Effect of reactive oxygen species scavengers, antiinflammatory drugs, and calcium-channel blockers on carbon dioxide pneumoperitoneum-enhanced adhesions in a laparoscopic mouse model

2007 ◽  
Vol 21 (10) ◽  
pp. 1826-1834 ◽  
Author(s):  
M. M. Binda ◽  
C. R. Molinas ◽  
A. Bastidas ◽  
P. R. Koninckx
2021 ◽  
Vol 22 (3) ◽  
pp. 1106
Author(s):  
Rayan Bou-Fakhredin ◽  
Batoul Dia ◽  
Hilda E. Ghadieh ◽  
Stefano Rivella ◽  
Maria Domenica Cappellini ◽  
...  

Oxidative damage by reactive oxygen species (ROS) is one of the main contributors to cell injury and tissue damage in thalassemia patients. Recent studies suggest that ROS generation in non-transfusion-dependent (NTDT) patients occurs as a result of iron overload. Among the different sources of ROS, the nicotinamide adenine dinucleotide phosphate (NADPH) oxidase family of enzymes and cytochrome P450 (CYP450) have been proposed to be major contributors for oxidative stress in several diseases. However, the sources of ROS in patients with NTDT remain poorly understood. In this study, Hbbth3/+ mice, a mouse model for β-thalassemia, were used. These mice exhibit an unchanged or decreased expression of the major NOX isoforms, NOX1, NOX2 and NOX4, when compared to their C57BL/6 control littermates. However, a significant increase in the protein synthesis of CYP4A and CYP4F was observed in the Hbbth3/+ mice when compared to the C57BL/6 control mice. These changes were paralleled by an increased production of 20-hydroxyeicosatetraenoic acid (20-HETE), a CYP4A and CYP4F metabolite. Furthermore, these changes corroborate with onset of ROS production concomitant with liver injury. To our knowledge, this is the first report indicating that CYP450 4A and 4F-induced 20-HETE production mediates reactive oxygen species overgeneration in Hbbth3/+ mice through an NADPH-dependent pathway.


2015 ◽  
Vol 411 (1-2) ◽  
pp. 317-330 ◽  
Author(s):  
Sergey Bolevich ◽  
Alekandr Haritonovic Kogan ◽  
Vladimir Zivkovic ◽  
Dusan Djuric ◽  
Aleksey Aleksejevic Novikov ◽  
...  

2017 ◽  
Vol 13 ◽  
pp. 174480691771390 ◽  
Author(s):  
Jun-Ho La ◽  
Jigong Wang ◽  
Alice Bittar ◽  
Hyun Soo Shim ◽  
Chilman Bae ◽  
...  

Blood ◽  
2008 ◽  
Vol 112 (11) ◽  
pp. 3631-3631
Author(s):  
Yang Jo Chung ◽  
Peter D. Aplan

Abstract The myelodysplastic syndromes (MDSs) comprise a group of pre-malignant hematologic disorders characterized by ineffective hematopoiesis, dysplasia, and transformation to acute myeloid leukemia (AML). The causative agents for de novo or therapy-related MDS (t-MDS) include exposure to known genotoxins, such as benzene and other solvents, anti-cancer chemotherapy, and ionizing radiation. It is known that MDS is closely related to chromosomal aberrations including deletions, amplifications, inversions, and translocations, however, the molecular mechanism of disease progression from MDS to AML not been completely elucidated. Recently, several studies have reported that oxidative stress contributes to the disease progression mechanism in MDS. Reactive oxygen species (ROS) play a role in regulating several biologic phenomena involving activation of signaling pathways in response to cytokines, and the gene expression induced by this signaling. ROS is also known to induce oxidative DNA damage which generates DNA base modifications and DNA helix alterations. Therefore, increased intracellular levels of ROS can induce mutations which lead to transformation of MDS to AML. A mouse model for MDS, generated by expressing a NUP98-HOXD13 (NHD13) fusion gene was exploited to investigate ROS levels in this study. Lineage negative (Linneg) bone marrow mononuclear cell (BMNC) from NHD13 mice with MDS had a 7.8±5.6 fold increased level of ROS compared with wild-type (WT) Linneg BMNC, while ROS levels of unfractionated BMNC were similar in NHD13 and WT mice. The increase ROS level in NHD13 Linneg BMNC was associated with an increase in G2/M phase (2 fold). Interleukin 3 (IL-3) dependent NHD13 ‘knock-in’ cell lines also showed an increased level of ROS (3 to 5 fold), in comparison with other IL-3 dependent cell lines, BaF3 and 32D. To determine if the increase in ROS could be generated by an NHD13 fusion, BaF3 and 32D cell were transfected with an NHD13 expression vector. The BaF3 and 32D transfectant showed 2 fold (2.1±0.3 and 1.9±0.6 fold) increase ROS level compared to controls transfected with an empty vector. These findings suggest that expression of an NHD13 fusion gene can induce ROS in hematopoietic progenitor cells, which may contribute to the development of MDS and subsequent disease progression to AML through DNA damages.


Sign in / Sign up

Export Citation Format

Share Document