Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System

The administration of therapeutics to peripheral nerve tissue is challenging due to the complexities of peripheral neuroanatomy and the limitations imposed by the blood–nerve barrier (BNB). Therefore, there is a pressing need to enhance delivery effectiveness and implement targeted delivery methods....

Full description

Bibliographic Details
Main Authors: Mena Asha Krishnan, Olawale A. Alimi, Tianshu Pan, Mitchell Kuss, Zeljka Korade, Guoku Hu, Bo Liu, Bin Duan
Format: Article
Language:English
Published: MDPI AG 2024-01-01
Series:Pharmaceutics
Subjects:
Online Access:https://www.mdpi.com/1999-4923/16/1/102
_version_ 1797342729918742528
author Mena Asha Krishnan
Olawale A. Alimi
Tianshu Pan
Mitchell Kuss
Zeljka Korade
Guoku Hu
Bo Liu
Bin Duan
author_facet Mena Asha Krishnan
Olawale A. Alimi
Tianshu Pan
Mitchell Kuss
Zeljka Korade
Guoku Hu
Bo Liu
Bin Duan
author_sort Mena Asha Krishnan
collection DOAJ
description The administration of therapeutics to peripheral nerve tissue is challenging due to the complexities of peripheral neuroanatomy and the limitations imposed by the blood–nerve barrier (BNB). Therefore, there is a pressing need to enhance delivery effectiveness and implement targeted delivery methods. Recently, erythrocyte-derived exosomes (Exos) have gained widespread attention as biocompatible vehicles for therapeutics in clinical applications. However, engineering targeted Exos for the peripheral nervous system (PNS) is still challenging. This study aims to develop a targeted Exo delivery system specifically designed for presynaptic terminals of peripheral nerve tissue. The clostridium neurotoxin, tetanus toxin-C fragment (TTC), was tethered to the surface of red blood cell (RBC)-derived Exos via a facile and efficient bio-orthogonal click chemistry method without a catalyst. Additionally, Cyanine5 (Cy5), a reactive fluorescent tag, was also conjugated to track Exo movement in both in vitro and in vivo models. Subsequently, Neuro-2a, a mouse neuronal cell line, was treated with dye-labeled Exos with/without TTC in vitro, and the results indicated that TTC-Exos exhibited more efficient accumulation along the soma and axonal circumference, compared to their unmodified counterparts. Further investigation, using a mouse model, revealed that within 72 h of intramuscular administration, engineered TTC-Exos were successfully transported into the neuromuscular junction and sciatic nerve tissues. These results indicated that TTC played a crucial role in the Exo delivery system, improving the affinity to peripheral nerves. These promising results underscore the potential of using targeted Exo carriers to deliver therapeutics for treating peripheral neuropathies.
first_indexed 2024-03-08T10:37:31Z
format Article
id doaj.art-73a7109c4b2646c38f89400bede28ebd
institution Directory Open Access Journal
issn 1999-4923
language English
last_indexed 2024-03-08T10:37:31Z
publishDate 2024-01-01
publisher MDPI AG
record_format Article
series Pharmaceutics
spelling doaj.art-73a7109c4b2646c38f89400bede28ebd2024-01-26T18:07:34ZengMDPI AGPharmaceutics1999-49232024-01-0116110210.3390/pharmaceutics16010102Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous SystemMena Asha Krishnan0Olawale A. Alimi1Tianshu Pan2Mitchell Kuss3Zeljka Korade4Guoku Hu5Bo Liu6Bin Duan7Mary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USAMary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USAMary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USAMary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USADepartment of Pediatrics, University of Nebraska Medical Center, Omaha, NE 68198, USADepartment of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USAMary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USAMary and Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE 68198, USAThe administration of therapeutics to peripheral nerve tissue is challenging due to the complexities of peripheral neuroanatomy and the limitations imposed by the blood–nerve barrier (BNB). Therefore, there is a pressing need to enhance delivery effectiveness and implement targeted delivery methods. Recently, erythrocyte-derived exosomes (Exos) have gained widespread attention as biocompatible vehicles for therapeutics in clinical applications. However, engineering targeted Exos for the peripheral nervous system (PNS) is still challenging. This study aims to develop a targeted Exo delivery system specifically designed for presynaptic terminals of peripheral nerve tissue. The clostridium neurotoxin, tetanus toxin-C fragment (TTC), was tethered to the surface of red blood cell (RBC)-derived Exos via a facile and efficient bio-orthogonal click chemistry method without a catalyst. Additionally, Cyanine5 (Cy5), a reactive fluorescent tag, was also conjugated to track Exo movement in both in vitro and in vivo models. Subsequently, Neuro-2a, a mouse neuronal cell line, was treated with dye-labeled Exos with/without TTC in vitro, and the results indicated that TTC-Exos exhibited more efficient accumulation along the soma and axonal circumference, compared to their unmodified counterparts. Further investigation, using a mouse model, revealed that within 72 h of intramuscular administration, engineered TTC-Exos were successfully transported into the neuromuscular junction and sciatic nerve tissues. These results indicated that TTC played a crucial role in the Exo delivery system, improving the affinity to peripheral nerves. These promising results underscore the potential of using targeted Exo carriers to deliver therapeutics for treating peripheral neuropathies.https://www.mdpi.com/1999-4923/16/1/102red blood cellstetanus toxin-C fragmentclick chemistrytarget deliveryperipheral nerve injury
spellingShingle Mena Asha Krishnan
Olawale A. Alimi
Tianshu Pan
Mitchell Kuss
Zeljka Korade
Guoku Hu
Bo Liu
Bin Duan
Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System
Pharmaceutics
red blood cells
tetanus toxin-C fragment
click chemistry
target delivery
peripheral nerve injury
title Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System
title_full Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System
title_fullStr Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System
title_full_unstemmed Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System
title_short Engineering Neurotoxin-Functionalized Exosomes for Targeted Delivery to the Peripheral Nervous System
title_sort engineering neurotoxin functionalized exosomes for targeted delivery to the peripheral nervous system
topic red blood cells
tetanus toxin-C fragment
click chemistry
target delivery
peripheral nerve injury
url https://www.mdpi.com/1999-4923/16/1/102
work_keys_str_mv AT menaashakrishnan engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT olawaleaalimi engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT tianshupan engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT mitchellkuss engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT zeljkakorade engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT guokuhu engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT boliu engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem
AT binduan engineeringneurotoxinfunctionalizedexosomesfortargeteddeliverytotheperipheralnervoussystem