L-arginine-nitric oxide metabolism. Glutamine: A new player in this metabolic game?

1998 ◽  
Vol 17 (1) ◽  
pp. 1-2 ◽  
Author(s):  
Erich Roth
2020 ◽  
Vol 26 (10) ◽  
pp. S17-S18
Author(s):  
Adil Yunis ◽  
Mehdi Nouraie ◽  
Noel Britton ◽  
Yingze Zhang ◽  
Nicole Helbling ◽  
...  

2006 ◽  
Vol 41 (7) ◽  
pp. 820-825 ◽  
Author(s):  
Eric A. G. Demoncheaux ◽  
David A. Elphick ◽  
Marc B. Dürner ◽  
Gail E. Higgins ◽  
David Crowther ◽  
...  

Life Sciences ◽  
2001 ◽  
Vol 68 (10) ◽  
pp. 1169-1179 ◽  
Author(s):  
Mercedes Ferrer ◽  
M Jesús Alonso ◽  
Mercedes Salaices ◽  
Jesús Marı́n ◽  
Gloria Balfagón

BIOCELL ◽  
2016 ◽  
Vol 40 (1) ◽  
pp. 55-58
Author(s):  
Tamara ZAOBORNYJ ◽  
Dar韔 E. IGLESIAS ◽  
Silvina S. BOMBICINO ◽  
Alberto BOVERIS ◽  
Laura B. VALDEZ

1997 ◽  
Vol 341 (1) ◽  
pp. 186-192 ◽  
Author(s):  
Y. Minamiyama ◽  
S. Takemura ◽  
M. Inoue

2004 ◽  
Vol 186 (23) ◽  
pp. 7980-7987 ◽  
Author(s):  
Andrea Büsch ◽  
Anne Pohlmann ◽  
Bärbel Friedrich ◽  
Rainer Cramm

ABSTRACT The σ54-dependent regulator NorR activates transcription of target genes in response to nitric oxide (NO) or NO-generating agents. In Ralstonia eutropha H16, NorR activates transcription of the dicistronic norAB operon that encodes NorA, a protein of unknown function, and NorB, a nitric oxide reductase. A constitutively activating NorR derivative (NorR′), in which the N-terminal signaling domain was replaced by MalE, specifically bound to the norAB upstream region as revealed by gel retardation analysis. Within a 73-bp DNA segment protected by MalE-NorR′ in a DNase I footprint assay, three conserved inverted repeats, GGT-(N7)-ACC (where N is any base), that we consider to be NorR-binding boxes were identified. Mutations altering the spacing or the base sequence of these repeats resulted in an 80 to 90% decrease of transcriptional activation by wild-type NorR. Genome database analyses demonstrate that the GT-(N7)-AC core of the inverted repeat is found in several proteobacteria upstream of gene loci encoding proteins of nitric oxide metabolism, including nitric oxide reductase (NorB), flavorubredoxin (NorV), NO dioxygenase (Hmp), and hybrid cluster protein (Hcp).


2018 ◽  
Vol 647 ◽  
pp. 47-53 ◽  
Author(s):  
Valeria Calabró ◽  
María C. Litterio ◽  
Cesar G. Fraga ◽  
Monica Galleano ◽  
Barbara Piotrkowski

Circulation ◽  
2014 ◽  
Vol 130 (suppl_2) ◽  
Author(s):  
Xiaoping Liu ◽  
Mohamed A El-Mahdy ◽  
Raed S Ismail ◽  
Sean Little ◽  
Le T Thuy ◽  
...  

Cytoglobin (Cygb) can effectively metabolize nitric oxide (NO), a potent vasodilator, in the presence of oxygen and reductants. Cygb in the vascular wall may affect cardiovascular functions by changing the rate of NO metabolism. In this study, we directly tested the vascular role of Cygb using Cygb knockout (Cygb-/-) mice. The mean blood pressure of Cygb-/- and C57BL/6 wild type (WT) mice was 65.3 ± 1.9 mmHg and 93.7 ± 1.5 mmHg, respectively (n=10). Using echocardiography, we observed that cardiac output (CO) was increased in Cygb-/- mice compared to WT with values of 29.8 ± 3.9 vs 17.7 ± 0.9 ml/min. The systemic vascular resistance (SVR) of Cygb-/- mice was decreased by ~60% vs that of WT mice (Fig. 1). Further, the inner diameter (id) of aorta of Cygb-/- mice was dilated compared to WT with values of 2.2 ± 0.1 mm vs 1.5 ± 0.05 mm (n=5), respectively. After treatment with the NO synthase inhibitor L-NAME, no difference in the aortic id remained between Cygb-/- (1.55 ± 0.03 mm) and WT (1.49 ± 0.02 mm) mice, indicating that the NO pathway is responsible for the difference in vascular inner diameters and tone. Myograph experiments show that the aortic vasodilation response of Cygb-/- mice is much more sensitive to acetylcholine (Ach) or the NO donor nitroprusside (SNP) (EC50 shifts from 13 nM and 2.9 nM (WT mice) to 0.33 nM and 0.16 nM (Cygb-/-) for Ach and SNP, respectively). Using NO electrodes to measure the rate of NO consumption by SMCs and quantitative imunoblotting to estimate Cygb content in RSMCs-AR and Cygb knockdown RSMCs, we observed that 90% of NO consumption by RSMCs-AR is caused by the intracellular Cygb. Our results indicate that Cygb deficiency in the vascular wall of Cygb-/- mice greatly reduces the rate of NO metabolism and increases vascular NO concentration, resulting in vasodilation, increase in vessel lumen diameter, and decrease in SVR. These results demonstrate that Cygb regulates cardiac function and vessel tone by controlling the rate of vascular NO metabolism.


Author(s):  
Ana Jiménez ◽  
Mª Carmen Martí ◽  
Daymi Camejo ◽  
Francisca Sevilla

Sign in / Sign up

Export Citation Format

Share Document