Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 12(1): 12031, 2022 07 14.
Article in English | MEDLINE | ID: mdl-35835907

ABSTRACT

The regression, or resolution, of inflammation in atherosclerotic plaques is impaired in diabetes. However, the factors mediating this effect remain incomplete. We identified protein arginine methyltransferase 2 (PRMT2) as a protein whose expression in macrophages is reduced in hyperglycemia and diabetes. PRMT2 catalyzes arginine methylation to target proteins to modulate gene expression. Because PRMT2 expression is reduced in cells in hyperglycemia, we wanted to determine whether PRMT2 plays a causal role in the impairment of atherosclerosis regression in diabetes. We, therefore, examined the consequence of deleting PRMT2 in myeloid cells during the regression of atherosclerosis in normal and diabetic mice. Remarkably, we found significant impairment of atherosclerosis regression under normoglycemic conditions in mice lacking PRMT2 (Prmt2-/-) in myeloid cells that mimic the decrease in regression of atherosclerosis in WT mice under diabetic conditions. This was associated with increased plaque macrophage retention, as well as increased apoptosis and necrosis. PRMT2-deficient plaque CD68+ cells under normoglycemic conditions showed increased expression of genes involved in cytokine signaling and inflammation compared to WT cells. Consistently, Prmt2-/- bone marrow-derived macrophages (BMDMs) showed an increased response of proinflammatory genes to LPS and a decreased response of inflammation resolving genes to IL-4. This increased response to LPS in Prmt2-/- BMDMs occurs via enhanced NF-kappa B activity. Thus, the loss of PRMT2 is causally linked to impaired atherosclerosis regression via a heightened inflammatory response in macrophages. That PRMT2 expression was lower in myeloid cells in plaques from human subjects with diabetes supports the relevance of our findings to human atherosclerosis.


Subject(s)
Atherosclerosis , Diabetes Mellitus, Experimental , Hyperglycemia , Plaque, Atherosclerotic , Animals , Atherosclerosis/metabolism , Diabetes Mellitus, Experimental/complications , Diabetes Mellitus, Experimental/genetics , Humans , Hyperglycemia/complications , Inflammation/complications , Inflammation/genetics , Intracellular Signaling Peptides and Proteins , Lipopolysaccharides , Mice , Myeloid Cells/metabolism , Plaque, Atherosclerotic/complications , Plaque, Atherosclerotic/genetics , Protein-Arginine N-Methyltransferases/genetics
2.
Anal Chem ; 93(29): 10090-10098, 2021 07 27.
Article in English | MEDLINE | ID: mdl-34269045

ABSTRACT

Peroxynitrite, a transient reactive oxygen species (ROS), is believed to play a deleterious role in physiological processes. Herein, we report a two-photon ratiometric fluorescent probe that selectively reacts with peroxynitrite yielding a >200-fold change upon reaction. The probe effectively visualized fluctuations in peroxynitrite generation by arginase 1 in vivo and in vitro. This provides evidence that arginase 1 is a critical regulator of peroxynitrite.


Subject(s)
Fluorescent Dyes , Peroxynitrous Acid , Arginase , Photons , Reactive Oxygen Species
3.
Redox Biol ; 17: 400-410, 2018 07.
Article in English | MEDLINE | ID: mdl-29879549

ABSTRACT

Autophagy has been shown to be stimulated in advanced atherosclerotic plaques by metabolic stress, inflammation and oxidized lipids. The lack of published studies addressing the potential stimulation of pro-survival autophagy by oxysterols, a family of cholesterol oxidation products, has prompted our study. Thus, the goal of the current study is to elucidate the molecular mechanism of the autophagy induced by 27-hydroxycholesterol (27-OH), that is one of the most abundant oxysterols in advanced atherosclerotic lesions, and to assess whether the pro-oxidant effect of the oxysterol is involved in the given response. Here we showed that 27-OH, in a low micromolar range, activates a pro-survival autophagic response in terms of increased LC3 II/LC3 I ratio and Beclin 1, that depends on the up-regulation of extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt pathways as a potential result of an intracellular reactive oxygen species increase provoked by the oxysterol in human promonocytic U937 cells. Moreover, 27-OH induced autophagy is dependent on the relation between nuclear factor erythroid 2 p45-related factor 2 (Nrf2)-dependent antioxidant response and p62. The data obtained highlight the involvement of cholesterol oxidation products in the pathogenesis of oxidative stress related chronic diseases like atherosclerosis. Therefore, deeply understanding the complex mechanism and generating synthetic or natural molecules targeting this survival mechanism might be very promising tools in the prevention of such diseases.


Subject(s)
Autophagy/drug effects , Cholesterol/metabolism , Hydroxycholesterols/pharmacology , NF-E2-Related Factor 2/genetics , RNA-Binding Proteins/genetics , Antioxidants/pharmacology , Apoptosis/drug effects , Atherosclerosis/metabolism , Atherosclerosis/pathology , Atherosclerosis/prevention & control , Autophagy/genetics , Cell Survival/drug effects , Humans , Monocyte-Macrophage Precursor Cells/drug effects , Monocyte-Macrophage Precursor Cells/metabolism , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , RNA-Binding Proteins/metabolism , Reactive Oxygen Species/metabolism
4.
Mol Aspects Med ; 49: 8-22, 2016 06.
Article in English | MEDLINE | ID: mdl-27017897

ABSTRACT

Oxysterols, a family of oxidation products of cholesterol, are increasingly drawing attention of scientists to their multifaceted biochemical properties, several of them of clear relevance to human pathophysiology. Taken up by cells through both vesicular and non-vesicular ways or often generated intracellularly, oxysterols contribute to modulate not only the inflammatory and immunological response but also cell viability, metabolism and function by modulating several signaling pathways. Moreover, they have been recognized as elective ligands for the most important nuclear receptors. The outcome of such a complex network of intracellular reactions promoted by these cholesterol oxidation products appears to be largely dependent not only on the type of cells, the dynamic conditions of the cellular and tissue environment but also on the concentration of the oxysterols. Here focus has been given to the cascade of molecular events exerted by relatively low concentrations of certain oxysterols that elicit survival and functional signals in the cells, with the aim to contribute to further expand the knowledge about the biological and physiological potential of the biochemical reactions triggered and modulated by oxysterols.


Subject(s)
Oxidation-Reduction , Oxysterols/metabolism , Signal Transduction , Animals , Autophagy , Cell Survival , Gene Expression Regulation , Humans , Lipid Metabolism , NF-E2-Related Factor 2/metabolism , Oxysterols/chemistry , Stress, Physiological , Transcription Factors/metabolism , Transcription, Genetic
5.
Free Radic Biol Med ; 91: 93-104, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26689473

ABSTRACT

Cholesterol oxidation products such as oxysterols are considered critical factors in the atherosclerotic plaque formation since they induce oxidative stress, inflammation and apoptotic cell death. 27-hydroxycholesterol (27-OH) is one of the most represented oxysterols in atherosclerotic lesions. We recently showed that relatively low concentrations of 27-OH generated a strong survival signaling through an early and transient increase of cellular ROS level, that enhanced MEK-ERK/PI3K-Akt phosphorylation, in turn responsible of a sustained quenching of ROS production. It remains to identify the link between ERK/Akt up-regulation and the consequent quenching effect on ROS intracellular level that efficiently and markedly delay the pro-apoptotic effect of the oxysterol. Here we report on the potent activation of Nrf2 redox-sensitive transcription factor by low micromolar amount of 27-OH added to U937 promonocytic cells. The 27-OH-exerted induction of Nrf2 and subsequently of the target genes, HO-1 and NQO-1, was proved to be: (i) dependent upon the activation of ERK and Akt pathways, (ii) directly responsible for the quenching of intracellular oxidative stress and by this way (iii) ultimately responsible for the observed oxysterol-induced pro-survival response.


Subject(s)
Hydroxycholesterols/pharmacology , Monocyte-Macrophage Precursor Cells/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus , Apoptosis , Cell Line , Cell Survival , Enzyme Induction/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Humans , MAP Kinase Signaling System , Monocyte-Macrophage Precursor Cells/drug effects , NAD(P)H Dehydrogenase (Quinone)/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species
6.
Free Radic Biol Med ; 77: 376-85, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25110320

ABSTRACT

The oxysterol 27-hydroxycholesterol (27-OH) is increasingly considered to be involved in a variety of pathophysiological processes, having been shown to modulate cell proliferation and metabolism, and also to exert proinflammatory and proapoptotic effects. This study aimed to elucidate the molecular pathways whereby 27-OH may generate survival signals in cells of the macrophage lineage, and to clarify whether its known prooxidant effect is involved in that process. A net up-regulation of survival signaling, involving the extracellular signal-regulated kinase (ERK) and phosphoinositide 3-kinase (PI3K)/Akt phosphorylation pathways, was observed in U937 promonocytic cells cultivated over time in the presence of a low micromolar concentration of the oxysterol. Interestingly, the up-regulation of both kinases was shown to be closely dependent on an early 27-OH-induced intracellular increase of reactive oxygen species (ROS). In turn, stimulation of ERK and PI3K/Akt both significantly quenched ROS steady state and markedly phosphorylated Bad, thereby determining a marked delay of the oxysterol׳s proapoptotic action. The 27-OH-induced survival pathways thus appear to be redox modulated and, if they occur within or nearby inflammatory cells during progression of chronic diseases such as cancer and atherosclerosis, they could significantly impact the growth and evolution of such diseases.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Hydroxycholesterols/pharmacology , Protein Processing, Post-Translational , Proto-Oncogene Proteins c-akt/metabolism , Apoptosis , Cell Line, Tumor , Cell Survival , Humans , Membrane Potential, Mitochondrial , Oxidation-Reduction , Phosphorylation , Reactive Oxygen Species/metabolism , Signal Transduction
7.
Free Radic Biol Med ; 52(1): 7-18, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-22019631

ABSTRACT

Tumor suppressor genes regulate diverse cellular activities including DNA damage repair, cell cycle arrest, mitogenic signaling, cell differentiation, migration, and programmed cell death. In this review the tumor suppressor genes p53, FoxO, retinoblastoma (RB), p21, p16, and breast cancer susceptibility genes 1 and 2 (BRCA1 and BRCA2) and their roles in oxidative stress are summarized with a focus on the links and interplay between their pathways and reactive oxygen species (ROS). The results of a number of studies have demonstrated an antioxidant role for tumor suppressor proteins, activating the expression of some well-known antioxidant genes in response to oxidative stress. On the other hand, recent studies have revealed a pro-oxidant role for p53 by which cellular ROS are increased by enhanced transcription of proapoptotic genes. A tightly regulated feedback loop between ROS and FoxO proteins, with ROS regulating FoxO activity through posttranslational modifications and protein interactions and FoxO controlling intracellular ROS levels, has been demonstrated. Furthermore, these studies have shown that FoxO transcription factors and p38 mitogen-activated protein kinases may interact with the RB pathway under stress conditions. In addition, cellular senescence studies established an unexpected role for ROS in inducing and maintaining senescence-induced tumor suppression that blocks cytokinesis to ensure senescent cells never divide again. p21 and p16 have been shown to act as tumor suppressor proteins and this function extends beyond cell cycle control and includes important roles in regulating oxidative stress. Consequently, these important interactions indicate a critical potential role for tumor suppressor genes in the cellular response against oxidative stress and emphasize links between ROS and tumor suppressor genes that might be therapeutic targets in oxidative damage-associated diseases.


Subject(s)
Antioxidants/metabolism , Feedback, Physiological/physiology , Forkhead Transcription Factors/metabolism , Oxidative Stress , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Tumor Suppressor Proteins/metabolism , Animals , Cell Communication , Cell Cycle Checkpoints , Cellular Senescence , DNA Repair , Forkhead Transcription Factors/genetics , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , Oxidation-Reduction , Tumor Suppressor Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...