File Download

There are no files associated with this item.

  Links for fulltext
     (May Require Subscription)
Supplementary

Article: Spatiotemporal tracking of gold nanorods after intranasal administration for brain targeting

TitleSpatiotemporal tracking of gold nanorods after intranasal administration for brain targeting
Authors
KeywordsBiodistribution
Brain region-specific uptake
Glioma
Gold nanorods
Intranasal administration
Multi-modal imaging
Issue Date2023
Citation
Journal of Controlled Release, 2023, v. 357, p. 606-619 How to Cite?
AbstractIntranasal administration is becoming increasingly more attractive as a fast delivery route to the brain for therapeutics circumventing the blood-brain barrier (BBB). Gold nanorods (AuNRs) demonstrate unique optical and biological properties compared to other gold nanostructures due to their high aspect ratio. In this study, we investigated for the first time the brain region-specific distribution of AuNRs and their potential as a drug delivery platform for central nervous system (CNS) therapy following intranasal administration to mice using a battery of analytical and imaging techniques. AuNRs were functionalized with a fluorescent dye (Cyanine5, Cy5) or a metal chelator (diethylenetriaminepentaacetic dianhydride, DTPA anhydride) to complex with Indium-111 via a PEG spacer for optical and nuclear imaging, respectively. Direct quantification of gold was achieved by inductively coupled plasma mass spectrometry. Rapid AuNRs uptake in mice brains was observed within 10 min following intranasal administration which gradually reduced over time. This was confirmed by the 3 imaging/analytical techniques. Autoradiography of sagittal brain sections suggested entry to the brain via the olfactory bulb followed by diffusion to other brain regions within 1 h of administration. The presence of AuNR in glioblastoma (GBM) tumors following intranasal administration was also proven which opens doors for AuNRs applications, as nose-to-brain drug delivery carriers, for treatment of a range of CNS diseases.
Persistent Identifierhttp://hdl.handle.net/10722/349897
ISSN
2023 Impact Factor: 10.5
2023 SCImago Journal Rankings: 2.157

 

DC FieldValueLanguage
dc.contributor.authorHan, Shunping-
dc.contributor.authorWang, Julie Tzu Wen-
dc.contributor.authorYavuz, Emine-
dc.contributor.authorZam, Alaa-
dc.contributor.authorRouatbi, Nadia-
dc.contributor.authorUtami, Rifka Nurul-
dc.contributor.authorLiam-Or, Revadee-
dc.contributor.authorGriffiths, Alexander-
dc.contributor.authorDickson, Wayne-
dc.contributor.authorSosabowski, Jane-
dc.contributor.authorAl-Jamal, Khuloud T.-
dc.date.accessioned2024-10-17T07:01:42Z-
dc.date.available2024-10-17T07:01:42Z-
dc.date.issued2023-
dc.identifier.citationJournal of Controlled Release, 2023, v. 357, p. 606-619-
dc.identifier.issn0168-3659-
dc.identifier.urihttp://hdl.handle.net/10722/349897-
dc.description.abstractIntranasal administration is becoming increasingly more attractive as a fast delivery route to the brain for therapeutics circumventing the blood-brain barrier (BBB). Gold nanorods (AuNRs) demonstrate unique optical and biological properties compared to other gold nanostructures due to their high aspect ratio. In this study, we investigated for the first time the brain region-specific distribution of AuNRs and their potential as a drug delivery platform for central nervous system (CNS) therapy following intranasal administration to mice using a battery of analytical and imaging techniques. AuNRs were functionalized with a fluorescent dye (Cyanine5, Cy5) or a metal chelator (diethylenetriaminepentaacetic dianhydride, DTPA anhydride) to complex with Indium-111 via a PEG spacer for optical and nuclear imaging, respectively. Direct quantification of gold was achieved by inductively coupled plasma mass spectrometry. Rapid AuNRs uptake in mice brains was observed within 10 min following intranasal administration which gradually reduced over time. This was confirmed by the 3 imaging/analytical techniques. Autoradiography of sagittal brain sections suggested entry to the brain via the olfactory bulb followed by diffusion to other brain regions within 1 h of administration. The presence of AuNR in glioblastoma (GBM) tumors following intranasal administration was also proven which opens doors for AuNRs applications, as nose-to-brain drug delivery carriers, for treatment of a range of CNS diseases.-
dc.languageeng-
dc.relation.ispartofJournal of Controlled Release-
dc.subjectBiodistribution-
dc.subjectBrain region-specific uptake-
dc.subjectGlioma-
dc.subjectGold nanorods-
dc.subjectIntranasal administration-
dc.subjectMulti-modal imaging-
dc.titleSpatiotemporal tracking of gold nanorods after intranasal administration for brain targeting-
dc.typeArticle-
dc.description.naturelink_to_subscribed_fulltext-
dc.identifier.doi10.1016/j.jconrel.2023.04.022-
dc.identifier.pmid37061195-
dc.identifier.scopuseid_2-s2.0-85153044974-
dc.identifier.volume357-
dc.identifier.spage606-
dc.identifier.epage619-
dc.identifier.eissn1873-4995-

Export via OAI-PMH Interface in XML Formats


OR


Export to Other Non-XML Formats