Enhanced paracellular transport of insulin can be achieved via transient induction of myosin light chain phosphorylation

Autor: Alistair Taverner, Randall J. Mrsny, Ian M. Eggleston, Ruggero Dondi, Floriane Laurent, Nikoletta Fotaki, Siân-Eleri Owens, Khaled Almansour
Rok vydání: 2015
Předmět:
Blood Glucose
Male
MLCP
Myosin light chain phosphatase

PK/PD
Pharmacokinetics/pharmacodynamics

medicine.medical_treatment
Pharmaceutical Science
Muscle Proteins
Peptide
02 engineering and technology
Protein–protein interactions
Paracellular transport
myosin light chain phosphatase
insulin delivery
cell penetrating peptide
protein-protein interactions
tight junction function
Myosin-Light-Chain Phosphatase
Phosphoprotein Phosphatases
Insulin
Phosphorylation
FD
Fluorescent dextran

Myosin light chain phosphatase
chemistry.chemical_classification
pMLC
Phosphorylated myosin light chain

0303 health sciences
Intracellular Signaling Peptides and Proteins
CPP
Cell Penetrating Peptide

PBS
Phosphate buffer saline

TJ
Tight junction

021001 nanoscience & nanotechnology
Intestinal epithelium
PKC
Protein kinase C

Biochemistry
FMC
9-fluorenylmethyloxycarbonyl

MLC
Myosin light chain

Myosin-light-chain phosphatase
MLCK
Myosin light chain kinase

0210 nano-technology
Oligopeptides
Myosin light-chain kinase
Myosin Light Chains
MYPT1
Myosin phosphatase target subunit

Tight junction function
Phosphatase
MTS
3-(4
5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium

Biology
Article
SPPS
Solid phase peptide synthesis

03 medical and health sciences
medicine
Animals
Humans
Rats
Wistar

030304 developmental biology
ILI
Intraluminal injection

Cell penetrating peptide
SC
Subcutaneous

Biological Transport
chemistry
Cell-penetrating peptide
Biophysics
Insulin delivery
Caco-2 Cells
DAPI
4′
6-diamidino-2-phenylindole
Zdroj: Journal of Controlled Release
Taverner, A, Dondi, R, Almansour, K, Laurent, F, Owens, S-E, Eggleston, I M, Fotaki, N & Mrsny, R J 2015, ' Enhanced paracellular transport of insulin can be achieved via transient induction of myosin light chain phosphorylation ', Journal of Controlled Release, vol. 210, pp. 189-197 . https://doi.org/10.1016/j.jconrel.2015.05.270
ISSN: 1873-4995
Popis: The intestinal epithelium functions to effectively restrict the causal uptake of luminal contents but has been demonstrated to transiently increase paracellular permeability properties to provide an additional entry route for dietary macromolecules. We have examined a method to emulate this endogenous mechanism as a means of enhancing the oral uptake of insulin. Two sets of stable Permeant Inhibitor of Phosphatase (PIP) peptides were rationally designed to stimulate phosphorylation of intracellular epithelial myosin light chain (MLC) and screened using Caco-2 monolayers in vitro. Apical application of PIP peptide 640, designed to disrupt protein–protein interactions between protein phosphatase 1 (PP1) and its regulator CPI-17, resulted in a reversible and non-toxic transient reduction in Caco-2 monolayer trans-epithelial electric resistance (TEER) and opening of the paracellular route to 4 kDa fluorescent dextran but not 70 kDa dextran in vitro. Apical application of PIP peptide 250, designed to impede MYPT1-mediated regulation of PP1, also decreased TEER in a reversible and non-toxic manner but transiently opened the paracellular route to both 4 and 70 kDa fluorescent dextrans. Direct injection of PIP peptides 640 or 250 with human insulin into the lumen of rat jejunum caused a decrease in blood glucose levels that was PIP peptide and insulin dose-dependent and correlated with increased pMLC levels. Systemic levels of insulin suggested approximately 3–4% of the dose injected into the intestinal lumen was absorbed, relative to a subcutaneous injection. Measurement of insulin levels in the portal vein showed a time window of absorption that was consistent with systemic concentration-time profiles and approximately 50% first-pass clearance by the liver. Monitoring the uptake of a fluorescent form of insulin suggested its uptake occurred via the paracellular route. Together, these studies add validation to the presence of an endogenous mechanism used by the intestinal epithelium to dynamically regulate its paracellular permeability properties and better define the potential to enhance the oral delivery of biopharmaceuticals via a transient regulation of an endogenous mechanism controlling the intestinal paracellular barrier.
Graphical abstract
Databáze: OpenAIRE