Previous Page  72 / 120 Next Page
Information
Show Menu
Previous Page 72 / 120 Next Page
Page Background

Liposomes, Exosomes, and Virosomes: From Modeling Complex

Membrane Processes to Medical Diagnostics and Drug Delivery

Poster Abstracts

67

46-POS

Board 23

On Chip Biodetection, Analysis and Nanometrology of Cell-Derived Vesicles

Sameh OBEID

1

, Gaël Belliot

2

, Benoit Le Roy de Boiseaumarié

1

, Adam Ceroi

3

, Guillaume

Mourey

3

, Philippe Saas

3

, Wilfrid Boireau

1

, Céline Elie-Caille

1

.

1

FEMTO-ST Institute, Besançon, France,

2

National Reference Center for Enteric Viruses, Public

Hospital, Dijon, France,

3

INSERM U1098, EFS Bourgogne Franche-Comté, UBFC, Besançon,

France.

Extracellular vesicles (EVs) are small vesicles (10-4000 nm) shed from different cells types upon

activation or apoptosis. EVs, including exosomes, microparticles and apoptotic bodies, are

recognized to play an important role in various biological processes and are also recognized as

potential biomarkers of various health disorders

1

.

Currently, the technic of choice used for the detection of EVs is flow cytometry (FC)

2

, but the

main drawback of FC is the lower detection limit (≈300nm at best); consequently, a big fraction

of vesicles cannot be detected.

Our work aims to use the potential of surface plasmon resonance (SPR) method to detect, and

qualify, specifically and without labeling, the EVs in their complex media, via their

(immune)capture onto the biochip-surface using different specific ligands.

Using two calibration standards of known size and concentration (Virus-Like Particles “VLP”

(50nm) & modified melamin resin MF beads (920nm)), we have shown that the capture level of

these entities by their specific ligands is linearly related to their concentration in the sample.

Dynamic ranges of concentrations (8x10

10

to 2x10

12

VLPs/ml and 3,25x10

7

to 3,25x10

8

beads/ml) have been established for 2 min injection time. These concentration ranges match the

typical range of EVs natural concentrations in plasma

3,4

. Additionally, by varying the injection

time, the dynamic range increases, which provides more sensibility for the detection of MPs in a

wide concentration range.

Moreover, a combination between SPR and atomic force microscopy (AFM) approaches appears

to be suitable for the metrological analysis of captured EVs, which allows identification of their

size subpopulations. Furthermore, an on-chip proteomic study was also engaged to discover

specific proteomic profiling of EVs in different conditions.

At last, we have adjusted this analytical platform based on a gold biochip to qualify and compare

resting and activated platelet MPs.