CD36 is a scavenger receptor known to play a critical role in the development of atherosclerosis by mediating the uptake of oxidized low-density lipoproteins (oxLDL) by macrophages. thus leading to foam cell formation. It is now generally recognized that the immune system has a pivotal role in the pathogenesis of atherosclerosis, whose progression is determined by ongoing inflammatory reactions. Recently, several studies pointed out that opioid peptides exert anti-inflammatory activities. Therefore the aim of the present study was to evaluate a possible endomorphin-1 (EM-1) immunomodulatory activity on human foam cells. Our results showed that EM-1 reduced Nile Red-stained lipid droplets content, decreased the expression of CD36 receptor and modulated tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) release from lipid-laden macrophages. Furthermore, Naloxone, an opioid receptors antagonist, reverted the anti-atherogenic and anti-inflammatory observed effects of EM-1. These data demonstrated, for the first time, an unprecedented ability of EM-1 to act as a novel modulator for macrophage-to-foam cell transformation, and for inflammatory cytokines profile, suggesting possible novel endomorphin-based anti-atherosclerotic approaches for the prevention and treatment of atherosclerosis. (C) 2010 Elsevier Inc. All rights reserved.

Chiurchiù, V., Izzi, V., D'Aquilio, F., Vismara, D., Carotenuto, F., Catanzaro, G., et al. (2011). Endomorphin-1 prevents lipid accumulation via CD36 down-regulation and modulates cytokines release from human lipid-laden macrophages. PEPTIDES, 32(1), 80-85 [10.1016/j.peptides.2010.09.024].

Endomorphin-1 prevents lipid accumulation via CD36 down-regulation and modulates cytokines release from human lipid-laden macrophages

Carotenuto F;Maccarrone M
2011-01-01

Abstract

CD36 is a scavenger receptor known to play a critical role in the development of atherosclerosis by mediating the uptake of oxidized low-density lipoproteins (oxLDL) by macrophages. thus leading to foam cell formation. It is now generally recognized that the immune system has a pivotal role in the pathogenesis of atherosclerosis, whose progression is determined by ongoing inflammatory reactions. Recently, several studies pointed out that opioid peptides exert anti-inflammatory activities. Therefore the aim of the present study was to evaluate a possible endomorphin-1 (EM-1) immunomodulatory activity on human foam cells. Our results showed that EM-1 reduced Nile Red-stained lipid droplets content, decreased the expression of CD36 receptor and modulated tumor necrosis factor-alpha (TNF-alpha) and interferon-gamma (IFN-gamma) release from lipid-laden macrophages. Furthermore, Naloxone, an opioid receptors antagonist, reverted the anti-atherogenic and anti-inflammatory observed effects of EM-1. These data demonstrated, for the first time, an unprecedented ability of EM-1 to act as a novel modulator for macrophage-to-foam cell transformation, and for inflammatory cytokines profile, suggesting possible novel endomorphin-based anti-atherosclerotic approaches for the prevention and treatment of atherosclerosis. (C) 2010 Elsevier Inc. All rights reserved.
2011
Pubblicato
Rilevanza internazionale
Articolo
Esperti anonimi
Settore MED/46 - SCIENZE TECNICHE DI MEDICINA E DI LABORATORIO
Settore BIO/13 - BIOLOGIA APPLICATA
Settore BIO/10 - BIOCHIMICA
English
Foam cells
Opioid peptides
Scavenger receptors
Atherosclerosis
Oxidized lipoproteins
CD36 Antigens
Cells, Cultured
Cytokines
Foam Cells
Humans
Lipoproteins, LDL
Microscopy, Confocal
Oligopeptides
Chiurchiù, V., Izzi, V., D'Aquilio, F., Vismara, D., Carotenuto, F., Catanzaro, G., et al. (2011). Endomorphin-1 prevents lipid accumulation via CD36 down-regulation and modulates cytokines release from human lipid-laden macrophages. PEPTIDES, 32(1), 80-85 [10.1016/j.peptides.2010.09.024].
Chiurchiù, V; Izzi, V; D'Aquilio, F; Vismara, D; Carotenuto, F; Catanzaro, G; Maccarrone, M
Articolo su rivista
File in questo prodotto:
Non ci sono file associati a questo prodotto.

I documenti in IRIS sono protetti da copyright e tutti i diritti sono riservati, salvo diversa indicazione.

Utilizza questo identificativo per citare o creare un link a questo documento: https://hdl.handle.net/2108/283374
Citazioni
  • ???jsp.display-item.citation.pmc??? 7
  • Scopus 11
  • ???jsp.display-item.citation.isi??? 10
social impact