Egyszerű nézet

dc.contributor.author Di Pietro, N
dc.contributor.author Giardinelli, A
dc.contributor.author Sirolli, V
dc.contributor.author Riganti, C
dc.contributor.author Di Tomo, P
dc.contributor.author Gazzano, E
dc.contributor.author Di Silvestre, S
dc.contributor.author Panknin, C
dc.contributor.author Cortese-Krott, MM
dc.contributor.author Csonka, Csaba
dc.contributor.author Kelm, M
dc.contributor.author Ferdinandy, Péter
dc.contributor.author Bonomini, M
dc.contributor.author Pandolfi, A
dc.date.accessioned 2021-12-11T14:17:27Z
dc.date.available 2021-12-11T14:17:27Z
dc.date.issued 2016
dc.identifier 84969900538
dc.identifier.citation journalVolume=417;journalIssueNumber=1-2;journalTitle=MOLECULAR AND CELLULAR BIOCHEMISTRY;pagerange=155-167;journalAbbreviatedTitle=MOL CELL BIOCHEM;
dc.identifier.uri http://repo.lib.semmelweis.hu//handle/123456789/8076
dc.identifier.uri doi:10.1007/s11010-016-2723-0
dc.description.abstract Red blood cells (RBCs) enzymatically produce nitric oxide (NO) by a functional RBC-nitric oxide synthase (RBC-NOS). NO is a vascular key regulatory molecule. In RBCs its generation is complex and influenced by several factors, including insulin, acetylcholine, and calcium. NO availability is reduced in end-stage renal disease (ESRD) and associated with endothelial dysfunction. We previously demonstrated that, through increased phosphatidylserine membrane exposure, ESRD-RBCs augmented their adhesion to human cultured endothelium, in which NO bioavailability decreased. Since RBC-NOS-dependent NO production in ESRD is unknown, this study aimed to investigate RBC-NOS levels/activation, NO production/bioavailability in RBCs from healthy control subjects (C, N = 18) and ESRD patients (N = 27). Although RBC-NOS expression was lower in ESRD-RBCs, NO, cyclic guanosine monophosphate (cGMP), RBC-NOS Serine1177 phosphorylation level and eNOS/Calmodulin (CaM)/Heat Shock Protein-90 (HSP90) interaction levels were higher in ESRD-RBCs, indicating increased enzyme activation. Conversely, following RBCs stimulation with insulin or ionomycin, NO and cGMP levels were significantly lower in ESRD- than in C-RBCs, suggesting that uremia might reduce the RBC-NOS response to further stimuli. Additionally, the activity of multidrug-resistance-associated protein-4 (MRP4; cGMP-membrane transporter) was significantly lower in ESRD-RBCs, suggesting a possible compromised efflux of cGMP across the ESRD-RBCs membrane. This study for the first time showed highest basal RBC-NOS activation in ESRD-RBCs, possibly to reduce the negative impact of decreased NOS expression. It is further conceivable that high NO production only partially affects cell function of ESRD-RBCs maybe because in vivo they are unable to respond to physiologic stimuli, such as calcium and/or insulin.
dc.format.extent 155-167
dc.relation.ispartof urn:issn:0300-8177; 1573-4919
dc.title Nitric oxide synthetic pathway and cGMP levels are altered in red blood cells from end-stage renal disease patients
dc.type Journal Article
dc.date.updated 2019-12-05T11:05:31Z
dc.language.rfc3066 en
dc.rights.holder NULL
dc.identifier.mtmt 3068545
dc.identifier.wos 000377593300015
dc.identifier.pubmed 27206740
dc.contributor.department SE/AOK/I/Farmakológiai és Farmakoterápiás Intézet
dc.contributor.institution Semmelweis Egyetem
dc.mtmt.swordnote Mario Bonomini, and Assunta Pandolfi have contributed equally to the study.


Kapcsolódó fájlok:

A fájl jelenleg csak egyetemi IP címről érhető el.

Megtekintés/Megnyitás

Ez a rekord az alábbi gyűjteményekben szerepel:

Egyszerű nézet