[go: up one dir, main page]

 
 

Topic Editors

Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
Department of Scienze della Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
Department of Scienzedella Vita e dell’Ambiente, Sezione di Scienze del Farmaco, University of Cagliari, Via Ospedale 72, 09124 Cagliari, Italy
Biomedical & Tissue Engineering Laboratory, Fundacion Investigacion Hospital General Universitario Valencia, Valencia, Spain
1. Departamento de Farmacia y Tecnología Farmacéutica y Parasitología, Facultad de Farmacia, Universitat de València, Av. Vicente Andrés Estellés s/n, Burjassot, 46100 Valencia, Spain
2. Instituto Interuniversitario de Investigación de Reconocimiento Molecular y Desarrollo Tecnológico (IDM), Universitat Politècnica de València, Universitat de València, 46100 Valencia, Spain

Oxidative Stress and Inflammation, 2nd Volume

Abstract submission deadline
closed (29 February 2024)
Manuscript submission deadline
closed (31 May 2024)
Viewed by
19375

Topic Information

Dear Colleagues,

Oxidative stress is viewed as an imbalance between the production of reactive oxygen species (ROS) and their elimination by protective mechanisms, which can lead to chronic inflammation. The incorrect lifestyle and the intake of external unhealthy chemicals are the major causes of age-related chronic diseases and cancer. Their study includes diseases pathology pathway, lifestyle, treatment, protection, and prevention of oxidative stress and inflammation. ROS are normally produced within the body in limited amount and are essential compounds involved in the regulation of processes capable of maintaining cell homeostasis and functions (signal transduction, gene expression, and activation of receptors). This imbalance may cause oxidative stress, which can lead to lipid peroxidation, gene mutation, inflammation and other complications. The harmful oxidative activity of ROS can be counteracted only by antioxidant/anti-inflammatory compounds, which may be both synthetic and natural, but are often characterized by several stability issues, such as poor water solubility and low bioavailability, that compromise their in vivo activities. The research papers of this Topic are looked forward to providing an overview of the status of research on both natural and synthetic products with antioxidant properties and capable of counteracting inflammatory diseases, including, but not necessarily restricted to, chemical compounds synthetic or of natural origin, vitamins, peptides, micronutrients, non-starch polysaccharides, probiotics, postbiotics, prebiotics formulations containing functional foods or nutraceutical or cosmeceutical products and their innovative oral, systemic or topical delivery capable to overcome their stability issues.

Dr. Maria Letizia Manca
Dr. Amparo Nacher
Dr. Mohamad Allaw
Dr. Matteo Perra
Dr. Ines Castangia
Topic Editors

Keywords

  • oxidative stress
  • inflammation
  • molecular mechanisms
  • antioxidant
  • cosmeceutics
  • nutraceutics
  • mitochondrial oxidative
  • ROS species
  • lifestyle
  • longevity
  • aging
  • drug delivery
  • skin delivery
  • oral delivery

Participating Journals

Journal Name Impact Factor CiteScore Launched Year First Decision (median) APC
Biomedicines
biomedicines
3.9 5.2 2013 15.3 Days CHF 2600
Cells
cells
5.1 9.9 2012 17.5 Days CHF 2700
International Journal of Molecular Sciences
ijms
4.9 8.1 2000 18.1 Days CHF 2900
Life
life
3.2 4.3 2011 18 Days CHF 2600
Oxygen
oxygen
- - 2021 23.1 Days CHF 1000

Preprints.org is a multidiscipline platform providing preprint service that is dedicated to sharing your research from the start and empowering your research journey.

MDPI Topics is cooperating with Preprints.org and has built a direct connection between MDPI journals and Preprints.org. Authors are encouraged to enjoy the benefits by posting a preprint at Preprints.org prior to publication:

  1. Immediately share your ideas ahead of publication and establish your research priority;
  2. Protect your idea from being stolen with this time-stamped preprint article;
  3. Enhance the exposure and impact of your research;
  4. Receive feedback from your peers in advance;
  5. Have it indexed in Web of Science (Preprint Citation Index), Google Scholar, Crossref, SHARE, PrePubMed, Scilit and Europe PMC.

Published Papers (9 papers)

Order results
Result details
Journals
Select all
Export citation of selected articles as:
18 pages, 12505 KiB  
Article
Reducing Oxidative Stress-Mediated Alcoholic Liver Injury by Multiplexed RNAi of Cyp2e1, Cyp4a10, and Cyp4a14
by Qi Zhang, Shuang Wu, Qiubing Chen, Yahong Zhang, Cai Zhang, Runting Yin, Zhen Ouyang and Yuan Wei
Biomedicines 2024, 12(7), 1505; https://doi.org/10.3390/biomedicines12071505 - 6 Jul 2024
Viewed by 1361
Abstract
The prevalence of excessive drinking-related alcoholic liver disease (ALD) is rising, yet therapeutic options remain limited. High alcohol consumption and consequent oxidative metabolism by cytochrome P450 (CYP) can lead to extremely high levels of reactive oxygen species, which overwhelm cellular defenses and harm [...] Read more.
The prevalence of excessive drinking-related alcoholic liver disease (ALD) is rising, yet therapeutic options remain limited. High alcohol consumption and consequent oxidative metabolism by cytochrome P450 (CYP) can lead to extremely high levels of reactive oxygen species, which overwhelm cellular defenses and harm hepatocytes. Our previous investigations showed that inhibiting Cyp2e1 using RNA interference reduced the incidence of ALD. However, compensatory mechanisms other than CYP2E1 contribute to oxidative stress in the liver. Therefore, we coupled triple siRNA lipid nanoparticles (LNPs) targeting Cyp2e1 with two isoenzymes Cyp4a10 and Cyp4a14 to treat ALD mouse models fed with Lieber–Decarli ethanol liquid diet for 12 weeks at the early (1st week), middle (5th week), and late (9th week) stages. The administration of triple siRNA LNPs significantly ameliorated chronic alcoholic liver injury in mice, and early treatment achieved the most profound effects. These effects can be attributed to a reduction in oxidative stress and increased expression of antioxidant genes, including Gsh-Px, Gsh-Rd, and Sod1. Moreover, we observed the alleviation of inflammation, evidenced by the downregulation of Il-1β, Il-6, Tnf-α, and Tgf-β, and the prevention of excessive lipid synthesis, evidenced by the restoration of the expression of Srebp1c, Acc, and Fas. Finally, triple siRNA treatment maintained normal metabolism in lipid oxidation. In brief, our research examined the possible targets for clinical intervention in ALD by examining the therapeutic effects of triple siRNA LNPs targeting Cyp2e1, Cyp4a10, and Cyp4a14. The in vivo knockdown of the three genes in this study is suggested as a promising siRNA therapeutic approach for ALD. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Diagram showing the administration of drugs for chronic alcoholic liver injury. LDC: Lieber–DeCarli; LNP: Lipid nanoparticle.</p>
Full article ">Figure 2
<p>Knockdown effects of triple siRNA LNPs in vivo. (<b>a</b>) Representative transmission electron microscopy images of LNPs encapsulated with triple siRNA. (<b>b</b>,<b>c</b>) Effect of knocking down <span class="html-italic">Cyp2e1</span> gene in an alcohol mouse model on CYP4A. (<span class="html-italic">n</span> = 4). ** <span class="html-italic">p</span> &lt; 0.01 versus pair-fed. (<b>d</b>) Changes over time in <span class="html-italic">Cyp2e1</span>, <span class="html-italic">Cyp4a10</span>, and <span class="html-italic">Cyp4a14</span> mRNA levels in mice injected with a single dose of triple siRNA LNPs. ** <span class="html-italic">p</span> &lt; 0.01 versus pair-fed. (<b>e</b>,<b>f</b>) Changes in CYP2E1 and CYP4A protein levels over time in mice injected with a single dose of triple siRNA LNPs. Data are expressed as mean ± standard deviation (SD) (<span class="html-italic">n</span> = 8). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 compared with Dulbecco’s phosphate-buffered saline (DPBS) as control; n.s. means not significant. LNPs: lipid nanoparticles.</p>
Full article ">Figure 3
<p>The role of triple gene knockdown in the treatment of alcoholic liver injury. (<b>a</b>) Gross morphology of the liver of mice in each group. (<b>b</b>) Effect of triple siRNA LNP administration on the liver index. (<b>c</b>,<b>d</b>) Effect of triple siRNA LNPs on serum AST and ALT levels in mice with alcoholic liver injury. (<b>e</b>,<b>f</b>) The protein expression levels of CYP2E1 and CYP4A. (<b>g</b>) Histopathological sections of the mouse liver were stained with hematoxylin and eosin (200×). (<b>h</b>) Sirius scarlet staining of the histopathological sections of mouse liver (200×). Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 8). ** <span class="html-italic">p</span> &lt; 0.01 versus pair-fed; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus alcohol-fed. PF/AF/ET/MT/LT: Pair-fed/Alcohol-fed/Early-treatment/Mid-treatment/Late-treatment.</p>
Full article ">Figure 4
<p>Antioxidant levels in liver tissue and the effect of triple siRNA LNPs on blood lipid levels in mice. (<b>a</b>–<b>f</b>) Effect of si-<span class="html-italic">Cyp2e1</span> LNP on hepatic glutathione peroxidase (GSH-PX), glutathione (GSH), reactive oxygen species (ROS), catalase (CAT), superoxide dismutase (SOD), and malondialdehyde (MDA) levels in alcoholic liver-injured mice. (<b>g</b>,<b>h</b>) Effect of triple siRNA LNPs on hepatic triglyceride (TG) and total cholesterol (TC) levels in mice with alcoholic liver injury. (<b>i</b>) Quantification of Oil Red O staining in different groups. (<b>j</b>) Histopathological sections of the mouse liver stained with Oil Red O (200×). Orange-red represents obvious fat accumulation in the liver. Data are expressed as the mean ± SD (<span class="html-italic">n</span> = 8). ** <span class="html-italic">p</span> &lt; 0.01 versus pair-fed; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus alcohol-fed. PF/AF/ET/MT/LT: Pair-fed/Alcohol-fed/Early-treatment/Mid-treatment/Late-treatment.</p>
Full article ">Figure 5
<p>Fluorescence immunohistochemistry of F4/80 (red) in the mouse liver; the nuclei are stained with DAPI (blue) (200×). ET/MT/LT: Early-treatment/Mid-treatment/Late-treatment.</p>
Full article ">Figure 6
<p>Role of triple siRNA LNPs in oxidative stress and levels of lipid- and inflammation-related genes and proteins. (<b>a</b>) mRNA expression levels of <span class="html-italic">Il-1β</span>, <span class="html-italic">Il-6</span>, <span class="html-italic">Tnf-α</span>, and <span class="html-italic">Tgf-β</span>. (<b>b</b>) mRNA expression levels of <span class="html-italic">Gsh-px</span>, <span class="html-italic">Gsh-rd</span>, and <span class="html-italic">Sod1</span>. (<b>c</b>) The mRNA expression levels of <span class="html-italic">Cpt1</span> and <span class="html-italic">Pgc-1α</span>. (<b>d</b>) The mRNA expression levels of <span class="html-italic">Fas</span>, <span class="html-italic">Srebp1c</span>, and <span class="html-italic">Acc</span>. (<b>e</b>,<b>f</b>) Relative protein expression levels of CPT1, SREBP1c, FAS, and PGC-1α. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 8). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 versus pair-fed; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus alcohol-fed. The mRNA levels are relative to the levels of the reference gene <span class="html-italic">Gapdh</span>, and the protein levels are relative to the levels of the reference protein GAPDH. ET: Early-treatment.</p>
Full article ">Figure 7
<p>Effects of triple siRNA LNPs on alcohol-induced ferroptosis in hepatocytes. (<b>a</b>) The mRNA expression levels of <span class="html-italic">Nrf2</span>, <span class="html-italic">HO-1</span>, and <span class="html-italic">GPX4</span>. (<b>b</b>,<b>c</b>) The relative protein expression levels of GPX4. Data are expressed as mean ± SD (<span class="html-italic">n</span> = 8). ** <span class="html-italic">p</span> &lt; 0.01 versus pair-fed; # <span class="html-italic">p</span> &lt; 0.05 and ## <span class="html-italic">p</span> &lt; 0.01 versus alcohol-fed. The mRNA levels are relative to the levels of the reference gene <span class="html-italic">Gapdh</span>, and the protein levels are relative to the levels of the reference protein GAPDH. ET: Early-treatment.</p>
Full article ">
15 pages, 4308 KiB  
Article
Therapeutic Effect of Donepezil on Neuroinflammation and Cognitive Impairment after Moderate Traumatic Brain Injury
by Dong Hyuk Youn, Younghyurk Lee, Sung Woo Han, Jong-Tae Kim, Harry Jung, Gui Seung Han, Jung In Yoon, Jae Jun Lee and Jin Pyeong Jeon
Life 2024, 14(7), 839; https://doi.org/10.3390/life14070839 - 1 Jul 2024
Cited by 1 | Viewed by 1233
Abstract
Background: Despite the important clinical issue of cognitive impairment after moderate traumatic brain injury (TBI), there is currently no suitable treatment. Here, we used in vitro and in vivo models to investigate the effect of Donepezil—an acetylcholinesterase (AChE) inhibitor—on cognitive impairment in the [...] Read more.
Background: Despite the important clinical issue of cognitive impairment after moderate traumatic brain injury (TBI), there is currently no suitable treatment. Here, we used in vitro and in vivo models to investigate the effect of Donepezil—an acetylcholinesterase (AChE) inhibitor—on cognitive impairment in the acute period following injury, while focusing on neuroinflammation and autophagy- and mitophagy-related markers. Methods: The purpose of the in vitro study was to investigate potential neuroprotective effects in TBI-induced cells after donepezil treatment, and the in vivo study, the purpose was to investigate therapeutic effects on cognitive impairment in the acute period after injury by analyzing neuroinflammation and autophagy- and mitophagy-related markers. The in vitro TBI model involved injuring SH-SY5Y cells using a cell-injury controller and then investigating the effect of donepezil at a concentration of 80 μM. The in vivo TBI model was made using a stereotaxic impactor for male C57BL/6J mice. Immuno-histochemical markers and cognitive functions were compared after 7 days of donepezil treatment (1 mg/kg/day). Mice were divided into four groups: sham operation with saline treatment, sham operation with donepezil treatment, TBI with saline treatment, and TBI with donepezil treatment (18 mice in each group). Donepezil treatment was administered within 4 h post-TBI. Results: In vitro, donepezil was found to lead to increased cell viability and 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimi-dazolylcarbocyanine iodide (JC-1), along with decreased reactive oxygen species (ROS), lactate-dehydrogenase (LDH), 2′-7′-dichlorodihydrofluorescein diacetate (DCFH-DA)-positive cells, and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells. The mRNA and protein expressions of neuroinflammation (Cyclooxygenase-2, COX-2; NOD-like receptor protein 3, NLRP3; Caspase-1; and Interleukin-1 beta, IL-1β), as well as autophagy- and mitophagy-related markers (death-associated protein kinase 1, DAPK1; PTEN-induced kinase 1, PINK1; BCL2/adenovirus E1B 19 kDa protein-interacting protein 3-like, BNIP3L; Beclin-1, BECN1; BCL2-associated X protein, BAX; microtubule-associated protein 1A/1B-light chain 3B (LC3B); Sequestosome-1; and p62) were all found to decrease after donepezil treatment. The in vivo study also showed that donepezil treatment resulted in decreased levels of cortical tissue losses and brain swelling in TBI compared to the TBI group without donepezil treatment. Donepezil treatment was also shown to decrease the mRNA and Western blotting expressions of all markers, and especially COX-2 and BNIP3L, which showed the most significant decreases. Moreover, TBI mice showed an decreased escape latency, increased alteration rate, and improved preference index, altogether pointing to better cognitive performance after donepezil treatment. Conclusions: Donepezil treatment may be beneficial in improving cognitive impairment in the early phase of moderate traumatic brain injury by ameliorating neuroinflammation, as well as autophagy and mitophagy. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Figure 1

Figure 1
<p>Results of the in vitro study. (<b>A</b>,<b>B</b>) Cell counting kit-8 (CCK-8) viability assay in SH-SY5Y cells with different dosage of donepezil (DPZ) and in in vitro traumatic brain injury model (n = 9, each group). (<b>C</b>–<b>F</b>) Comparison of lactate dehydrogenase (LDH) test (n = 9, each group), 5,5′,6,6′-tetrachloro-1,1′,3,3′-tetraethylbenzimi-dazolylcarbocyanine iodide (JC-1) (n = 9, each group), reactive oxygen species (ROS) scavenging (n = 6, each group), and 2′-7′-dichlorodihydrofluorescein diacetate (DCFH-DA) staining (n = 6 in each group). (<b>G</b>–<b>P</b>) Differences in mRNA expression levels, Western blotting (n = 4, each group), and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL)-positive cells (green, n = 6, each group) according to donepezil treatment. All in vitro experiments were repeated three times. Scale bar = 200 μm. Error bars indicate SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.005. Superoxide dismutase 2, SOD2; tumor necrosis factor, TNF-α; Interleukin-6, IL-6; Interleukin-10, IL-10; Cyclooxygenase-2, COX-2; NOD-like receptor protein 3, NLRP3; Caspase-1; Interleukin-1 beta, IL-1β.</p>
Full article ">Figure 2
<p>mRNA expression (n = 4, each group) (<b>A</b>–<b>G</b>) and Western blotting (n = 3, each group) (<b>H</b>,<b>I</b>) in autophagy- and mitophagy-related markers based on 80 μM donepezil (DZP) treatment in in vitro model of traumatic brain injury using SH-SY5Y. Error bars indicate SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.005. Death-associated protein kinase 1, DAPK1; PTEN-induced kinase 1, PINK1; BCL2/adenovirus E1B 19 kDa protein-interacting protein 3-like, BNIP3L; Beclin 1, BECN1; Bcl-2-associated X-protein, BAX; microtubule-associated proteins 1A/1B light-chain 3B (LC3B); Sequestosome 1; p62/SQSTM1; Actin, β-actin.</p>
Full article ">Figure 3
<p>Results of the in vivo study using traumatic brain injury model. (<b>A</b>,<b>B</b>) Comparison of optical visible image of mouse brains (n = 5, each group) and brain water content (n = 6, each group). (<b>C</b>,<b>D</b>) Representative images of FJB-positive cells and their differences (n = 6, each group). Scale bar = 200 μm. (<b>E</b>–<b>I</b>) mRNA expression (n = 4, each group) and Western blotting of COX-2, NLRP3, Caspase-1, and IL-1β according to donepezil (DZP) treatment (n = 3, each group). Scale bar = 200 μm. Error bars indicate SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.005. Fluoro-jade, FJB; Cyclooxygenase-2, COX-2; NOD-like receptor protein 3, NLRP3; Caspase-1; Interleukin-1 beta, IL-1β.</p>
Full article ">Figure 4
<p>Differences in mRNA expression in (n = 8, each group) (<b>A</b>–<b>G</b>) and Western blotting (n = 3, each group) (<b>H</b>,<b>I</b>) in autophagy- and mitophagy-related markers based on donepezil (DZP) treatment in in vivo model of traumatic brain injury (TBI). Error bars indicate SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.005. Death-associated protein kinase 1, DAPK1; PTEN-induced kinase 1, PINK1; BCL2/adenovirus E1B 19 kDa protein-interacting protein 3-like, BNIP3L; Beclin 1, BECN1; Bcl-2-associated X-protein, BAX; microtubule-associated proteins 1A/1B light-chain 3B, LC3B; Sequestosome 1; p62/SQSTM1; Actin, β-actin.</p>
Full article ">Figure 5
<p>Cognitive function tests after donepezil (DZP) treatment in in vivo mouse model of traumatic brain injury (TBI) (n = 9, each group). The images are representative of the group. Error bars indicate SEM. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, and *** <span class="html-italic">p</span> &lt; 0.005. Novel object recognition, NOR. Red and blue indicate the highest or lowest colors in heatmaps.</p>
Full article ">
13 pages, 1887 KiB  
Article
Protective Effect of Hyperprolactinemia on Oxidative Stress in Patients with Psychotic Disorder on Atypical Antipsychotics Risperidone and Paliperidone: A Cross-Sectional Study
by Milena Stojkovic, Mirjana Jovanovic, Vladimir Jakovljevic, Vladimir Zivkovic, Natasa Djordjevic, Aleksandar Kocovic, Marina Nikolic, Aleksandra Stojanovic, Natasa Minic, Vesna Ignjatovic, Vladimir Vukomanovic, Danijela Nastic, Natasa Zdravkovic, Olivera Radmanovic, Milan Djordjic, Sasa Babic and Branimir Radmanovic
Biomedicines 2024, 12(7), 1418; https://doi.org/10.3390/biomedicines12071418 - 26 Jun 2024
Viewed by 1081
Abstract
Several studies indicate the impact of antipsychotics like risperidone and paliperidone on oxidative stress parameters, yet data remain inconsistent. We investigated the link between these medications, hyperprolactinemia (HPRL), and oxidative stress. This study was conducted at the Psychiatry Clinic, University Clinical Center, Kragujevac, [...] Read more.
Several studies indicate the impact of antipsychotics like risperidone and paliperidone on oxidative stress parameters, yet data remain inconsistent. We investigated the link between these medications, hyperprolactinemia (HPRL), and oxidative stress. This study was conducted at the Psychiatry Clinic, University Clinical Center, Kragujevac, between November 2022 and August 2023. Inclusion criteria comprised diagnosed psychotic disorders from the ICD-10-based F20-F29 spectrum and clinical stability on risperidone/paliperidone for ≥12 weeks with no recent dose adjustments. Exclusion criteria included pregnancy, breastfeeding, relevant medical conditions, or co-therapy with prolactin-secreting drugs. Data encompassed drug choice, administration method, therapy duration, and daily dose. Prolactin (PRL) levels, oxidative stress parameters (TBARS, H2O2, O2, NO2), and antioxidant system (CAT, GSH, SOD) were assessed. Of 155 subjects, women exhibited significantly higher PRL levels (p < 0.001) and symptomatic HPRL (p < 0.001). Drug choice and regimen significantly influenced TBARS (p < 0.001), NO2 (p < 0.001), O2 (p = 0.002), CAT (p = 0.04), and GSH (p < 0.001) levels. NO2 levels were affected by drug dose (p = 0.038). TBARS (p < 0.001), O2 (p < 0.001), and SOD (p = 0.022) inversely correlated with PRL levels, suggesting PRL’s protective role against oxidative stress. The female sex association with higher PRL levels implies additional factors influencing PRL’s antioxidant role. Antipsychotic choice and dosage impact PRL and oxidative stress markers, necessitating further exploration. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Figure 1

Figure 1
<p>Comparison of the mean values of the parameters of oxidation stress between sexes and in total. TBARS—index of lipid peroxidation (graph (<b>A</b>)); NO—nitric oxide (graph (<b>B</b>)); O<sub>2</sub><sup>−</sup>—superoxide anion radical (graph (<b>C</b>)); H<sub>2</sub>O<sub>2</sub>—hydrogen peroxide (graph (<b>D</b>)); SOD—superoxide anion radical (graph (<b>E</b>)); CAT—catalase (graph (<b>F</b>)); GSH—reduced glutathione (graph (<b>G</b>)).</p>
Full article ">Figure 2
<p>Comparison of the mean values of the parameters of oxidation stress between different therapy regimens. TBARS—index of lipid peroxidation (graph (<b>A</b>); NO—nitric oxide (graph (<b>B</b>)); O<sub>2</sub><sup>−</sup>—superoxide anion radical (graph (<b>C</b>)); H<sub>2</sub>O<sub>2</sub>—hydrogen peroxide (graph (<b>D</b>)); SOD—superoxide anion radical (graph (<b>E</b>)); CAT—catalase (graph (<b>F</b>)); GSH—reduced glutathione (graph (<b>G</b>)).</p>
Full article ">Figure 3
<p>Comparison of the mean values of the parameters of oxidation stress between different categories of drug dosage. TBARS—index of lipid peroxidation (graph (<b>A</b>)); NO—nitric oxide (graph (<b>B</b>)); O<sub>2</sub><sup>−</sup>—superoxide anion radical (graph (<b>C</b>)); H<sub>2</sub>O<sub>2</sub>—hydrogen peroxide (graph (<b>D</b>)). The dose of the drug was categorized as small (2–4 mg/day), medium (5–6 mg/day), and high (7–8 mg/day).</p>
Full article ">Figure 4
<p>Comparison of the mean values of the parameters of oxidation stress between groups categorized by the level of prolactin and symptom manifestation. TBARS—index of lipid peroxidation (graph (<b>A</b>)); NO—nitric oxide (graph (<b>B</b>)); O<sub>2</sub><sup>−</sup>—superoxide anion radical (graph (<b>C</b>)); H<sub>2</sub>O<sub>2</sub>—hydrogen peroxide (graph (<b>D</b>)); SOD—superoxide anion radical (graph (<b>E</b>)); CAT—catalase (graph (<b>F</b>)); GSH—reduced glutathione (graph (<b>G</b>)); PRL—prolactin; HPRL—hyperprolctinemia.</p>
Full article ">Figure 5
<p>The association of the drug used in therapy, level of prolactin, and symptom manifestation. PRL—prolactin; HPRL—hyperprolctinemia.</p>
Full article ">
23 pages, 3076 KiB  
Article
A Nonclinical Safety Evaluation of Cold Atmospheric Plasma for Medical Applications: The Role of Genotoxicity and Mutagenicity Studies
by Piimwara Yarangsee, Supakit Khacha-ananda, Pornsiri Pitchakarn, Unchisa Intayoung, Sirikhwan Sriuan, Jirarat Karinchai, Apiwat Wijaikhum and Dheerawan Boonyawan
Life 2024, 14(6), 759; https://doi.org/10.3390/life14060759 - 13 Jun 2024
Viewed by 849
Abstract
Atmospheric nonthermal plasma (ANTP) has rapidly evolved as an innovative tool in biomedicine with various applications, especially in treating skin diseases. In particular, the formation of reactive oxygen species (ROS) and nitrogen species (RNS), which are generated by ANTP, plays an important role [...] Read more.
Atmospheric nonthermal plasma (ANTP) has rapidly evolved as an innovative tool in biomedicine with various applications, especially in treating skin diseases. In particular, the formation of reactive oxygen species (ROS) and nitrogen species (RNS), which are generated by ANTP, plays an important role in the biological signaling pathways of human cells. Unfortunately, excessive amounts of these reactive species significantly result in cellular damage and cell death induction. To ensure the safe application of ANTP, preclinical in vitro studies must be conducted before proceeding to in vivo or clinical trials involving humans. Our study aimed to investigate adverse effects on genetic substances in murine fibroblast cells exposed to ANTP. Cell viability and proliferation were markedly reduced after exposing the cells with plasma. Both extracellular and intracellular reactive species, especially RNS, were significantly increased upon plasma exposure in the culture medium and the cells. Notably, significant DNA damage in the cells was observed in the cells exposed to plasma. However, plasma was not classified as a mutagen in the Ames test. This suggested that plasma led to the generation of both extracellular and intracellular reactive species, particularly nitrogen species, which affect cell proliferation and are also known to induce genetic damage in fibroblast cells. These results highlight the genotoxic and mutagenic effects of ANTP, emphasizing the need for the cautious selection of plasma intensity in specific applications to avoid adverse side effects resulting from reactive species production. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Figure 1

Figure 1
<p>The Nightingale instrument. (<b>A</b>) The instrument consisted of an air filter channel to pump atmospheric air into the plasma head containing an electrode, in which plasma was generated under a high electromagnetic field and released through the plasma head via four channels of air jets. (<b>B</b>) The direct exposure of plasma to the NIH3T3 murine fibroblasts which were cultured in DMEM medium at a distance of approximately 1.6 cm from the surface of the cell lines.</p>
Full article ">Figure 2
<p>The cell viability of fibroblast cells after exposure with plasma. The cells were exposed to different air flow rates coupled with plasma. The percentage of fibroblast cell viability is expressed as mean and standard deviation. * and ** indicate a statistical difference in cell viability, comparing plasma intensities of 4, 7, and 10 pulses to a plasma intensity of 0 pulses at the same air flow rate (* <span class="html-italic">p</span> value &lt; 0.05; ** <span class="html-italic">p</span> value &lt; 0.01). The lower-case letters indicate a statistical difference in cell viability comparing the same plasma intensity. Data are representative of three independent experiments.</p>
Full article ">Figure 3
<p>The suppression of cell proliferation after exposure to plasma. (<b>A</b>) The proliferative effect of plasma on the plasma-exposed cells demonstrated by a colony formation assay. The surviving colonies were stained and counted. (<b>B</b>) The percentage of colony formation is expressed as mean and standard deviation. *, **, and *** indicate a statistical difference in the percentage of colony formation, comparing plasma intensities of 4, 7, and 10 pulses to a plasma intensity of 0 pulses at the same flow rate (* <span class="html-italic">p</span> value &lt; 0.05; ** <span class="html-italic">p</span> value &lt; 0.01; *** <span class="html-italic">p</span> value &lt; 0.001). The lower-case letters indicate a statistical difference in cell viability comparing the same plasma intensity. Data are representative of three independent experiments.</p>
Full article ">Figure 4
<p>Modulation of intracellular ROS and RNS. (<b>A</b>) Percentage of ROS-positive cells in plasma-exposed cells. The treated cells were harvested to stain them with 2′,7′-dichlorofluorescin diacetate. The stained cells were counted by a flow cytometer. (<b>B</b>) Percentage of RNS-positive cells in plasma-exposed cells. The treated cells were harvested to stain them with 4-amino-5-methylamino-2′,7′-difluorofluorescein diacetate. The stained cells were counted by a flow cytometer. Hydrogen peroxide and diethylamine NONOate sodium salt were used as positive controls for ROS and RNS, respectively. The percentage of ROS- and RNS-positive cells was expressed as mean and standard deviation. ** and *** indicate a statistical difference, comparing plasma intensities of 4, 7, and 10 pulses as well as the positive control to a plasma intensity of 0 pulses (** <span class="html-italic">p</span> value &lt; 0.01; *** <span class="html-italic">p</span> value &lt; 0.001). Data are representative of three independent experiments.</p>
Full article ">Figure 5
<p>The quantification of extracellular (<b>A</b>) H<sub>2</sub>O<sub>2</sub> and (<b>B</b>) NO in the plasma-exposed culture medium. The culture medium was directly exposed to plasma for 30 s; then, the culture medium was harvested and reacted with the specific reagent. The amount of H<sub>2</sub>O<sub>2</sub> and NO was calculated from the standard curve and expressed as mean and standard deviation. *** indicates a statistical difference, comparing plasma intensities of 4, 7, and 10 pulses to a plasma intensity of 0 pulses at the same flow rate (*** <span class="html-italic">p</span> value &lt; 0.001). Data are representative of three independent experiments.</p>
Full article ">Figure 6
<p>The DNA damage was measured by (<b>A</b>) the percentage of TUNEL-positive cells and (<b>B</b>) the amount of intracellular 8-OHdG in plasma-exposed cells. The TUNEL assay was used for investigating DNA strand break. The commercial ELISA kit was also chosen for the quantification of 8-OHdG. H<sub>2</sub>O<sub>2</sub> was used as a positive control. The data were expressed as mean and standard deviation. * and *** indicate a statistical difference, comparing plasma intensities of 4, 7, and 10 pulses as well as the positive control to a plasma intensity of 0 pulses (* <span class="html-italic">p</span> value &lt; 0.05 and *** <span class="html-italic">p</span> value &lt; 0.001). <sup>##</sup> indicates a statistical difference, comparing air flow rates of 3 and 5 L/min (<sup>##</sup> <span class="html-italic">p</span> value &lt; 0.01). Data are representative of three independent experiments.</p>
Full article ">Figure 7
<p>The immunofluorescent staining of gamma H2AX (γH2AX) on plasma-exposed cells. (<b>A</b>) Control cells were stained with gamma H2AX at 0 and 24 h. The nucleus was counterstained with DAPI. The fluorescent-staining cells were observed and photographed under an inverted microscope. (<b>B</b>) Plasma-exposed cells were exposed to a CAPJ at an intensity of 10 pulses and under an air flow rate of 5 L/min for 30 s, then further incubated for 12 and 24 h. The cells were stained with gamma H2AX (γH2AX) and counterstained with DAPI. Green dot shows γ-H2AX foci, blue nuclei stained with DAPI.</p>
Full article ">Figure 8
<p>The growth rate of plasma-exposed bacteria was assessed to test the effect of plasma on the bacterial growth of (<b>A</b>) strain TA98 and (<b>B</b>) strain TA100. After exposure to two strains of bacteria with plasma, the plasma-exposed bacteria were diluted and added to a sterile 96-well plate. The plate was then incubated at 37 °C for 24 h. The optical density (OD) at a wavelength of 600 nm was measured every 10 min. The growth curve is represented with the <span class="html-italic">x</span>-axis indicating time of incubation and the <span class="html-italic">y</span>-axis indicating OD600 nm. The results are expressed as the average and standard deviation of triplicate experiments.</p>
Full article ">Figure 9
<p>The mutagenicity of plasma was tested on (<b>A</b>) <span class="html-italic">Salmonella typhimurium</span> strain TA98 and (<b>B</b>) <span class="html-italic">S. typhimurium</span> strain TA100. The bacteria were directly exposed to plasma for 30 s. After that, the treated bacteria were cultured on a culture medium. The number of revertant colonies was counted. The mutagenic index was calculated and is expressed as mean and standard deviation. The mutagenicity of the test substance was classified by a mutagenic index of more than 2 (over the dot line). Data are representative of three independent experiments. The dotted line represents the cut-off to indicate the test substance is not classified as mutagens (mutagenic index less than 2).</p>
Full article ">
17 pages, 5450 KiB  
Article
Saliva as a Diagnostic Tool for Early Detection of Exercise-Induced Oxidative Damage in Female Athletes
by Aleksandr N. Ovchinnikov and Antonio Paoli
Biomedicines 2024, 12(5), 1006; https://doi.org/10.3390/biomedicines12051006 - 2 May 2024
Viewed by 1479
Abstract
Although blood still remains the most commonly utilized medium to detect increased levels of oxidative damage induced by exercise, saliva diagnostics have gained increasing popularity due to their non-invasive nature and athlete-friendly collection process. Given that the contribution of various phases of the [...] Read more.
Although blood still remains the most commonly utilized medium to detect increased levels of oxidative damage induced by exercise, saliva diagnostics have gained increasing popularity due to their non-invasive nature and athlete-friendly collection process. Given that the contribution of various phases of the menstrual cycle to the levels of oxidative damage may differ, the aim of this study was to evaluate an agreement between salivary and plasmatic levels of lipid peroxidation products in female swimmers in both the follicular (F) and luteal (L) phases of the menstrual cycle at rest and following exercise. Twelve well-trained female swimmers aged 19.6 ± 1.1 years old were examined. We measured diene conjugates (DCs), triene conjugates (TCs), and Schiff bases (SBs) in lipids immediately after their extraction from both saliva and blood plasma. All female swimmers were studied two times each, in the two different phases of one menstrual cycle, before and after high-intensity interval exercise (HIIE). Salivary and plasmatic levels of DCs, TCs, and SBs significantly increased post-exercise compared to pre-exercise, in both the F and L phases. A high positive correlation was observed between the concentrations of DCs, TCs, and SBs in the saliva and blood plasma of participants in the F and L phases, both at rest and following HIIE. Ordinary least products regression analysis indicates that there was no proportional and differential bias in the data. The Bland–Altman method also declares that there was no differential bias, since the line of equality was within the 95% confidence interval of the mean difference between salivary and plasmatic levels of DCs, TCs, and SBs in female swimmers, in both the F and L phases, before and after HIIE. There was also no proportional bias in the Bland–Altman plots. Thus, this is the first study to report a high agreement between the quantifications of DCs, TCs, and SBs in the saliva and blood plasma of female swimmers in both the F and L phases, at rest and following HIIE. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Salivary and plasmatic levels of DCs, TCs, and SBs in female swimmers in the follicular and luteal phases of the menstrual cycle at rest. Data are expressed as means, augmented with the medians and interquartile ranges, and are compared using paired-sample <span class="html-italic">t</span>-tests. DCs, diene conjugates; TCs, triene conjugates; SBs, Schiff bases.</p>
Full article ">Figure 2
<p>Salivary and plasmatic levels of DCs, TCs, and SBs in female swimmers in the follicular and luteal phases of the menstrual cycle following high-intensity interval exercise. Data are expressed as means, augmented with the medians and interquartile ranges, and are compared using paired-sample <span class="html-italic">t</span>-tests. DCs, diene conjugates; TCs, triene conjugates; SBs, Schiff bases.</p>
Full article ">Figure 3
<p>Salivary and plasmatic levels of DCs, TCs, and SBs in female swimmers in the follicular phase of the menstrual cycle before and immediately after high-intensity interval exercise. Data are expressed as means, augmented with the medians and interquartile ranges, and are compared using paired-sample <span class="html-italic">t</span>-tests. DCs, diene conjugates; TCs, triene conjugates; SBs, Schiff bases.</p>
Full article ">Figure 4
<p>Salivary and plasmatic levels of DCs, TCs, and SBs in female swimmers in the luteal phase of the menstrual cycle before and immediately after high-intensity interval exercise. Data are expressed as means, augmented with the medians and interquartile ranges, and are compared using paired-sample <span class="html-italic">t</span>-tests. DCs, diene conjugates; TCs, triene conjugates; SBs, Schiff bases.</p>
Full article ">Figure 5
<p>Correlation between plasmatic and salivary levels of DCs, TCs, and SBs in female swimmers in the follicular phase of the menstrual cycle at both pre-exercise and post-exercise. The solid black lines represent the ordinary least squares regression of salivary levels on plasmatic levels. The grey shaded regions indicate the 95% confidence intervals. DCs, diene conjugates; TCs, triene conjugates; SBs, Schiff bases.</p>
Full article ">Figure 6
<p>Correlation between plasmatic and salivary levels of DCs, TCs, and SBs in female swimmers in the luteal phase of the menstrual cycle at both pre-exercise and post-exercise. The solid black lines represent the ordinary least squares regression of salivary levels on plasmatic levels. The grey shaded regions indicate the 95% confidence intervals. DCs, diene conjugates; TCs, triene conjugates; SBs, Schiff bases.</p>
Full article ">Figure 7
<p>Bland–Altman plots of differences against means for comparisons of salivary and plasmatic levels of the LPO products measured in female swimmers in the follicular phase of the menstrual cycle both pre-exercise and post-exercise. The mean difference is represented by a dashed line (inside the blue area) parallel to the <span class="html-italic">x</span> axis. The limits of agreement are represented by dashed lines parallel to the <span class="html-italic">x</span> axis at −1.96 SD (inside the red area) and +1.96 SD (inside the green area). Shaded areas represent the 95% confidence interval limits for the mean difference (blue shading) and for the agreement limits (red and green shading). The solid blue lines represent the ordinary least squares regression of differences on means. The grey shaded regions indicate the 95% confidence intervals. DC, diene conjugate; TC, triene conjugate; SB, Schiff base.</p>
Full article ">Figure 8
<p>Bland–Altman plots of differences against means for comparisons of salivary and plasmatic levels of the LPO products measured in female swimmers in the luteal phase of the menstrual cycle both pre-exercise and post-exercise. The mean difference is represented by a dashed line (inside the blue area) parallel to the <span class="html-italic">x</span> axis. The limits of agreement are represented by dashed lines parallel to the <span class="html-italic">x</span> axis at −1.96 SD (inside the red area) and +1.96 SD (inside the green area). Shaded areas represent the 95% confidence interval limits for the mean difference (blue shading) and for the agreement limits (red and green shading). The solid blue lines represent the ordinary least squares regression of differences on means. The grey shaded regions indicate the 95% confidence intervals. DC, diene conjugate; TC, triene conjugate; SB, Schiff base.</p>
Full article ">
24 pages, 5974 KiB  
Article
Human Macrophages Activate Bystander Neutrophils’ Metabolism and Effector Functions When Challenged with Mycobacterium tuberculosis
by Dearbhla M. Murphy, Anastasija Walsh, Laura Stein, Andreea Petrasca, Donal J. Cox, Kevin Brown, Emily Duffin, Gráinne Jameson, Sarah A. Connolly, Fiona O’Connell, Jacintha O’Sullivan, Sharee A. Basdeo, Joseph Keane and James J. Phelan
Int. J. Mol. Sci. 2024, 25(5), 2898; https://doi.org/10.3390/ijms25052898 - 1 Mar 2024
Cited by 1 | Viewed by 2486
Abstract
Neutrophils are dynamic cells, playing a critical role in pathogen clearance; however, neutrophil infiltration into the tissue can act as a double-edged sword. They are one of the primary sources of excessive inflammation during infection, which has been observed in many infectious diseases [...] Read more.
Neutrophils are dynamic cells, playing a critical role in pathogen clearance; however, neutrophil infiltration into the tissue can act as a double-edged sword. They are one of the primary sources of excessive inflammation during infection, which has been observed in many infectious diseases including pneumonia and active tuberculosis (TB). Neutrophil function is influenced by interactions with other immune cells within the inflammatory lung milieu; however, how these interactions affect neutrophil function is unclear. Our study examined the macrophage–neutrophil axis by assessing the effects of conditioned medium (MΦ-CM) from primary human monocyte-derived macrophages (hMDMs) stimulated with LPS or a whole bacterium (Mycobacterium tuberculosis) on neutrophil function. Stimulated hMDM-derived MΦ-CM boosts neutrophil activation, heightening oxidative and glycolytic metabolism, but diminishes migratory potential. These neutrophils exhibit increased ROS production, elevated NET formation, and heightened CXCL8, IL-13, and IL-6 compared to untreated or unstimulated hMDM-treated neutrophils. Collectively, these data show that MΦ-CM from stimulated hMDMs activates neutrophils, bolsters their energetic profile, increase effector and inflammatory functions, and sequester them at sites of infection by decreasing their migratory capacity. These data may aid in the design of novel immunotherapies for severe pneumonia, active tuberculosis and other diseases driven by pathological inflammation mediated by the macrophage–neutrophil axis. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Figure 1

Figure 1
<p>Methods Summary. (<b>A</b>) Macrophage Stage: hMDMs, differentiated from PBMCs isolated from healthy blood donors over 7 days, were stimulated with Mtb for three hours and washed to remove unphagocytosed Mtb (hMDMs were also left unstimulated and treated with 100 ng/mL LPS in parallel). Then, 24 h post-stimulation, the hMDM-conditioned medium (MΦ-CM) was processed and stored at −80 °C for later use. MΦ-CM from a minimum of three independent donors was pooled and was used for all subsequent experimentation techniques. Neutrophil Stage: On a subsequent day, fresh CD15+ cells were isolated, purified, and treated with 20% MΦ-CM (80% fresh cRPMI) for one hour. Flow cytometry, real-time extracellular Seahorse flux analysis, and MSD ELISAs were subsequently carried out to determine the effect of the MΦ-CM on neutrophil function. (<b>B</b>) All treatments with MΦ-CM consisted of freshly isolated neutrophils being exposed to 20% cRPMI (RPMI supplemented with 10% human serum), 20% MΦ-CM from unstimulated hMDMs (UsCoM), 20% MΦ-CM from Mtb-stimulated hMDMs (MtbCoM), and 20% MΦ-CM from LPS-stimulated hMDMs (LPSCoM).</p>
Full article ">Figure 2
<p>Characterizing secreted cytokine and chemokine levels in MΦ-CM (UsCoM, MtbCoM, and LPSCoM) from unstimulated, Mtb-stimulated, and LPS-stimulated hMDMs. hMDMs, differentiated from PBMCs isolated from healthy blood donors, were stimulated with iH37Rv Mtb for three hours and were washed to remove unphagocytosed Mtb. hMDMs were also left unstimulated and treated with LPS (100 ng/mL) in parallel. (<b>A</b>) 24 h post-stimulation, the secreted levels of the cytokines TNF-α, IL-1β, IL-10, IL-4, IL-2, CXCL8, IL-13, IFN-γ, and IL-6 were quantified using MSD Multi-Array technology, (<b>B</b>) along with the chemokines IP-10, MCP-4, TARC, MIP-1α, MDC, Eotaxin, Eotaxin-3, MIP-1β, and MCP-1. Bars denote mean ± SEM (n = 18 independent donors). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001, ns = no significance (Friedman ANOVA test with Dunn’s multiple comparisons tests).</p>
Full article ">Figure 3
<p>Examining if MΦ-CM from Mtb-stimulated or LPS-stimulated hMDMs affects human neutrophil activation and migration. (<b>A</b>) Neutrophils exposed to 20% MΦ-CM were visualised under light microscopy (40×) to examine if the MΦ-CM affects neutrophil morphology. (<b>B</b>) Flow cytometric analyses were carried out to examine if neutrophils exposed to MΦ-CM could be activated, first by gating on single cells (FSC-A vs. FSC-H), granular cells (FSC-A vs. SSC-A), and finally viable CD15<sup>+</sup> neutrophils (AmCyan vs. PE-Cy7). (<b>C</b>) Neutrophil activation was assessed by determining the expression of the granulation marker CD62L and (<b>D</b>) the degranulation marker CD63 in viable CD15<sup>+</sup> neutrophils in response to cRPMI, UsCoM, MtbCoM, and LPSCoM (n = 5). (<b>E</b>) As activation status is a good surrogate indicator of migration in neutrophils, the ability of neutrophils to migrate was also assessed. Cells were treated with 20% MΦ-CM for one hour as before, placed into the top chamber of 5 µm transwell inserts, and allowed to migrate toward 1.25 ng/mL fMLP in the bottom chamber for 15 min. Samples were then stained for CD15 expression and viability, an equal number of Precision<sup>TM</sup> Counting Beads were added, and the numbers of migrated neutrophils (cells/µL) were quantified by flow cytometry (n = 6). (<b>F</b>) Expression of the cell surface migration marker CXCR2 was examined in neutrophils exposed to the MΦ-CM treatments (n = 5). * <span class="html-italic">p</span> &lt; 0.05 and ** <span class="html-italic">p</span> &lt; 0.01 (Friedman ANOVA test with Dunn’s multiple comparisons tests). MFI: Median fluorescence intensity.</p>
Full article ">Figure 4
<p>Investigating if MΦ-CM from Mtb-stimulated or LPS-stimulated hMDMs affects baseline glycolysis and oxidative phosphorylation in unstimulated human neutrophils. The effect of MΦ-CM on the real-time baseline (<b>A</b>) OCR (n = 14) and (<b>B</b>) ECAR (n = 18), representing oxidative phosphorylation and glycolysis, respectively, was determined utilising Seahorse extracellular flux assays in neutrophils treated for one hour with 20% cRPMI, UsCoM, MtbCoM, and LPSCoM. (<b>C</b>) The immunometabolic shift from UsCoM treatment to MtbCoM and LPSCoM treatment can be illustrated by the metabolic phenograms. ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 (Friedman ANOVA test with Dunn’s multiple comparisons tests).</p>
Full article ">Figure 5
<p>Assessing if MΦ-CM from Mtb-stimulated or LPS-stimulated hMDMs affects bioenergetics in fMPL-stimulated and Mtb-stimulated human neutrophils. (<b>A</b>) Neutrophils were seeded into Labtek II chamber slides, unstimulated or stimulated with iH37Rv-Mtb, washed, and phagocytosed Mtb was fluorescently visualised through auramine (FITC) and hoechst (DAPI) staining. (<b>B</b>) Baseline metabolic OCR (left) and ECAR (right) values were examined in neutrophils in response to iH37Rv-Mtb (MOI: 1–10), (<b>C</b>) prior to determining if 20% cRPMI, UsCoM, MtbCoM, and LPSCoM pre-treatment exhibited an effect on OCR and ECAR profiles in neutrophils stimulated with iH37Rv (n = 3). After neutrophils were treated for one hour with 20% cRPMI, UsCoM, MtbCoM, and LPSCoM, cells were also stimulated with fMLP (1.25 ng/mL) and real-time baseline (<b>D</b>) OCR and (<b>E</b>) ECAR values were determined utilising Seahorse extracellular flux assays (n = 5). (<b>D</b>,<b>E</b>) OCR and ECAR rates were plotted and graphed (right-hand side graphs) at the time point reflecting the highest metabolic response to fMLP stimulation (40 min). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 ((<b>A</b>,<b>B</b>): Friedman ANOVA test with Dunn’s multiple comparisons tests; (<b>D</b>,<b>E</b>): Two-way RM ANOVA with Šídák’s multiple comparisons test).</p>
Full article ">Figure 6
<p>Elucidating if MΦ-CM from Mtb-stimulated or LPS-stimulated hMDMs alters cytokine release in primary human neutrophils. Human neutrophils were treated with 20% cRPMI, UsCoM, MtbCoM, and LPSCoM for 1 h. Following incubation, neutrophils were left unstimulated (<b>A</b>) and stimulated with 1.25 ng/mL fMLP (<b>B</b>). 4 h later, the secreted levels of CXCL8, IL-13, and IL-6 were assessed using MSD-ELISA (n = 13). * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, *** <span class="html-italic">p</span> &lt; 0.001, and **** <span class="html-italic">p</span> &lt; 0.0001 ((<b>A</b>): Friedman ANOVA test with Dunn’s multiple comparisons tests; (<b>B</b>): Two-way ANOVA with mixed effects model multiple comparisons test).</p>
Full article ">Figure 7
<p>Determining if MΦ-CM from Mtb-stimulated or LPS-stimulated hMDMs alters NETosis, reactive oxygen species (ROS) production, glucose uptake, and apoptosis in primary human neutrophils. Flow cytometric analyses were carried out to examine if neutrophils exposed to MΦ-CM exhibited altered NETosis, ROS production, and glucose uptake. This was achieved by first gating on single cells (FSC-A vs. FSC-H), granular cells (FSC-A vs. SSC-A), and finally, viable CD15+ neutrophils (AmCyan vs. PE-Cy7). (<b>A</b>) Flow cytometric dot plots illustrating the effect of 20% MΦ-CM on NETosis in a representative donor (NETosis is characterized as being double positive in the Sytox red (APC) and DAPI (Pacific Blue) channels). Histogram shifts in (<b>B</b>) DHR-123 (FITC) and (<b>C</b>) 2-NBDG (FITC) expression are characterized by changes in ROS production and glucose uptake in MΦ-CM-treated CD15+ neutrophils, respectively. Using these flow cytometric tools, NETosis (n = 10), ROS production (n = 16), and glucose uptake (n = 4) were assessed in CD15+ neutrophils one hour post-treatment with 20% MΦ-CM. (<b>D</b>) Early apoptosis (characterized as CD15+/Annexin V+) and late apoptosis (characterized as CD15+/Annexin V+/Zombie+) were also determined to examine the effect of the MΦ-CM on cell viability. A representative flow cytometric dot plot is shown. * <span class="html-italic">p</span> &lt; 0.05, ** <span class="html-italic">p</span> &lt; 0.01, **** <span class="html-italic">p</span> &lt; 0.0001 (Friedman ANOVA test with Dunn’s multiple comparisons tests).</p>
Full article ">Figure 8
<p>A graphical summary of the study. Freshly isolated CD15+ neutrophils were exposed to four different conditions, complete RPMI media (cRPMI), media from unconditioned MDMs (UsCoM), media from MDMs infected with Mtb (MtbCoM), and media from MDMs stimulated with LPS (LPSCoM). Glucose uptake and apoptosis were unaffected by MtbCoM and LPSCoM. Effects on several aspects of neutrophil function were examined via several methods, including flow cytometry, Agilent Seahorse assays, MSD ELISA, and transwell migration assays. Our data suggests an augmentation of many neutrophil functions as a result of MtbCoM and LPSCoM, such as NETosis, ROS generation, and inflammatory cytokine production. Neutrophils exposed to the inflammatory conditioned media also exhibited a significant energetic shift, with glycolysis and oxidative phosphorylation significantly augmented. MtbCoM and LPSCoM also decrease neutrophil chemotaxis, as shown both via the quantification of the chemotactic receptor CXCR2 and via transwell migration assays. Overall, our data demonstrated the importance of the macrophage–neutrophil axis in infection.</p>
Full article ">
22 pages, 18029 KiB  
Article
Reactive Oxygen Species-Dependent Activation of EGFR/Akt/p38 Mitogen-Activated Protein Kinase and JNK1/2/FoxO1 and AP-1 Pathways in Human Pulmonary Alveolar Epithelial Cells Leads to Up-Regulation of COX-2/PGE2 Induced by Silica Nanoparticles
by Yan-Jyun Lin, Chien-Chung Yang, I-Ta Lee, Wen-Bin Wu, Chih-Chung Lin, Li-Der Hsiao and Chuen-Mao Yang
Biomedicines 2023, 11(10), 2628; https://doi.org/10.3390/biomedicines11102628 - 25 Sep 2023
Cited by 2 | Viewed by 1697
Abstract
The risk of lung exposure to silica nanoparticles (SiNPs) and related lung inflammatory injury is increasing with the wide application of SiNPs in a variety of industries. A growing body of research has revealed that cyclooxygenase (COX)-2/prostaglandin E2 (PGE2) up-regulated [...] Read more.
The risk of lung exposure to silica nanoparticles (SiNPs) and related lung inflammatory injury is increasing with the wide application of SiNPs in a variety of industries. A growing body of research has revealed that cyclooxygenase (COX)-2/prostaglandin E2 (PGE2) up-regulated by SiNP toxicity has a role during pulmonary inflammation. The detailed mechanisms underlying SiNP-induced COX-2 expression and PGE2 synthesis remain unknown. The present study aims to dissect the molecular components involved in COX-2/PGE2 up-regulated by SiNPs in human pulmonary alveolar epithelial cells (HPAEpiCs) which are one of the major targets while SiNPs are inhaled. In the present study, we demonstrated that SiNPs induced COX-2 expression and PGE2 release, which were inhibited by pretreatment with a reactive oxygen species (ROS) scavenger (edaravone) or the inhibitors of proline-rich tyrosine kinase 2 (Pyk2, PF-431396), epidermal growth factor receptor (EGFR, AG1478), phosphatidylinositol 3-kinase (PI3K, LY294002), protein kinase B (Akt, Akt inhibitor VIII), p38 mitogen-activated protein kinase (MAPK) (p38 MAPK inhibitor VIII), c-Jun N-terminal kinases (JNK)1/2 (SP600125), Forkhead Box O1 (FoxO1, AS1842856), and activator protein 1 (AP-1, Tanshinone IIA). In addition, we also found that SiNPs induced ROS-dependent Pyk2, EGFR, Akt, p38 MAPK, and JNK1/2 activation in these cells. These signaling pathways induced by SiNPs could further cause c-Jun and FoxO1 activation and translocation from the cytosol to the nucleus. AP-1 and FoxO1 activation could increase COX-2 and PGE2 levels induced by SiNPs. Finally, the COX-2/PGE2 axis might promote the inflammatory responses in HPAEpiCs. In conclusion, we suggested that SiNPs induced COX-2 expression accompanied by PGE2 synthesis mediated via ROS/Pyk2/EGFR/PI3K/Akt/p38 MAPK- and JNK1/2-dependent FoxO1 and AP-1 activation in HPAEpiCs. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Figure 1

Figure 1
<p><b>SiNPs induce COX-2 protein and mRNA expression, as well as PGE<sub>2</sub> production in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were incubated with different concentrations (10, 25, and 50 μg/mL) of SiNPs for the indicated time intervals (0, 4, 6, 8, 10, and 12 h). The levels of COX-2 and GAPDH protein were determined by Western blot. (<b>B</b>) HPAEpiCs were treated with 25 μg/mL SiNPs for the indicated time intervals (0, 2, 4, 6, 8, 10 h). The COX-2 and GAPDH mRNA levels were determined by quantitative real-time PCR. (<b>C</b>) The culture media from (<b>A</b>), treated with 25 μg/mL SiNPs, were collected to determine the levels of PGE<sub>2</sub> using a PGE<sub>2</sub> ELISA kit. (<b>D</b>) Cells were incubated with different concentrations (5, 10, 25, 50, and 100 μg/mL) of SiNPs for 12 h. Cell viability was determined using the Cell Counting Kit 8. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel.</p>
Full article ">Figure 2
<p><b>SiNP-induced COX-2 expression and PGE<sub>2</sub> production require ongoing transcription and translation in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of Act. D (1, 10, and 100 nM) or CHI (0.01, 0.1, and 1 μM) for 1 h, respectively, and then incubated with 25 μg/mL SiNPs for 12 h. COX-2 and GAPDH protein expression were determined by Western blot. (<b>B</b>) HPAEpiCs were pretreated with Act. D (100 nM) or CHI (1 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were analyzed by quantitative real-time PCR. (<b>C</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel.</p>
Full article ">Figure 3
<p><b>ROS generation induced by SiNPs mediates COX-2 expression and PGE<sub>2</sub> production in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of edaravone (1, 10, and 100 nM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. COX-2 and GAPDH protein expression were determined by Western blot. (<b>B</b>) Cells were pretreated with edaravone (0.1 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The COX-2 and GAPDH mRNA levels were determined by quantitative real-time PCR. (<b>C</b>) Cells were stimulated with 25 μg/mL SiNPs for 15, 30, and 60 min (upper panel) and were pretreated without or with edaravone (100 nM) for 1 h before exposure to 25 μg/mL SiNPs for 30 min (lower panel), followed by incubation with H<sub>2</sub>DCF-DA (5 μM) for 30 min. The fluorescence intensity of cells was measured using a fluorescent microplate reader. (<b>D</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel.</p>
Full article ">Figure 4
<p><b>SiNPs induced COX-2 expression and PGE<sub>2</sub> production via Pyk2 activation in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of PF-431396 (0.1, 1, and 3 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) HPAEpiCs were pretreated with PF-431396 (3 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were analyzed by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled or Pyk2 siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, Pyk2, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as a control and Pyk2 siRNA, and pretreated without or with 100 nM edaravone for 1 h. They were then incubated with 25 μg/mL SiNPs for the indicated time intervals (15, 30, and 60 min). The protein levels of phospho-Pyk2, total Pyk2, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 5
<p><b>EGFR is required for SiNP-induced COX-2 expression and PGE<sub>2</sub> production in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of AG1478 (0.1, 1, and 10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) HPAEpiCs were pretreated with AG1478 (10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were analyzed by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled or EGFR siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, EGFR, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as a control and EGFR or Pyk2 siRNA, and then incubated with 25 μg/mL SiNPs for the indicated time intervals (15, 30, and 60 min). The protein levels of phospho-Pyk2, total Pyk2, phospho-EGFR, total EGFR, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 6
<p><b>PI3K/Akt signaling involvement in SiNP-induced COX-2 expression and PGE<sub>2</sub> secretion in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of LY294002 (0.3, 3, and 30 μM) or Akt inhibitor VIII (0.1, 1, and 10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) Cells were pretreated with LY294002 (30 μM) or Akt inhibitor VIII (10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were determined by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled, p110, or Akt siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, p110, Akt, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as a control, and p110, Akt, or EGFR siRNA, and then incubated with 25 μg/mL SiNPs for the indicated time intervals (90, 120, and 150 min). The protein levels of phospho-Akt, total Akt, phospho-EGFR, total EGFR, total p110, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 7
<p><b>p38 MAPK involvement in SiNP-induced COX-2 expression and PGE<sub>2</sub> production in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of p38 MAPK inhibitor VIII (0.1, 1, and 10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) HPAEpiCs were pretreated with p38 MAPK inhibitor VIII (10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were analyzed by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled or p38 siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, p38, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as control, and p38 siRNA or Akt siRNA, and then incubated with 25 μg/mL SiNPs for the indicated time intervals (90, 120, and 150 min). The protein levels of phospho-Akt, total Akt, phospho-p38, total p38, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 8
<p><b>JNK1/2 contributes to SiNP-induced COX-2 expression and PGE<sub>2</sub> production in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of SP600125 (0.1, 1, and 10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) HPAEpiCs were pretreated with SP600125 (10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were analyzed by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled or JNK2 siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, JNK2, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as control, and JNK2 or Akt siRNA, and then incubated with 25 μg/mL SiNPs for the indicated time intervals (90, 120, and 150 min). The protein levels of phospho-Akt, total Akt, phospho-JNK1/2, total JNK1/2, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 9
<p><b>Involvement of FoxO1 in SiNP-induced COX-2 expression and PGE<sub>2</sub> production in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of AS1842856 (0.1, 1, and 10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) Cells were pretreated with AS1842856 (10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were determined by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled or FoxO1 siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, FoxO1, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as control, and FoxO1, JNK2, or p38 siRNA, and then incubated with 25 μg/mL SiNPs for the indicated time intervals (90, 120, and 150 min). The protein levels of phospho-FoxO1, total FoxO1, phospho-JNK1/2, total JNK1/2, phospho-p38, total p38, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 10
<p><b>c-Jun/AP-1 regulates COX-2 expression and PGE<sub>2</sub> production induced by SiNPs in HPAEpiCs.</b> (<b>A</b>) HPAEpiCs were pretreated with various concentrations of Tanshinone IIA (0.1, 1, and 10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 12 h. The protein expression of COX-2 and GAPDH was determined by Western blot. (<b>B</b>) Cells were pretreated with Tanshinone IIA (10 μM) for 1 h, and then incubated with 25 μg/mL SiNPs for 6 h. The levels of COX-2 and GAPDH mRNA were determined by quantitative real-time PCR. (<b>C</b>) The cells were transfected with scrambled or c-Jun siRNA, and then incubated with 25 μg/mL SiNPs for 12 h. The protein levels of COX-2, c-Jun, and GAPDH were analyzed by Western blot. (<b>D</b>) The cells were transfected with scrambled siRNA as control, and c-Jun siRNA, JNK2 siRNA, or p38 siRNA, and then incubated with 25 μg/mL SiNPs for the indicated time intervals (90, 120, and 150 min). The protein levels of phospho-c-Jun, total c-Jun, phospho-JNK1/2, total JNK1/2, phospho-p38, total p38, and GAPDH were analyzed by Western blot. (<b>E</b>) The culture media from (<b>A</b>) were collected to determine the levels of PGE<sub>2</sub> synthesis using a PGE<sub>2</sub> ELISA kit. Data are presented as mean ± S.E.M. from at least three independent experiments as indicated on each panel. # <span class="html-italic">p</span> &lt; 0.01, as compared between the two indicated groups.</p>
Full article ">Figure 11
<p><b>Schematic signaling pathways contributing to SiNP-induced COX-2 expression and PGE<sub>2</sub> production in HPAEpiCs.</b> SiNP-induced up-regulation of COX-2 and synthesis of PGE<sub>2</sub> are, at least in part, mediated through ROS accumulation, which leads to the transactivation of EGFR by Pyk2. Subsequently, the activation of the PI3K/Akt pathway is triggered by EGFR, resulting in the activation and translocation of c-Jun and FoxO1 from the cytosol to the nucleus in a p38 MAPK- and JNK1/2-dependent manner. The activation of AP-1 and FoxO1 contributes to the increased levels of COX-2 and PGE<sub>2</sub> induced by SiNPs. Ultimately, the COX-2/PGE<sub>2</sub> axis may promote inflammatory responses in HPAEpiCs.</p>
Full article ">
13 pages, 7445 KiB  
Article
Chlorogenic Acid Alleviates LPS-Induced Inflammation and Oxidative Stress by Modulating CD36/AMPK/PGC-1α in RAW264.7 Macrophages
by Tiantian Gu, Zhiguo Zhang, Jinyu Liu, Li Chen, Yong Tian, Wenwu Xu, Tao Zeng, Weicheng Wu and Lizhi Lu
Int. J. Mol. Sci. 2023, 24(17), 13516; https://doi.org/10.3390/ijms241713516 - 31 Aug 2023
Cited by 11 | Viewed by 2011
Abstract
Chlorogenic acid (CGA) is a bioactive substance with anti-inflammatory activities. Clusters of CD36 have been suggested to be widely involved in inflammatory damage. However, the mechanism of CGA protecting against LPS-induced inflammation involving the CD36 regulation is unclear. Here, we demonstrated that CGA [...] Read more.
Chlorogenic acid (CGA) is a bioactive substance with anti-inflammatory activities. Clusters of CD36 have been suggested to be widely involved in inflammatory damage. However, the mechanism of CGA protecting against LPS-induced inflammation involving the CD36 regulation is unclear. Here, we demonstrated that CGA protected against LPS-induced cell death and decreased the production of ROS. Moreover, the SOD, CAT, and GSH-Px activities were also upregulated in CGA-treated cells during LPS stimulation. CGA reduced COX-2 and iNOS expression and IL-1β, IL-6, and TNF-α secretion in LPS-stimulated RAW264.7 macrophages. In addition, CGA treatment widely involved in immune-related signaling pathways, including NF-κB signaling, NOD-like receptor signaling, and IL-17 signaling using transcriptomic analysis and CD36 also markedly reduced during CGA pretreatment in LPS-induced RAW264.7 cells. Furthermore, the CD36 inhibitor SSO attenuated inflammation and oxidative stress by enabling activation of the AMPK/PGC-1α cascade. These results indicate that CGA might provide benefits for the regulation of inflammatory diseases by modulating CD36/AMPK/PGC-1α to alleviate oxidative stress. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Figure 1

Figure 1
<p>Effect of chlorogenic acid (CGA) on cell proliferation. (<b>A</b>) The chemical structure of CGA. (<b>B</b>) CCK-8 assays of cell proliferation. Results are shown as the mean ± SD (<span class="html-italic">n</span> = 6). Different letters indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 2
<p>CGA reduced LPS-induced oxidative stress. (<b>A</b>) ROS production. (<b>B</b>) The activity of SOD. (<b>C</b>) The activity of CAT activity, and (<b>D</b>) the activity of GSH-Px activity in different groups. Results are shown as the mean ± SD (<span class="html-italic">n</span> = 3). Different letters indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 3
<p>CGA reduced LPS-induced inflammatory mediators and cytokines expression. (<b>A</b>,<b>B</b>) The secretion of IL-6 and TNF-α. (<b>C</b>–<b>G</b>) mRNA level and (<b>H</b>,<b>I</b>) protein expression of IL-6, TNF-α, IL-1β, COX-2, and iNOS. Results are shown as the mean ± SD (<span class="html-italic">n</span> = 3). Different letters indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 4
<p>Transcriptomic analysis reveals that CGA alleviates LPS-induced inflammation and oxidative stress by gene regulation. (<b>A</b>) A heatmap of differentially expressed genes. (<b>B</b>) Correlation analysis. (<b>C</b>) Principal component analysis of the samples of mock−treated, LPS−treated, and CGA+LPS−treated cells. (<b>D</b>) Gene ontology analysis results. (<b>E</b>) KEGG analysis results. (<b>F</b>) qPCR validation of selected genes. Different letters (a, b, c) indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05). Results are shown as the mean ± SD (<span class="html-italic">n</span> = 3). Different letters and * indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 5
<p>CGA treatment inhibited CD36 expression. (<b>A</b>) A Venn analysis of mock-treated vs. LPS−treated cells and LPS−treated vs. LPS+CGA−treated cells with 613 shared DEGs with opposite trends selected. (<b>B</b>) Protein–protein interaction analysis of CD36. Red indicated <span class="html-italic">Cd36</span>. (<b>C</b>–<b>E</b>) The protein expression of CD36, phosphorylated AMPK, and PGC-1α. Results are shown as the mean ± SD (<span class="html-italic">n</span> = 3). Different letters indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 6
<p>CD36 inhibition defended anti-inflammatory and oxidative stress functions through the CD36/AMPK/PGC-1α cascade. (<b>A</b>) The phosphorylated AMPKα and PGC-1α protein expression. (<b>B</b>) ROS levels. (<b>C</b>–<b>E</b>) Oxidative stress parameters (SOD, CAT, and GSH-Px activities). (<b>F</b>,<b>G</b>) Inflammatory cytokine (IL-6 and TNF-α) expression. (<b>H</b>) Inflammatory mediators and cytokines (IL-1β, IL-6, TNF-α, COX-2, and iNOS) protein expression. Results are shown as the mean ± SD (<span class="html-italic">n</span> = 3). Different letters indicate significant differences between groups (<span class="html-italic">p</span> &lt; 0.05).</p>
Full article ">Figure 7
<p>CGA reduces LPS−induced oxidative stress and inflammation by activating the CD36/AMPK/PGC-1α pathway.</p>
Full article ">
16 pages, 1024 KiB  
Review
Antioxidant Therapy as an Effective Strategy against Noise-Induced Hearing Loss: From Experimental Models to Clinic
by Anna Pisani, Fabiola Paciello, Raffaele Montuoro, Rolando Rolesi, Jacopo Galli and Anna Rita Fetoni
Life 2023, 13(4), 1035; https://doi.org/10.3390/life13041035 - 17 Apr 2023
Cited by 6 | Viewed by 4734
Abstract
Cochlear redox unbalance is the main mechanism of damage involved in the pathogenesis of noise-induced-hearing loss. Indeed, the increased free radical production, in conjunction with a reduced efficacy of the endogenous antioxidant system, plays a key role in cochlear damage induced by noise [...] Read more.
Cochlear redox unbalance is the main mechanism of damage involved in the pathogenesis of noise-induced-hearing loss. Indeed, the increased free radical production, in conjunction with a reduced efficacy of the endogenous antioxidant system, plays a key role in cochlear damage induced by noise exposure. For this reason, several studies focused on the possibility to use exogenous antioxidant to prevent or attenuate noise-induce injury. Thus, several antioxidant molecules, alone or in combination with other compounds, have been tested in both experimental and clinical settings. In our findings, we tested the protective effects of several antioxidant enzymes, spanning from organic compounds to natural compounds, such as nutraceuticals of polyphenols. In this review, we summarize and discuss the strengths and weaknesses of antioxidant supplementation focusing on polyphenols, Q-Ter, the soluble form of CoQ10, Vitamin E and N-acetil-cysteine, which showed great otoprotective effects in different animal models of noise induced hearing loss and which has been proposed in clinical trials. Full article
(This article belongs to the Topic Oxidative Stress and Inflammation, 2nd Volume)
Show Figures

Graphical abstract

Graphical abstract
Full article ">Figure 1
<p>Mechanisms of cochlear oxidative damage in NIHL. (<b>A</b>) schematic representation showing the damaging effects of ROS production, including mitochondrial damage, apoptotic pathway activation leading to cell death, lipid peroxidation and reducing endogenous antioxidants defense. O<sub>2</sub>: Superoxide; <sup>1</sup>O<sub>2</sub>: Singlet oxygen; H<sub>2</sub>O<sub>2</sub>: hydrogen peroxide; SOD: superoxide dismutase; HO-1: heme-oxygenase-1; GSH: glutathione; Nrf2: nuclear factor erythroid 2–related factor 2. (<b>B</b>) Schematic representation of the Organ of Corti showing the sensory cells (hair cells), consisting of three rows of outer hair cells (OHCs) and one row of inner hair cell (IHCs), and the neuronal afferent fibers (AF) forming synaptic contact with both OHCs and IHCs. Noise exposure induced a drastic increase of ROS production, leading to redox imbalance and decreasing the endogenous antioxidant defenses.</p>
Full article ">Figure 2
<p>Schematic protective effect of antioxidants compounds against cochlear ROS increased amount induced by noise exposure. Q10 soluble form (Q-Ter), N-acetil-cysteine (NAC) and Vitamin E (VIT E) act directly as ROS scavenging molecules. Phenolic compounds (ferulic acid—FA, rosmarinic acid—RA, caffeic acid—CA) act indirectly by activating the redox-sensitive factor Nrf2. By reestablishing cochlear redox balance, antioxidant protection can counteract molecular mechanisms underlying hair cell dysfunction, attenuating cochlear damage, and restoring hearing loss.</p>
Full article ">
Back to TopTop