1 whereas inhuman breast cancer cell line it triggered apoptosis at con-centration > 0.2 mg mL−1. These nanocomposites entered

    the cell through endocytosis and further could be used forthe in vivo imaging studies.170 Another anticancer com-pound Ru complex has also been successfully immobilizedto the ferritin cages by His residue present on the ferritinsurface.204

    A bio-inspired nanoconstruct have recently been devel-oped using an apoferritin-gold nanoconstruct loadedwith anticancer drug 5-fluorouracil (5-FU) that exhib-ited a high selectivity towards cancerous cells and alsoincreased the cellular uptake of 5-FU via receptor-mediated endocytosis.153 Moreover, a genetically modifiedferritin (RFRTs) nanocages having a tumor targeting pep-tide Cys-Asp-Cys-Arg-Gly-Asp-Cys-Phe-Cys (RGD4C)attached on its surface has been used for the deliveryof anticancer drug doxorubicin as shown in Figure 2.Such nanoconstruct showed high drug loading efficiencyin presence of Cu(II) as a helper agent. This nanoconstructhave improved tumor suppression ability and reduced car-diotoxicity, when studied on U87MG subcutaneous tumormodels.199

    Other Therapeutic ApplicationsIron is one of the essential elements for all the livingbeings, but if present in excess becomes toxic. Humanbody is incapable to remove this excess iron, which leadsto their accumulation in the liver and other organs lead-ing to serious health complications and eventually death.213

    To remove this excess of iron, Desferrioxamine B (DFO)drug produced by Streptomyces pylosus is used for ironchelation therapy by encapsulating it inside the apoferritincage, which upon further reaction with Fe III gives rise toencapsulated [DFOFe] complex within the apoferritin.142

    This nanocontainer can also be utilized for the treatmentof other infectious disease. As in a newly developed strat-egy in which silver (I) ions were loaded into apoferritinto function as an antimicrobial agent.157 Recently, a noveltheranostic agent has been constructed utilizing the apofer-ritin cage to simultaneously deliver the therapeutic agent(curcumin) and imaging agent (GdHPDO3A) to hepato-cyte in mice. This nanoconstruct can be used to preventhepatocellular damage in the thioacetamide-induced hep-atitis and can simultaneously evaluate the drug deliveryefficiency via Magnetic resonance imaging (MRI), as apo-ferritin cage is efficiently taken up by hepatocyte scav-enger receptor class A type 5 from blood via the ferritintransporting route.161

    Tumor ImagingIn order to improve the quality and accuracy of dis-ease management, a fused technique has been developedby coupling the multiple imaging techniques as shownin Figure 3. Near-infrared fluorescence (NIRF) imagingand positron emission tomography (PET) are combinedin order to minimize the chances of misdiagnosis andused for in vivo imaging. A chimeric ferritin nanocagehas been developed by introducing RGD4C and Cy5.5 on

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2961

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    Figure 2. (a) Schematic illustration of D-RFRTs. Dox was precomplexed with Cu, and then encapsulated into RFRTs. (b) Gel-filtration chromatography analysis of RFRTs and D-RFRTs. The same peak at around 27.4 min was observed for both RFRTs andD-RFRTs. (c) Cumulative drug release curves of D-RFRTs in PBS (pH 7.4) and FBS. (d) Therapy studies performed on U87MGtumor-bearing nude mice (n = 5/group). On day 18, significant difference in tumor growth was found between D-RFRT treatedmice and those treated with PBS, RFRTs and free Dox (P < 0�05). Eighteen days after the onset of the treatment, a TGI rate of89.6% was observed for D-RFRTs, in comparison to that of 74.0% for free Dox. Reprinted with permission from [199], Z. Zhen,et al., RGD-modified apoferritin nanoparticles for efficient drug delivery to tumors. ACS Nano 7, 4830 (2013). © 2013, AmericanChemical Society.

    the exterior surfaces of hybrid ferritin cage via geneticand chemical means. These nanocages loaded with 64Cuonto heavy chain of ferritins have a potential as mul-tifunctional loading and multimodality imaging probes.This hybrid nanoprobe has both PET and NIRF func-tionalities for tumor imaging in conjugation to integrinspecific targeting, when injected intravenously into tumor-bearing mice.103 Similarly, it has been demonstrated thatthe engineered human ferritin protein cages conjugatedwith either fluorescent Cy5.5 molecule or encapsulatingmagnetite nanoparticles, can serve as a nano-platform toimage vascular inflammation in vivo. They can be success-fully taken up by the macrophages in murine atheroscle-rotic carotid arteries and thus served as a novel platform asMR or Near-infrared (NIR) contrast agents for detecting

    macrophage infiltration within atherosclerotic plaques todetect high-risk atherosclerotic plaques.130

    In a recent study, a multifunctional ferritin cage-basednanostructure has been developed for the fluorescence andMR imaging and for detection of �v�3 integrin upregula-tion in tumor cells by attaching green fluorescent protein(GFP) and Arg–Gly–Asp (RGD) peptide on the exteriorsurface of the ferritin cages and ferrimagnetic iron oxidenanoparticles to the interior cavity.132 Paired gold clustershave been synthesized within the interior cavity of apofer-ritin cage with tunable fluorescent emissions, suggestingthe occurrence of fluorescence resonance energy transfer(FRET) effects between the clusters and use of these novelbiomolecule-metal complexes for in vivo kidney targetingand biomedical imaging.145

    2962 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    Figure 3. (a) Schematic illustration of the process of triple-loading. First, we introduced RGD4C and Cy5.5 onto the surfacesof two sets of ferritins, via genetic and chemical means. These two ferritins were then mixed and broken down into subunits atpH = 2 and incubated with 64CuCl2 to achieve radiolabeling. The pH was then adjusted back to 7.4 to facilitate the reformationof nanostructures. The reconstituted chimeric ferritin nanocages have both RGD4C and Cy5.5 on their surfaces and 64Cu loadedin their cavities. In vivo (b) PET and (c) NIRF images after the administration of ferritin probes. In the comparison group, ablocking dose of c (RGDyK) was injected 30 min prior to the ferritin probe administration. Reprinted with permission from [103],X. Lin, et al., Chimeric ferritin nanocages for multiple function loading and multimodal imaging. Nano Lett. 11, 814 (2011). © 2011,American Chemical Society.

    In a similar way, gadolinium 1,4,7-tris(carboxymethyl)-10-(2′-hydroxypropyl)-1,4,7,10-tetraazacyclododecane (Gd-HP-DO3A) loaded apoferritin probe has been used for MRvisualization of tumor blood vessels (tumor angiogenesis)in a mouse model by utilizing biotin-streptavidin affinityand targeting neural cell adhesion molecules.165 Recently, aartificial luminescent protein has been developed by encap-sulating a strongly luminescent Eu3+ complex, N ,N ,N1,N1-[40-(1-naphthyl)-2,20:60,200-terpyridine-6,60 0-diyl]bis(methylenenitrilo) tetrakis(acetic acid) (NTTA–Eu3+�into cavity of apoferritin which act as a bioprobe fortime-gated luminescence bioimaging. This bioprobe canbe used to understand the distribution and function ofapoferritin inside complex living systems.203

    Tumor TargetingProtein based NP systems are the promising tool for thetargeted delivery of imaging and therapeutic agents. Theadvantage of these NPs over other conventional systemslies in their ease to undergo cage modification and extendsto the wide possibility for loading a variety of moieties

    for diagnostic and therapeutic purposes. These functionalmoieties include targeting agents that can effectively rec-ognize the receptor, over expressed by specific cells andtissues.Magnetic nanoparticles loaded apoferritin conjugated

    with fluorescently labeled RGD-4C peptide can be takenup by macrophages more efficiently due to their spe-cific affinity with amelanotic melanoma cells and THP-1monocyte cells, which are known to overexpress integrin�v�3.

    214 Similarly, a multifunctional NPs have been for-mulated for cell specific targeting by encapsulating ironoxide (magnetite) NPs within the interior cavity of genet-ically engineered human H-chain ferritin (HFn) and a flu-orescent dye, Fluorescein- 5-maleimide along with cellspecific targeting peptide, RGD-4C as shown in Figure 4.RGD-4C were attached on its exterior surface whichenabled specific binding to �v�3 integrins upregulated ontumor vasculature and C32 melanoma cells in vitro.122

    Recently, multifunctional nanoparticles have been devel-oped by genetically and chemically modifying the heavychain of the human protein ferritin (HFt), stabilizing andmasking them with polyethylene glycol (PEG) molecules,

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2963

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    Figure 4. TEM images (left) and DLS analysis (right; insets are the corresponding correlation functions) of empty HFn andRGD4C-Fn. Both HFn and RGD4C-Fn show 12–14 nm in diameter. Reprinted with permission from [122], M. Uchida, et al., Tar-geting of cancer cells with ferrimagnetic ferritin cage nanoparticles. J. Am. Chem. Soc. 128, 16629 (2006). © 2006, AmericanChemical Society.

    rhodamine fluorophores and magnetic resonance imag-ing contrasting agents for selective melanoma-targetingboth in vitro and in vivo. These constructs were specifi-cally targeted to the melanoma cell by attaching selectivetargeting moiety, such as �-melanocyte stimulating hor-mone (�-MSH) peptide on the surface of protein, whichbinds to the receptors expressed only by melanoma cellsand to some extend by melanocytes. In this study, therewas considerable reduction in non-specific recognitionand uptake by the reticuloendothelial and mononuclearphagocytic systems as HFt-MSH-PEG were easily recog-nized and taken by the melanoma cells and not by otherhuman cancer cells or mouse tissues (expect by dedicatedphagocytes).128

    Cellular UptakeFerritin in natural conditions enters into cell though recep-tor mediated endocytosis due to the presence of endoge-nous ferritin receptors and for site specific targeting offerritin their exterior surface could be modified. The recep-tor for ferritin varies with the type of cell and tissue andon their developmental stages. The ferritin receptors werefound on different types of cells including lymphocytes,215

    placental microvilli,216 and erythroid precursors.217 Theseare also found on various cell lines, such as giant HeLacells,218 K562 cells,219 and human intestinal carcinoma

    Caco-2 cells, which can even internalize plant ferritin.220

    In absence of transferrin receptors, ferritin L-chain recep-tors (scara 5) have been found on developing kidney forthe iron uptake.221

    It has been previously reported that ferritin bind to themembrane of HeLa cells and is internalized through fer-ritin receptors via endocytosis.218 For example, in embryoTim2 is reported as the receptor for H-ferritin,222 whereasin many other cell lines such as HeLa cells and immuno-genic cells such as mitogen-activated T- and B-cells, cel-lular uptake is facilitated via human transferrin receptor-1(TfR1).223

    Apoferritin nanocage can act as a natural and bio-compatible carrier for the cellular delivery of bioac-tive molecules through receptor-mediated endocytosis andprovide a non-destructive (to the cell membrane) andswitchable control of their cellular uptake by inhibitionof endocytosis which make them a highly flexible andpractical nanocarrier for drug delivery. The control of thedelivery system was tested on human intestinal epithelialCaco-2 cells, as they exhibit ferritin receptors.143

    BioassaysIn the modern era of nanotechnology, use of nanoparticleshas emerged as an important tool in the field of biomedicalapplications because of their simplicity, high surface area

    2964 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    and unique physiochemical properties at the nanoscale.They have been widely utilized in development of highlysensitive bioassays for biomolecular diagnosis. Apoferritinin combination with other metal NPs have been extensivelystudied for molecular diagnosis, bioimaging, targeted drugdelivery and therapeutics.A fluorescence marker (fluorescein anion) and a redox

    marker [hexacyanoferrate (III)] loaded apoferritin hasbeen synthesized that can be used as bioassay labels for

    Figure 5. (A) Magnetic beads and electrochemical sandwichimmunoassay protocol based on biotin-functionalized hexa-cyanoferrate MLAN labels. (B) Typical square wave voltam-mograms of electrochemical immunoassay with increasingconcentration of the IgG (from a to e, 0.1, 0.5, 2, 10, and20 ng mL−1 IgG, respectively). A baseline correction of theresulting voltammogram was performed using the “linearbaseline correction” mode of the CHI 660 (CH Instruments)software. Also shown (insets), (top) the resulting calibrationplot and (bottom) the square wave voltammograms (with-out baseline correction) of 0.1 and 0 ng mL−1 (control) IgG.After the sandwich hybridization assay, the magnetic bead-hexacyanoferrate loaded apoferritin hybrid was dispersed in50 �L of 0.1 M HCl/KCl to release the captured hexacyano-ferrate. Following a magnetic separation, the solution wastransferred to a SPE surface for SWV scanning. Reprintedwith permission from [180], G. Liu, et al., Versatile apoferritinnanoparticle labels for assay of protein. Anal. Chem. 78, 7423(2006). © 2006, American Chemical Society.

    microscopic fluorescence immunoassay and electrochem-ical immunoassay, respectively as shown in Figure 5. Itsdetection limits were estimated to be of 0.06 (0.39 pM)and 0.08 ng mL−1 (0.52 pM) IgG with fluorescein andhexacyanoferrate, respectively.180�181 The biologically pro-duced functionalized NPs were also used as labelingagents in bioaffinity assay. In this study, Eu3+ ions wereused as labeling agent and were loaded inside the fer-ritin, while a binding moiety i.e., single chain Fv fragment(scFv) of an antibody was attached on its surface in orderto aid their specific binding to the thyroid stimulating hor-mone (TSH).202

    A new highly sensitive and selective magnetic parti-cle (MP)-based electrochemical immunoassay has beendemonstrated, having a detection limit of 0.01 ng/mLusing carbon nanospheres (NS) and lead phosphate loadedprotein cage nanoparticles (PCN) for signal amplification.This system has been used to analyze the phosphorylatedprotein human phospho-p5315, a potential biomarker ofgamma-radiation exposure.167

    In a similar way, a co-reactant based highly sensitiveelectro chemiluminescence (ECL) immunoassay approachhas been devised based on PEI loaded apoferritin NPs,probes for the specific quantification of human chorionicgonadotrophin (HCG) by enhancing the ECL of ruthe-nium (II) tris(2,2′-bipyridyl) (Ru(bpy)+32 ).

    201 Moreover, forrapid, sensitive, selective and inexpensive quantification oforganophosphorylated acetylcholinesterase (OP-AChE), anexposure biomarker of organophosphate based pesticides.A new sandwich type electrochemical immunoassay hasbeen developed using apoferritin templated lead phosphatelabel for quantification of OP-AChE, having a detectionlimit of 0.02 nM.168 These new apoferritin based nanopar-ticle labels hold great promise in the field of biomoleculedetection and in enhancing the sensitivity of various otherbioassays.

    BiosensorsThe metal encapsulated apoferritin NPs can be used invariety of nanodevices, such as single electron transistor,catalysis and floating gate memory. In similar way, semi-conductor NPs such as CdSe, ZnSe, and CdS encapsulatedapoferritin can be used as quantum dots and photofluores-cence markers. ZnSe is a n-type semiconductor that couldbe used as fluorescent labels for biological applications astheir fluorescent light does not quench easily.Ferritin molecules have redox property which remains

    unchangeable until their electrochemical surrounding isfixed.224 In recent years, protein electrochemistry hasemerged as an interesting area in the development ofbiosensors and bioreactors. Various electron transfer reac-tion studies of ferritin have been conducted, such aselectron transfer of ferritin on bare gold electrode.225

    The electrochemical behavior of ferritin adsorbed onindium–tin oxide (ITO) glass and single wall nano-tubes (SWNT)/ferritin composite on glassy carbon (GC)

    J. Biomed. Nanotechnol. 10, 2950–2976, 2014 2965

  • Ferritin Nanocages: A Novel Platform for Biomedical Applications Bhushan et al.

    disk electrode had been studied for nanoelectronicapplications.226�227 Moreover, electrochemical studies onferritin immobilized onto a self-assembled monolayer-modified gold electrode have been already reported.228 Thedirect electron transfer of ferritin in Dihexadecyl phos-phate (DHP) on Au film electrode was also evaluated.229

    A ferritin/DNA complex was successfully constructedby chemically attaching maleimide modified DNA(M-DNA) to the exterior surface of a ferritin mutant pro-tein, which can be sterically attached to the complementaryDNA-functionalized GNPs. This complex can be utilizedin photo electrochemical biosensor fabrication as it canserve as a mediator between the DNA/RNA responsiblefor disease and dye-labeled photo reporter probe.137

    A highly sensitive electrochemical approach have beenreported having a linear range from 2�0× 10−16 to 1�0×10−14 M and the detection limit was 5�1×10−17 M underoptimum condition, based on signal dual-amplificationwith Au NPs and marker-loaded apoferritin NPs for thesequence-specific DNA detection. The concentration oftarget DNA is quantified by electrochemical stripping anal-ysis of the electroactive cadmium markers released fromapoferritin NPs in acidic buffers. This proposed DNAbiosensor has high sensitivity, good reproducibility andselectivity even against two-base mismatched DNA.176

    Recently, a direct electron transfer has been investigatedbetween cobalt NPs loaded apoferritin and a glassy carbonelectrode in thin film of dihexadecyl phosphate (DHP) bycyclic voltammetry (CV) in order to design a biosensingdevice that can be used in detection of various chemicaland biological analytes.112

    In addition to this, an electrochemical approach has beendeveloped using metal phosphate nanoparticles loadedmonobase-conjugated apoferritin probe for the detection ofindividual single nucleotide polymorphisms (SNPs). Thebiotinylated DNA probes get hybridized with mutant andcomplementary DNA and the duplex DNA helix formwere captured on the surface magnetic beads by biotin-streptavidin based affinity binding. Signals were gener-ated and detected by electrochemical stripping analyses,when the probes get coupled to the mutant sites of formedduplex DNA by DNA polymerase, as each mutation cap-tures different nucleotide-conjugated apoferritin probe andgenerates distinct potential voltammogram peaks relativeto mismatch.156

    An array of charged storage nodes in floating gate mem-ory had been developed using ferritin encapsulated NPs.230

    Apoferritin loaded with Ni atoms catalyzed the fabricationof high quality polycrystalline silicon (Si) thin film froman amorphous Si thin film.231

    In general, electrochemical biosensors utilize thepotentiometric and amperometric transducers that con-vert the biosensing information into the measurablesignal. Recently, apoferritin encapsulated gold nanoparti-cles have been utilized to perform electrochemical DNAbiosensing having a sensitivity up to 51 aM.176 Apoferritin

    bionanomaterial also enhances electron transfer reactionsof hemoglobin in a wide pH range. Since, the Hb exhibitcatalytic activity toward H2O2, the construct can be usedfor the development of H2O2 biosensor.

    232

    BiocatalystApoferritin loaded nanoparticles also found their rolein catalyzing various chemical reactions. As shown inFigure 6, polymerization of phenylacetylene has been cat-alyzed by the Rhodium (Rh(nbd)) complexes immobilizedwithin the discrete space of apoferritin that can be usefulin investigating the behavior of a single polymer chain iso-lated within a nano-sized space.60 Similarly, they have alsodemonstrated that the ferrocenes and Pd(allyl) complexeswere immobilized on the interior surface of apoferritin.The Pd(allyl) complexes immobilized by forming a thiol-bridged dinuclear complexes and catalyzed the redox andSuzuki coupling reactions.101�233

    Figure 6. Polymerization of phenylacetylene catalyzedby Rh(nbd) · apo-Fr. (a) Solution of Rh(nbd) · apo-Fr priorto addition of phenylacetylene (b) Reaction mixtureof Rh(nbd) ·apo-Fr and phenylacetylene after stirring for 3 h at25 �C. (c) Reaction mixture of [Rh(nbd)Cl]2 and phenylacety-lene under the same conditions. (d)–(f) Elution profiles fromsize-exclusion chromatography of (d) Rh(nbd) · apo-Fr afterthe reaction, (e) Rh(nbd) · apo-Fr, and (f) apo-Fr. Elutionwas monitored at both 280 nm (black line) and 383 nm (redline). Reprinted with permission from [60], S. Abe, et al.,Polymerization of phenylacetylene by rhodium complexeswithin a discrete space of apo-ferritin. J. Am. Chem. Soc. 131,6959 (2009). © 2009, American Chemical Society.

    2966 J. Biomed. Nanotechnol. 10, 2950–2976, 2014

  • Bhushan et al. Ferritin Nanocages: A Novel Platform for Biomedical Applications

    In a similar way, Pd has been encapsulated within thecore of a hyperthermophilic ferritin cages (from pyro-coccus furiosus) to form a hybrid catalysts that can beused for highly specific aerobic oxidation of alcohols inwater.99 In addition to this, it has been demonstrated thatthe apoferritin encapsulated Pd nanocluster catalyzes thesize-selective olefin hydrogenation.100

    The Fe(O)OH-mineralized iron storage protein fer-ritin was used to catalyze the photoreduction of aque-ous Cr-(VI) species to Cr(III), Cu(II) to form a stable,air sensitive, colloidal dispersion of Cu(0) and reductionof cytochrome c and viologens as well as the oxidationof carboxylic acids, thiol compounds, and sulfite. Fer-ritin act as photocatalyst in presence of UV/visible lightand can be utilized for future photocatalytic applications,such as in environmental remediation chemistry.125�126�104

    These NPs also acts as a catalyst for the growth of single-walled carbon nanotubes.124 Similarly, it has been reportedthat 1–2 nm and 3–5 nm diameter range discrete cat-alytic nanoparticles synthesized in apoferritin cavity canbe used for the growth of SWNTs on substrate by chem-ical vapor deposition (CVD) and diameter of nanotubeswas controlled by getting hold on the structure of catalyticNPs in core.127 Moreover, apoferritin encapsulated AuNPs exhibited catalytic synthesis of single-walled carbonnanotubes (SWCNTs) on various substrates by chemicalvapor deposition.146 In another similar attempt, ferritincage loaded with catalytic Au NPs were immobilized toa silicon substrate for the growth of silicon nanowire(SiNW) by CVD.147

    The apoferritin encapsulated homogeneous gold-silveralloy NPs aid in the catalytic reduction of 4-nitrophenolinto 4-aminophenol in the presence of NaBH4.

    152

    A bimetallic nanoreactor is prepared by loading Au–PdNPs in apoferritin core that shows 2.5-fold higher catalyticreactivity of olefin hydrogenation as compared to Pd0 NPsin the cage.151

    Enzyme ImmobilizationNow a days, researchers are interested in stabilizingenzymes and retaining their activity as they are promisingtools for wide range of applications including biocatalysis,bioassay, bioenergy conversion and environmental reme-diation. A large number of techniques are available forthe enzyme immobilization but most of them have certainlimitations, which include loss of enzymatic activity dur-ing immobilization, stability and low efficiency. Therefore,there is a need for development of new novel immobi-lization technique. A large number of inorganic materi-als are used for enzyme immobilization but they are notbiocompatible.Apoferritin provide a biocompatible nanosized con-

    tainer for the synthesis of biomaterials. It has beenrecently shown that the apoferritin can also be usedin stabilizing enzymes and also to enhance their activ-ity. Immobilization of glucose oxidase (GOx) has been

    reported on the surface of apoferritin by green syntheticapproach. A glucose oxidase–biotin/streptavidin/biotin–apoferritin conjugate (Apo–GOx) was formed by bridgingwith streptavidin. The Apo-GOx formed shows enhancedthermal and chemical stabilities.200

    Artificial AntioxidantA naturally occurring antioxidant enzyme includes theendogenous superoxide dismutase (SOD), but it is foundto be incapable in protecting the cells from sudden oxida-tive damage. Therefore, current research is now focus-ing on the development of artificial antioxidant havinga high ROS-scavenging capability and low cytotoxicity.Nanoceria (nano-CeO2) is now being studied because oftheir SOD mimetic activity and other properties, such asreversibility and auto regeneration.234�235

    Recently, a nano-CeO2 has been constructed withinthe cavity of apoferritin protein cage, which improvesbiocompatibility and manipulate electron localization onthe surface of nanoparticles thereby improving the ROS-scavenging activity of this nanocomposite. It was sug-gested that the increase in redox activity of CeO2 is dueto change in the surface morphology/surface defect orvacancies due to the charge transfer process that changethe electron localization on the surface of nano-CeO2,which e