Previous Page  5 / 18 Next Page
Information
Show Menu
Previous Page 5 / 18 Next Page
Page Background

Nanobiotechnology 2018

Page 23

Nano Research & Applications

ISSN: 2471-9838

E u r o S c i C o n C o n f e r e n c e o n

Nanotech & Nanobiotechnology

J u l y 1 2 - 1 3 , 2 0 1 8

P a r i s , F r a n c e

T

he intranasal route of drug delivery has traditionally been used to

administer small, lipophilic drugs that are rapidly absorbed into capillaries

of the nasal epithelium, resulting in rapid onset of CNS actions. Many neuro-

therapeutic agents, especially polynucleotides and proteins, do not readily

cross the blood-brain barrier and cannot survive intact in the gut or blood.

Hence, gene therapy for brain disorders has required direct neurosurgical

microinjection or infusion into brain or cerebrospinal fluid. However, recent

research has demonstrated direct nose-to-brain delivery of relatively large

molecules, including neurotrophins (NGF and insulin-like growth factor [IGF]-

1), neuropeptides, cytokines (interferon β-1b and erythropoietin as well as

polynucleotides (DNA plasmids and genes). The present report describes

development of a novel manganese-chelate nanocarrier system for direct nose-

to-brain delivery of small interfering RNA (siRNA) or DNA. The manganese

(Mn) chelate Mangafodopir served 2 functions: 1) as a marker of the NPs

for intracerebral tracking with magnetic resonance imaging (MRI) and 2) as

a cross-linker of the chitosan matrix in the nanocarrier structure. Using high

field small animal MRI, the Mn-tagged NPs were visualized on T1-weighted

images and were found to penetrate from nasal epitheliaum into olfactory bulb

and across brain regions following intranasal instillation of the nanocarriers.

In addition, Mn content of the nanocarrier did not impede the functional activity

of siRNA directed against green fluorescent protein eGFP in transgenic green

mice. Expression of eGFP mRNA in transgenic green mice was decreased by at

least 50% in four brain regions. Those brain regions also exhibited significantly

increased Mn signal in T1-weighted MR images. In separate experiments, we

showed that mNPs loaded with dsDNA encoding the red fluorescent protein

(RFP) was expressed in corpus striatum and other regions following intranasal

administration. Hence, this novel nanocarrier system permitted

in vivo

tracking

of the therapeutic agent and was effective in delivering nucleic acid payloads

that exhibited the expected activity in brain tissue.

Biography

Juan Sanchez-Ramos received a PhD in Pharmacology and

Physiology from the University of Chicago and aMedical Degree

(MD) from the University of Illinois. He trained in Neurology

at the University of Chicago and as a Fellow in Movement

Disorders at the University of Miami. Currently, he is a Professor

of Neurology at the University of South Florida in Tampa where

he holds the Helen Ellis Endowed Chair for Parkinson's disease

Research and is Director of the HDSA Center of Excellence

for Huntington’s disease. He is also Medical Director of the

non-profit Parkinson Research Foundation based in Sarasota

FL. In addition to teaching and attending to patients with

Movement Disorders, he has directed basic research projects

in neurodegeneration, neurotoxicology, adult stem cell biology

and presently is focused on novel approaches for non-invasive

delivery of gene therapy to brain.

jsramos@health.usf.edu

Nanocarriers for nose-to-brain, non-invasive delivery of gene

therapy

Juan Sanchez-Ramos

1

, Shijie Song

1

, Xiaoyuan Kong

1

, Gary

Martinez

2

, Shyam Mohapatra

3

, Subra Mohapatra

4

, Reka A

Haraszti

5

, Anastasia Khvorova

5

, Neil Aronin

5

, Vasyl Sava

1

1

University of South Florida, USA

2

Moffitt Cancer Center and Research Institute, USA

3

Nanomedicine Research Center, USA

4

Department of Molecular Medicine, USA

5

RNA Therapeutics Institute-UMASS, USA

Juan Sanchez-Ramos et al., Nano Res Appl 2018, Volume 4

DOI: 10.21767/2471-9838-C2-011