%0 Journal Article %T CAR/FoxP3-engineered T regulatory cells target the CNS and suppress EAE upon intranasal delivery %A Moa Fransson %A Elena Piras %A Joachim Burman %A Berith Nilsson %A Magnus Essand %A BinFeng Lu %A Robert A Harris %A Peetra U Magnusson %A Eva Brittebo %A Angelica SI Loskog %J Journal of Neuroinflammation %D 2012 %I BioMed Central %R 10.1186/1742-2094-9-112 %X CD4+ T cells were modified utilizing a lentiviral vector system to express a chimeric antigen receptor (CAR) targeting myelin oligodendrocyte glycoprotein (MOG) in trans with the murine FoxP3 gene that drives Treg differentiation. The cells were evaluated in vitro for suppressive capacity and in C57BL/6 mice to treat EAE. Cells were administered by intranasal (i.n.) cell delivery.The engineered Tregs demonstrated suppressive capacity in vitro and could efficiently access various regions in the brain via i.n cell delivery. Clinical score 3 EAE mice were treated and the engineered Tregs suppressed ongoing encephalomyelitis as demonstrated by reduced disease symptoms as well as decreased IL-12 and IFNgamma mRNAs in brain tissue. Immunohistochemical markers for myelination (MBP) and reactive astrogliosis (GFAP) confirmed recovery in mice treated with engineered Tregs compared to controls. Symptom-free mice were rechallenged with a second EAE-inducing inoculum but remained healthy, demonstrating the sustained effect of engineered Tregs.CNS-targeting Tregs delivered i.n. localized to the CNS and efficiently suppressed ongoing inflammation leading to diminished disease symptoms. %K MS %K redirected cells %K T regulatory cells %K EAE %K FoxP3 %K Myelin oligodendrocyte glycoprotein (MOG) %U http://www.jneuroinflammation.com/content/9/1/112/abstract