scholarly journals Simultaneous NAD(P)H and FAD fluorescence lifetime microscopy of long UVA–induced metabolic stress in reconstructed human skin

2021 ◽  
Vol 11 (1) ◽  
Author(s):  
Thi Phuong Lien Ung ◽  
Seongbin Lim ◽  
Xavier Solinas ◽  
Pierre Mahou ◽  
Anatole Chessel ◽  
...  

AbstractSolar ultraviolet longwave UVA1 exposure of human skin has short-term consequences at cellular and molecular level, leading at long-term to photoaging. Following exposure, reactive oxygen species (ROS) are generated, inducing oxidative stress that might impair cellular metabolic activity. However, the dynamic of UVA1 impact on cellular metabolism remains unknown because of lacking adequate live imaging techniques. Here we assess the UVA1-induced metabolic stress response in reconstructed human skin with multicolor two-photon fluorescence lifetime microscopy (FLIM). Simultaneous imaging of nicotinamide adenine dinucleotide (NAD(P)H) and flavin adenine dinucleotide (FAD) by wavelength mixing allows quantifying cellular metabolism in function of NAD(P)+/NAD(P)H and FAD/FADH2 redox ratios. After UVA1 exposure, we observe an increase of fraction of bound NAD(P)H and decrease of fraction of bound FAD indicating a metabolic switch from glycolysis to oxidative phosphorylation or oxidative stress possibly correlated to ROS generation. NAD(P)H and FAD biomarkers have unique temporal dynamic and sensitivity to skin cell types and UVA1 dose. While the FAD biomarker is UVA1 dose-dependent in keratinocytes, the NAD(P)H biomarker shows no dose dependence in keratinocytes, but is directly affected after exposure in fibroblasts, thus reflecting different skin cells sensitivities to oxidative stress. Finally, we show that a sunscreen including a UVA1 filter prevents UVA1 metabolic stress response from occurring.

Author(s):  
Divya Lodha ◽  
Jamuna R. Subramaniam

Abstract Objectives The main aim of this study is to identify the deleterious effects of indiscriminately consumed high fructose on motor neurons that are critically affected in many neurological conditions causing movement disorders including paralysis. Materials and Methods Neuroblastoma x mouse spinal cord motor neuron cell line (NSC-34) motor neuron cell lines were treated with high fructose and oxygen supplementation (18.8%) and assayed for cell proliferation/death, reactive oxygen species (ROS) generation, and oxidative stress response induction Statistical Analysis Mean and standard deviation, significance with and without high fructose (F)-5%, were estimated by t-tests using GraphPad Prism ver. 8.2.1 Results F-5% along with O2 (18.8%) annihilates the cells (∼85%) by day10 and inhibits cell division as observed by the presence of multinucleated cells. Unexpectedly, 1 to 2% of cells that survived, differentiated and displayed progressive neurite extension. Though not healthy, they were viable up to 80 days. F-5% increased ROS levels (∼34%) not accompanied by concomitant enhanced expression of oxidative stress response regulator, the transcription factor, nrf-2, or downstream effector, sod-1. Conclusion High fructose is extremely harmful to NSC-34 motor neuron cell line.


2021 ◽  
Vol 2021 ◽  
pp. 1-25
Author(s):  
Ting Hong ◽  
Yiyan Chen ◽  
Xiaoying Li ◽  
Yan Lu

The overproduction of reactive oxygen species (ROS) and consequent oxidative stress contribute to the pathogenesis of acute and chronic liver diseases. It is now acknowledged that nonalcoholic fatty liver disease (NAFLD) is characterized as a redox-centered disease due to the role of ROS in hepatic metabolism. However, the underlying mechanisms accounting for these alternations are not completely understood. Several nuclear receptors (NRs) are dysregulated in NAFLD, and have a direct influence on the expression of a set of genes relating to the progress of hepatic lipid homeostasis and ROS generation. Meanwhile, the NRs act as redox sensors in response to metabolic stress. Therefore, targeting NRs may represent a promising strategy for improving oxidation damage and treating NAFLD. This review summarizes the link between impaired lipid metabolism and oxidative stress and highlights some NRs involved in regulating oxidant/antioxidant turnover in the context of NAFLD, shedding light on potential therapies based on NR-mediated modulation of ROS generation and lipid accumulation.


2009 ◽  
Vol 296 (4) ◽  
pp. H946-H956 ◽  
Author(s):  
Nazar Labinskyy ◽  
Partha Mukhopadhyay ◽  
Janos Toth ◽  
Gabor Szalai ◽  
Monika Veres ◽  
...  

Vascular aging is characterized by increased oxidative stress and proinflammatory phenotypic alterations. Metabolic stress, such as hyperglycemia in diabetes, is known to increase the production of ROS and promote inflammatory gene expression, accelerating vascular aging. The oxidative stress hypothesis of aging predicts that vascular cells of long-lived species exhibit lower steady-state production of ROS and/or superior resistance to the prooxidant effects of metabolic stress. We tested this hypothesis using two taxonomically related rodents, the white-footed mouse ( Peromyscus leucopus) and the house mouse ( Mus musculus), which show a more than twofold difference in maximum lifespan potential (8.2 and 3.5 yr, respectively). We compared interspecies differences in steady-state and high glucose (HG; 30 mmol/l)-induced production of O2•− and H2O2, endothelial function, mitochondrial ROS generation, and inflammatory gene expression in cultured aortic segments. In P. leucopus aortas, steady-state endothelial O2•− and H2O2 production and ROS generation by mitochondria were less than in M. musculus vessels. Furthermore, vessels of P. leucopus were more resistant to the prooxidant effects of HG. Primary fibroblasts from P. leucopus also exhibited less steady-state and HG-induced ROS production than M. musculus cells. In M. musculus arteries, HG elicited significant upregulation of inflammatory markers (TNF-α, IL-6, ICAM-1, VCAM, and monocyte chemoattractant protein-1). In contrast, the proinflammatory effects of HG were blunted in P. leucopus vessels. Thus, increased life span potential in P. leucopus is associated with decreased cellular ROS generation and increased resistance to prooxidant and proinflammatory effects of metabolic stress, which accord with predictions of the oxidative stress hypothesis of aging.


Blood ◽  
2019 ◽  
Vol 134 (Supplement_1) ◽  
pp. 1201-1201
Author(s):  
Vinothini Govindarajah ◽  
Jung-Mi Lee ◽  
Michael Solomon ◽  
Bryan Goddard ◽  
Ramesh C. Nayak ◽  
...  

Lifelong maintenance of the blood system requires the preservation of a healthy hematopoietic stem cell (HSC) pool. Integrity of the HSC compartment is disrupted by severe homeostatic perturbations following acute infection or irradiation, which result in HSC loss and lead to dramatic hematological dysfunctions. HSC fitness is also affected by organismal patho-physiological conditions, which are associated with chronic low-grade stress. These chronic conditions present, over time, a unique challenge to the maintenance of the long-lived HSCs. However, the mechanisms by which the HSC pool adapts to chronic, low-grade stress conditions remain largely unknown. We focus on obesity as a model of chronic stress that potentially affects the health of the HSC compartment. Obesity is a chronic pathological state, which is established and persists over long period of time and therefore, is a prime candidate to alter long-lived HSCs. Obesity becomes progressively associated with an array of systemic and local alterations that include metabolic dysregulations, unresolved low-grade inflammation andalteration of BM microenvironment, all conditions that have been linked to HSC defects (Nagareddy et al, Cell Metabol., 2013; Naveiras et al, Nature, 2009; Luo et al, Cell Metabol., 2015; Ambrosi et al, Cell Stem Cell, 2017).Consistent with these studies, we recently showed that obesity alters the composition of the HSC-SLAM (Lin- ckit+ Sca1+ CD48+ CD150-) compartment and leads to a progressive loss of HSC fitness upon serial competitive transplantation assays (Lee et al, J. Exp. Med.,2018). Mechanistically, we established that the oxidative stress induced by obesity dysregulates the expression of the transcription factor Gfi1 and contributes to the long-term alteration of the HSC functions. Following this work, we confirmed that obesity mainly affects the short and long-term stress response of the most primitive HSC compartment (defined as HSC-SLAM CD34- CD49b-) but not of the downstream short-term HSC subsets, therefore indicating that obesitydisrupts intrinsic mechanisms regulating self-renewal activity. Genome-wide gene expression analyses indicated that HSCs isolated from obese mice dysregulate multiples genes involved in the phosphatidylinositol signaling pathway (e.g. Pik3c2a, Pik3c2b, Pi3kap1, Pi3kip1), upstream of the Akt signaling molecule. Phospho-flow cytometry analyses showed that HSCs freshly isolated from obese mice display constitutive Akt activation. In vivopharmacological Akt inhibition did not significantly change the size or phenotype of the HSC-SLAM compartment in obesity but led to the normalization of their functions in condition of transplantation-induced regenerative stress. These results indicate that the aberrant Akt activation in obesity-primed HSCs directly contributes to their hyperactivity upon transplantation, which ultimately results in the erosion of their self-renewal potential. We next explored the mechanisms that allow HSCs to remain quiescent in obesity despite the constitutive activation of Akt and the continued presence of oxidative stress condition. We focused on FoxO3, which is a key transcriptional regulator of HSC quiescence and direct Akt target. We found that FoxO3 in obesity-primed HSCs become insensitive to its normal upstream regulators such as Akt and p38, leading to the maintenance of its nuclear location in oxidative stress condition. As consequence, we observed that obesity-primed HSCs were more resistant to oxidative stress than their normal counterparts, suggesting an increase protection against ROS-mediated senescence and apoptosis. Altogether, these results suggest that chronic metabolic stresses associated with obesity progressively affect the wiring of the HSCs and modify their oxidative stress response. The uncoupling of FoxO3 to its environmental regulators in obesity could be viewed as a key adaptive strategy to ensure the survival and function of the HSC compartment in condition of chronic metabolic stress. More broadly, these results highlight how patho-physiological conditions associated with chronic low-grade stress (such as obesity) could shape the characteristic of the HSC compartment at steady state. Progressive alteration of the normal HSC stress response in such conditions could be an unrecognized contributing factor for the development of hematological diseases. Disclosures Cancelas: Cerus Co.: Research Funding; TerumoBCT: Consultancy, Research Funding; Macopharma Inc: Research Funding; Cytosorbents: Research Funding; Cellphire: Research Funding; Velico: Consultancy, Research Funding; Fresenius-Kabi: Research Funding; Hemanext: Consultancy, Research Funding.


2020 ◽  
Vol 140 (7) ◽  
pp. S36
Author(s):  
K. Marengo ◽  
J. Oldach ◽  
A. Armento ◽  
A. Langerveld ◽  
E. Lehigh ◽  
...  

2017 ◽  
Vol 2017 ◽  
pp. 1-10 ◽  
Author(s):  
Mariola Herbet ◽  
Agnieszka Korga ◽  
Monika Gawrońska-Grzywacz ◽  
Magdalena Izdebska ◽  
Iwona Piątkowska-Chmiel ◽  
...  

Chronic environmental stress is associated with reactive oxygen species (ROS) overproduction and the pathogenesis of depression. The purpose of this study was to evaluate biochemical and molecular changes associated with ROS generation in the brains of rats submitted to chronic variable stress. Male Wistar rats (50–55 days old, weighing 200–250 g) were divided in two groups (n=10): control and stressed. Rats in the stressed group were exposed to stress conditions for 40 days. The animals were decapitated and the brain samples were collected. In prefrontal cortex, we measured the following biochemical parameters: lipid peroxidation and concentration of glutathione—GSH, GSSG, GSH/GSSG ratio, glutathione peroxidase, and glutathione reductase activities. In the hippocampus marker of DNA, oxidative damage and expression of DNA-repairing genes (Ogg1,MsrA) and gene-encoding antioxidative transcriptional factor (Nrf2) were determined. The results demonstrate indirect evidence of ROS overproduction and presence of oxidative stress. They also reveal disruption of oxidative defense systems (decreased GR activity, diminished GSH/GSSG ratio, and decreasedNrf2expression) and activation of the oxidative DNA repair system (increasedOgg1andMsrAexpression). Together, the presented data suggest that independent activation of oxidative stress response genes occurs in chronic variable stress conditions.


Sign in / Sign up

Export Citation Format

Share Document