Please use this identifier to cite or link to this item: http://hdl.handle.net/10603/522591
Title: Characterization of oral and subcutaneous efficacy of GMFBI 1 in the suppression of Experimental Autoimmune Encephalomyelitis progression
Researcher: Sunitha Subhramanian
Guide(s): Krishnakumar Menon and Gopi Mohan C
Keywords: Life Sciences
Molecular Biology and Genetics; Pharmacokinetics; Multiple Sclerosis
University: Amrita Vishwa Vidyapeetham University
Completed Date: 2023
Abstract: In the emerging significance of research on gut-brain control of multiple sclerosis (MS), therapeutic targeting of gut-brain inflammation modulation at a fundamental level is more enlivening, wherein the immunomodulator glia maturation factor-and#946; (GMF-and#946;) plays an important role in proinflammation mediation. Activation of GMF-and#946; leads to GMF-and#946;-Ser-83 phosphorylation which results in proinflammation and augments experimental autoimmune encephalomyelitis (EAE). Remarkably, there were no EAE symptoms in GMF-and#946;-/- mice. Consequently, our group recognized 1H-Indazole4-yl-methanol (GMFBI.1) inhibitor which prevented GMF-and#946;-Ser-83 phosphorylation in EAE suppression. Moreover, to ascertain the role of gut GMF-and#946; in EAE suppression and the impact of gut-brain implication in neurodegenerative diseases, we amended the gut bioavailability of GMFBI.1 as a parameter of EAE suppression. Primarily, we pinpointed Miglyol 812N as an appropriate biocompatible GMFBI.1 carrier in contrast to other FDA approved carriers screened using in silico molecular docking. GMFBI.1 Miglyol 812N formulation enhanced the retention/brain penetration of the inhibitor. Consequently, administration of GMFBI.1-Miglyol 812N at varying doses by subcutaneous/oral routes resulted in differential GMFBI.1 bioavailability in both gut and brain determined by pharmacokinetic approach indicating the effect of local GMFBI.1 inhibitor bioavailability in EAE reversal. Insufficient gut GMFBI.1 bioavailability caused incomplete EAE reversal despite having adequate GMFBI.1 inhibitor in the systemic circulation to hamper brain GMF-and#946; activity. On the other hand, adequate GMFBI.1 gut bioavailability led to absolute EAE reversal. The extent of GMF-and#946;-Ser-83 phosphorylation/GM-CSF expressions in the enteric glial cells and the partial/full recovery from EAE is linked to GMFBI.1 compound bioavailability. In addition, up-regulation of Treg cells within the periphery following GMFBI.1 p.o. treatment abrogated EAE clinical progression. Necessarily, dual targeting of GMF-and#946; in both..
Pagination: ix, 134
URI: http://hdl.handle.net/10603/522591
Appears in Departments:Amrita Centre for Nanosciences and Molecular Medicine

Files in This Item:
File Description SizeFormat 
01_title.pdfAttached File226.19 kBAdobe PDFView/Open
02_preliminary page.pdf709.5 kBAdobe PDFView/Open
03_contents.pdf230.97 kBAdobe PDFView/Open
04_abstract.pdf179.95 kBAdobe PDFView/Open
05_chapter 1.pdf7.63 MBAdobe PDFView/Open
06_chapter 2.pdf3.49 MBAdobe PDFView/Open
07_chapter 3.pdf14.18 MBAdobe PDFView/Open
08_chapter 4.pdf312.62 kBAdobe PDFView/Open
09_chapter 5.pdf1.55 MBAdobe PDFView/Open
10_annexure.pdf402.21 kBAdobe PDFView/Open
80_recommendation.pdf1.77 MBAdobe PDFView/Open
Show full item record


Items in Shodhganga are licensed under Creative Commons Licence Attribution-NonCommercial-ShareAlike 4.0 International (CC BY-NC-SA 4.0).

Altmetric Badge: