51
|
Vassilopoulou E, Guibas GV, Papadopoulos NG. Mediterranean-Type Diets as a Protective Factor for Asthma and Atopy. Nutrients 2022; 14:1825. [PMID: 35565792 PMCID: PMC9105881 DOI: 10.3390/nu14091825] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 04/25/2022] [Accepted: 04/26/2022] [Indexed: 12/25/2022] Open
Abstract
We are currently riding the second wave of the allergy epidemic, which is ongoing in affluent societies, but now also affecting developing countries. This increase in the prevalence of atopy/asthma in the Western world has coincided with a rapid improvement in living conditions and radical changes in lifestyle, suggesting that this upward trend in allergic manifestations may be associated with cultural and environmental factors. Diet is a prominent environmental exposure that has undergone major changes, with a substantial increase in the consumption of processed foods, all across the globe. On this basis, the potential effects of dietary habits on atopy and asthma have been researched rigorously, but even with a considerable body of evidence, clear associations are far from established. Many factors converge to obscure the potential relationship, including methodological, pathophysiological and cultural differences. To date, the most commonly researched, and highly promising, candidate for exerting a protective effect is the so-called Mediterranean diet (MedDi). This dietary pattern has been the subject of investigation since the mid twentieth century, and the evidence regarding its beneficial health effects is overwhelming, although data on a correlation between MedDi and the incidence and severity of asthma and atopy are inconclusive. As the prevalence of asthma appears to be lower in some Mediterranean populations, it can be speculated that the MedDi dietary pattern could indeed have a place in a preventive strategy for asthma/atopy. This is a review of the current evidence of the associations between the constituents of the MedDi and asthma/atopy, with emphasis on the pathophysiological links between MedDi and disease outcomes and the research pitfalls and methodological caveats which may hinder identification of causality. MedDi, as a dietary pattern, rather than short-term supplementation or excessive focus on single nutrient effects, may be a rational option for preventive intervention against atopy and asthma.
Collapse
Affiliation(s)
- Emilia Vassilopoulou
- Department of Nutritional Sciences and Dietetics, International Hellenic University, 57400 Thessaloniki, Greece
| | - George V. Guibas
- Department of Allergy and Clinical Immunology, Royal Preston Hospital, Lancashire Teaching Hospitals NHS Foundation Trust, Preston PR2 9HT, UK;
- School of Biological Sciences, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
| | - Nikolaos G. Papadopoulos
- School of Biological Sciences, Medicine and Health, University of Manchester, Manchester M13 9PL, UK;
- Allergy Department, 2nd Pediatric Clinic, National and Kapodistrian University of Athens, Thivon and Levadias 1, 11527 Athens, Greece
| |
Collapse
|
52
|
GlyNAC (Glycine and N-Acetylcysteine) Supplementation in Mice Increases Length of Life by Correcting Glutathione Deficiency, Oxidative Stress, Mitochondrial Dysfunction, Abnormalities in Mitophagy and Nutrient Sensing, and Genomic Damage. Nutrients 2022; 14:nu14051114. [PMID: 35268089 PMCID: PMC8912885 DOI: 10.3390/nu14051114] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/02/2022] [Accepted: 03/03/2022] [Indexed: 02/01/2023] Open
Abstract
Determinants of length of life are not well understood, and therefore increasing lifespan is a challenge. Cardinal theories of aging suggest that oxidative stress (OxS) and mitochondrial dysfunction contribute to the aging process, but it is unclear if they could also impact lifespan. Glutathione (GSH), the most abundant intracellular antioxidant, protects cells from OxS and is necessary for maintaining mitochondrial health, but GSH levels decline with aging. Based on published human studies where we found that supplementing glycine and N-acetylcysteine (GlyNAC) improved/corrected GSH deficiency, OxS and mitochondrial dysfunction, we hypothesized that GlyNAC supplementation could increase longevity. We tested our hypothesis by evaluating the effect of supplementing GlyNAC vs. placebo in C57BL/6J mice on (a) length of life; and (b) age-associated GSH deficiency, OxS, mitochondrial dysfunction, abnormal mitophagy and nutrient-sensing, and genomic-damage in the heart, liver and kidneys. Results showed that mice receiving GlyNAC supplementation (1) lived 24% longer than control mice; (2) improved/corrected impaired GSH synthesis, GSH deficiency, OxS, mitochondrial dysfunction, abnormal mitophagy and nutrient-sensing, and genomic-damage. These studies provide proof-of-concept that GlyNAC supplementation can increase lifespan and improve multiple age-associated defects. GlyNAC could be a novel and simple nutritional supplement to improve lifespan and healthspan, and warrants additional investigation.
Collapse
|
53
|
Nucera F, Mumby S, Paudel KR, Dharwal V, DI Stefano A, Casolaro V, Hansbro PM, Adcock IM, Caramori G. Role of oxidative stress in the pathogenesis of COPD. Minerva Med 2022; 113:370-404. [PMID: 35142479 DOI: 10.23736/s0026-4806.22.07972-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Chronic inhalation of cigarette smoke is a prominent cause of chronic obstructive pulmonary disease (COPD) and provides an important source of exogenous oxidants. In addition, several inflammatory and structural cells are a source of endogenous oxidants in the lower airways of COPD patients, even in former smokers. This suggests that oxidants play a key role in the pathogenesis of COPD. This oxidative stress is counterbalanced by the protective effects of the various endogenous antioxidant defenses of the lower airways. A large amount of data from animal models and patients with COPD have shown that both the stable phase of the disease, and during exacerbations, have increased oxidative stress in the lower airways compared with age-matched smokers with normal lung function. Thus, counteracting the increased oxidative stress may produce clinical benefits in COPD patients. Smoking cessation is currently the most effective treatment of COPD patients and reduces oxidative stress in the lower airways. In addition, many drugs used to treat COPD have some antioxidant effects, however, it is still unclear if their clinical efficacy is related to pharmacological modulation of the oxidant/antioxidant balance. Several new antioxidant compounds are in development for the treatment of COPD.
Collapse
Affiliation(s)
- Francesco Nucera
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy -
| | - Sharon Mumby
- Airways Diseases Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, UK
| | - Keshav R Paudel
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Vivek Dharwal
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Antonino DI Stefano
- Divisione di Pneumologia e Laboratorio di Citoimmunopatologia dell'Apparato Cardio Respiratorio, Istituti Clinici Scientifici Maugeri, IRCCS, Veruno, Novara, Italy
| | - Vincenzo Casolaro
- Department of Medicine, Surgery and Dentistry Scuola Medica Salernitana, University of Salerno, Salerno, Italy
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute and University of Technology Sydney, School of Life Sciences, Faculty of Science, Sydney, Australia
| | - Ian M Adcock
- Airways Diseases Section, Faculty of Medicine, Imperial College London, National Heart and Lung Institute, London, UK
| | - Gaetano Caramori
- Pneumologia, Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali (BIOMORF), Università degli Studi di Messina, Messina, Italy
| |
Collapse
|
54
|
Kaloumenou M, Skotadis E, Lagopati N, Efstathopoulos E, Tsoukalas D. Breath Analysis: A Promising Tool for Disease Diagnosis-The Role of Sensors. SENSORS (BASEL, SWITZERLAND) 2022; 22:1238. [PMID: 35161984 PMCID: PMC8840008 DOI: 10.3390/s22031238] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 01/30/2022] [Accepted: 02/01/2022] [Indexed: 05/07/2023]
Abstract
Early-stage disease diagnosis is of particular importance for effective patient identification as well as their treatment. Lack of patient compliance for the existing diagnostic methods, however, limits prompt diagnosis, rendering the development of non-invasive diagnostic tools mandatory. One of the most promising non-invasive diagnostic methods that has also attracted great research interest during the last years is breath analysis; the method detects gas-analytes such as exhaled volatile organic compounds (VOCs) and inorganic gases that are considered to be important biomarkers for various disease-types. The diagnostic ability of gas-pattern detection using analytical techniques and especially sensors has been widely discussed in the literature; however, the incorporation of novel nanomaterials in sensor-development has also proved to enhance sensor performance, for both selective and cross-reactive applications. The aim of the first part of this review is to provide an up-to-date overview of the main categories of sensors studied for disease diagnosis applications via the detection of exhaled gas-analytes and to highlight the role of nanomaterials. The second and most novel part of this review concentrates on the remarkable applicability of breath analysis in differential diagnosis, phenotyping, and the staging of several disease-types, which are currently amongst the most pressing challenges in the field.
Collapse
Affiliation(s)
- Maria Kaloumenou
- Department of Applied Physics, National Technical University of Athens, 15780 Athens, Greece; (M.K.); (D.T.)
| | - Evangelos Skotadis
- Department of Applied Physics, National Technical University of Athens, 15780 Athens, Greece; (M.K.); (D.T.)
| | - Nefeli Lagopati
- Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., Goudi, 11527 Athens, Greece; (N.L.); (E.E.)
| | - Efstathios Efstathopoulos
- Medical School, National and Kapodistrian University of Athens, 75, Mikras Asias Str., Goudi, 11527 Athens, Greece; (N.L.); (E.E.)
| | - Dimitris Tsoukalas
- Department of Applied Physics, National Technical University of Athens, 15780 Athens, Greece; (M.K.); (D.T.)
| |
Collapse
|
55
|
Xin L, Sun J, Zhai X, Chen X, Wan J, Tian H. Repeated radon exposure induced lung damage via oxidative stress-mediated mitophagy in human bronchial epithelial cells and mice. ENVIRONMENTAL TOXICOLOGY AND PHARMACOLOGY 2022; 90:103812. [PMID: 35033684 DOI: 10.1016/j.etap.2022.103812] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 01/06/2022] [Accepted: 01/11/2022] [Indexed: 06/14/2023]
Abstract
This study aimed to investigate the potential molecular mechanism underlying radon-induced lung damage. Our results showed that long-term radon exposure induced mitochondrial damage and redox imbalance in BEAS-2B cells and a time-dependent lung pathological injury in mice. The activation of Nrf-2 and its down-stream antioxidants, and the gene expression of the indicated markers at different stages of autophagy were found to be induced with the increasing of radon exposure time. Changes in the gene expression of PINK-1, Parkin, and p62 induced by radon showed differences in mechanisms of mitophagy activation and profiles of autophagic flux between BEAS-2B cells and mice. Our findings not only demonstrated that long-term radon exposure induced damages to bronchial epithelial cells and the mice lung through increasing oxidative stress, decreasing mitochondrial function and activating mitophagy with different profiles of autophagic flux, but also revealed Nrf-2 as a central regulator of mitochondrial homeostasis and lung damage.
Collapse
Affiliation(s)
- Lili Xin
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Jiaojiao Sun
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Xuedi Zhai
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Xiaoyu Chen
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Jianmei Wan
- Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China
| | - Hailin Tian
- School of Public Health, Medical College of Soochow University, 199 Renai Road, Suzhou 215123, Jiangsu, China.
| |
Collapse
|
56
|
Evaluation of Thiol/Disulfide Homeostasis in Bronchiectasis. Can Respir J 2022; 2022:8340450. [PMID: 35132344 PMCID: PMC8817889 DOI: 10.1155/2022/8340450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 12/14/2021] [Accepted: 01/13/2022] [Indexed: 11/25/2022] Open
Abstract
Purpose Thiols are sulfhydryl-containing organic compounds that have an important role in preventing cellular oxidative stress. This study compares the blood oxidative stress marker levels in bronchiectasis cases during their stable periods with healthy controls. Materials and Methods Seventy-seven patients (49 patients with stable bronchiectasis/28 healthy controls), followed up by the chest disease clinic, were included in the study. Peripheral blood thiol-disulfide parameters (NT: native thiol (−SH); TT: total thiol (−SH + SS); SS: disulfide (−SS); SS-SH: disulfide/native thiol index; SS-TT: disulphide/total thiol index; SH-TT: native thiol/total thiol index), and ischemia-modified albumin (IMA) levels were examined in the stable bronchiectasis group and the control group. Thiol-disulfide homeostasis was evaluated using a novel and automated assay. Findings and Result. Blood native thiol levels in patients with stable bronchiectasis were found to be significantly higher compared with healthy controls. A positive correlation between the total airway disease score and IMA levels was present. Our findings revealed that native thiol levels, which constitute a part of the antioxidant defense system, are increased in patients with stable bronchiectasis.
Collapse
|
57
|
Zhang WT, Lu Q, Ding JJ, Gu M. Association of asymmetric dimethylarginine with the pathological process of persistent pulmonary hypertension of the newborn. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2022; 24:54-59. [PMID: 35177176 PMCID: PMC8802391 DOI: 10.7499/j.issn.1008-8830.2108145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Accepted: 12/01/2021] [Indexed: 06/14/2023]
Abstract
OBJECTIVES To study the change in asymmetric dimethylarginine (ADMA) in the circulation system of full-term infants with persistent pulmonary hypertension of the newborn (PPHN) and its association with treatment response, as well as the possibility of ADMA as a therapeutic target and a marker for treatment response. METHODS A prospective study was performed. A total of 30 full-term neonates who were diagnosed with PPHN within 3 days after birth were enrolled as the PPHN group, and the neonates without PPHN, matched for gestational age and age, who were treated or observed in the department of neonatology were enrolled as the control group. Serum samples were collected on days 1, 7, and 14 of treatment. The high-performance liquid chromatography-tandem mass spectrometry was used to measure the serum concentrations of L-arginine, ADMA, and its isomer symmetric dimethylarginine (SDMA). RESULTS For the neonates in the control group, the serum concentrations of ADMA and L-arginine continuously increased and the serum concentration of SDMA continuously decreased within the first 14 days of treatment. On days 1 and 14, there was no significant difference in the serum concentration of ADMA between the control and PPHN groups (P>0.05). On day 7, the PPHN group had a significantly higher serum concentration of ADMA than the control group (P<0.05), while there were no significant differences in serum concentrations of SDMA or L-arginine (P>0.05). Moreover, after 7 days of treatment, the PPHN neonates with a systolic pulmonary arterial pressure (sPAP) of >35 mmHg had a significantly higher serum concentration of ADMA than those with an sPAP of ≤35 mm Hg. CONCLUSIONS There are continuous increases in the ADMA concentration and the ADMA/SDMA ratio in the circulation system of full-term infants within the first 2 weeks after birth, and this process is accelerated by the pathological process of PPHN, suggesting that ADMA may be involved in the pathologic process of PPHN. A high level of ADMA is associated with the resistance to PPHN treatment, suggesting that inhibition of ADMA might be a potential target of drug intervention to improve the treatment response of PPHN.
Collapse
Affiliation(s)
- Wen-Ting Zhang
- Department of Pediatric Laboratory, Changzhou Children's Hospital Affiliated to Nantong University, Changzhou, Jiangsu 213003, China
| | | | - Jie-Jun Ding
- Department of Pediatric Laboratory, Changzhou Children's Hospital Affiliated to Nantong University, Changzhou, Jiangsu 213003, China
| | - Meng Gu
- Department of Pediatric Laboratory, Changzhou Children's Hospital Affiliated to Nantong University, Changzhou, Jiangsu 213003, China
| |
Collapse
|
58
|
Lan H, Gui Z, Zeng Z, Li D, Qian B, Qin LY, Dai L, Song JL. Oral administration of Lactobacillus plantarum CQPC11 attenuated the airway inflammation in an ovalbumin (OVA)-induced Balb/c mouse model of asthma. J Food Biochem 2022; 46:e14036. [PMID: 34981513 DOI: 10.1111/jfbc.14036] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2021] [Revised: 11/14/2021] [Accepted: 11/18/2021] [Indexed: 12/11/2022]
Abstract
This study investigated the antiasthmatic and anti-inflammatory effects of Lactobacillus plantarum-CQPC11 (LP-CQPC11) on ovalbumin (OVA)-induced asthmatic Balb/c mice. Administration of different doses of LP-CQPC11 (105 , 107 , and 109 colony-forming unit [CFU]/mouse) effectively reduced airway hyperresponsiveness (AHR) and the lung W/D ratio in asthmatic mice. LP-CQPC11 treatment reduced the accumulation of inflammatory cells in the BALF and attenuated histologic edema in asthmatic mice. Administration of LP-CQPC11 decreased the serum levels of OVA-specific IgE, IgE, and OVA-specific IgG1. LP-CQPC11 treatment decreased the levels of inflammatory cytokines (TNF-α, IL-4, IL-13, IL-5, and IL-6) in the BALF of asthmatic mice. In addition, LP-CQPC11 also elevated the mRNA levels of Foxp3 and T-bet and decreased the mRNA levels of Gata3 and RORγt in asthmatic mice lungs. Administration of LP-CQPC11 also reduced OVA-induced oxidative stress by improving the activities of GSH-Px, SOD, and catalase in the lungs. Finally, LP-CQPC11 treatment also significantly decreased the activation of the NF-κB pathway to modulate the inflammatory reaction in the lungs of asthmatic mice. The results from this study clearly demonstrated that oral administration of LP-CQPC11 exhibited outstanding activity in attenuating OVA-induced asthma in a mouse model. Furthermore, LP-CQPC11 may be an effective microecologic agent in preventing allergic asthma in the future. PRACTICAL APPLICATIONS: Allergic asthma is a common chronic inflammation-associated respiratory disease. Lactic acid bacteria (LAB) are known as a health product involved in modulating immune tolerance and play important roles in disease prevention and treatment. Many studies have reported that LAB, as probiotics, exhibits great antioxidation, anticancer, and anti-inflammatory activities and have health benefits in gastrointestinal disorders. In fact, human studies have confirmed that Lactobacillus rhamnosus strains have an effective activity to reduce the risk of allergic asthma. LP-CQPC11 was isolated from Sichuan pickled cabbages (a type of LAB-fermented vegetable product, also called Sichuan paocai) and was reported to reduce d-galactose-induced aging in mice in our previous study. However, the antiasthmatic and anti-inflammatory activities of LP-CQPC11 are unclear. The current study investigated the antiasthmatic and anti-inflammatory effects of LP-CQPC11 on OVA-induced asthmatic Balb/c mice.
Collapse
Affiliation(s)
- Huan Lan
- Department of Analytical Chemistry & Drug Analysis, College of Pharmacy, Guilin Medical University, Guilin, China
| | - Zhongyu Gui
- Department of Nutrition and Food Hygiene, College of Public Health, Guilin Medical University, Guilin, China.,Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, Guilin Medical University, Guilin, China.,Guangxi Key Laboratory of Environmental Exposureomics and Entire Lifecycle Health, Guilin Medical University, Guilin, China
| | - Zhen Zeng
- Department of Nutrition and Food Hygiene, College of Public Health, Guilin Medical University, Guilin, China.,Department of Pediatrics and Maternal and Child Health, Xiangya College of Public Health, Central South University, Changsha, China
| | - Dayu Li
- Department of Parasitology, College of Basic Medicine, Guilin Medical University, Guilin, China
| | - Bo Qian
- Department of Nutrition and Food Hygiene, College of Public Health, Guilin Medical University, Guilin, China.,Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, Guilin Medical University, Guilin, China.,Guangxi Key Laboratory of Environmental Exposureomics and Entire Lifecycle Health, Guilin Medical University, Guilin, China
| | - Lin-Yuan Qin
- Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, Guilin Medical University, Guilin, China.,Guangxi Key Laboratory of Environmental Exposureomics and Entire Lifecycle Health, Guilin Medical University, Guilin, China.,Department of Epidemiology and Statistics, College of Public Health, Guilin Medical University, Guilin, China
| | - Ling Dai
- Center of Mental Health Education and Counseling, Guilin Medical University, Guilin, China
| | - Jia-Le Song
- Department of Nutrition and Food Hygiene, College of Public Health, Guilin Medical University, Guilin, China.,Guangxi Health Commission Key Laboratory of Entire Lifecycle Health and Care, Guilin Medical University, Guilin, China.,Guangxi Key Laboratory of Environmental Exposureomics and Entire Lifecycle Health, Guilin Medical University, Guilin, China.,Department of Clinical Nutrition, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
| |
Collapse
|
59
|
Yamamoto A, Sly PD, Henningham A, Begum N, Yeo AJ, Fantino E. Redox Homeostasis in Well-differentiated Primary Human Nasal Epithelial Cells. JOURNAL OF CELLULAR SIGNALING 2022; 3:193-206. [PMID: 36777036 PMCID: PMC9912202 DOI: 10.33696/signaling.3.083] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Oxidative stress (OS) in the airway epithelium is associated with inflammation, cell damage, and mitochondrial dysfunction that may initiate or worsen respiratory disease. Redox regulation maintains the equilibrium of pro-oxidant/antioxidant reactions but can be disturbed by environmental exposures. The mechanism(s) underlying the induction and impact of OS on airway epithelium and how these influences on respiratory disease is poorly understood. The aim of this study was to develop a stress response model in primary human nasal epithelial cells (NECs) grown at the air-liquid interface (ALI) into a well-differentiated epithelium and to use this model to investigate the mechanisms underlying OS. Hydrogen peroxide (H2O2) was used to induce acute OS and the responses were measured with trans epithelial electrical resistance (TEER), membrane permeability, cell death (LDH release), mitochondrial reactive oxygen species (mtROS) generation, redox status (GSH/GSSG ratio), cellular ATP, and signaling pathways (SIRT1, FOXO3, p53, p21, PINK1, PARKIN, NRF2). Following 25 mM (sensitive) or 50mM (resistant) H2O2 exposure, cell integrity decreased (p<0.05), GSH/GSSG ratio reduced (p<0.05), and ATP production declined by 83% (p<0.05) in the sensitive and 55% (p<0.05) in the resistant group; mtROS production increased 3.4-fold (p<0.001). Significant inter-individual differences between healthy humans with regards to susceptibility to OS, and differential activation of various pathways (FOXO3, PARKIN) were observed. These intra-individual differences in susceptibility to OS may be attributed to resistant individuals having more mitochondria or greater mitochondrial function.
Collapse
Affiliation(s)
- Ayaho Yamamoto
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia,Correspondence should be addressed to Ayaho Yamamoto,
| | - Peter D. Sly
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| | - Anna Henningham
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| | - Nelufa Begum
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| | - Abrey J. Yeo
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia,The University of Queensland Centre for Clinical Research, Herston, Queensland 4029, Australia
| | - Emmanuelle Fantino
- Child Health Research Centre, The University of Queensland, South Brisbane, Queensland 4101, Australia
| |
Collapse
|
60
|
Severe Glutathione Deficiency, Oxidative Stress and Oxidant Damage in Adults Hospitalized with COVID-19: Implications for GlyNAC (Glycine and N-Acetylcysteine) Supplementation. Antioxidants (Basel) 2021; 11:antiox11010050. [PMID: 35052554 PMCID: PMC8773164 DOI: 10.3390/antiox11010050] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 12/21/2021] [Accepted: 12/21/2021] [Indexed: 12/15/2022] Open
Abstract
Humanity is battling a respiratory pandemic pneumonia named COVID-19 which has resulted in millions of hospitalizations and deaths. COVID-19 exacerbations occur in waves that continually challenge healthcare systems globally. Therefore, there is an urgent need to understand all mechanisms by which COVID-19 results in health deterioration to facilitate the development of protective strategies. Oxidative stress (OxS) is a harmful condition caused by excess reactive-oxygen species (ROS) and is normally neutralized by antioxidants among which Glutathione (GSH) is the most abundant. GSH deficiency results in amplified OxS due to compromised antioxidant defenses. Because little is known about GSH or OxS in COVID-19 infection, we measured GSH, TBARS (a marker of OxS) and F2-isoprostane (marker of oxidant damage) concentrations in 60 adult patients hospitalized with COVID-19. Compared to uninfected controls, COVID-19 patients of all age groups had severe GSH deficiency, increased OxS and elevated oxidant damage which worsened with advancing age. These defects were also present in younger age groups, where they do not normally occur. Because GlyNAC (combination of glycine and N-acetylcysteine) supplementation has been shown in clinical trials to rapidly improve GSH deficiency, OxS and oxidant damage, GlyNAC supplementation has implications for combating these defects in COVID-19 infected patients and warrants urgent investigation.
Collapse
|
61
|
Caldeira DDAF, Weiss DJ, Rocco PRM, Silva PL, Cruz FF. Mitochondria in Focus: From Function to Therapeutic Strategies in Chronic Lung Diseases. Front Immunol 2021; 12:782074. [PMID: 34887870 PMCID: PMC8649841 DOI: 10.3389/fimmu.2021.782074] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Accepted: 10/29/2021] [Indexed: 01/14/2023] Open
Abstract
Mitochondria are essential organelles for cell metabolism, growth, and function. Mitochondria in lung cells have important roles in regulating surfactant production, mucociliary function, mucus secretion, senescence, immunologic defense, and regeneration. Disruption in mitochondrial physiology can be the central point in several pathophysiologic pathways of chronic lung diseases such as chronic obstructive pulmonary disease, idiopathic pulmonary fibrosis, and asthma. In this review, we summarize how mitochondria morphology, dynamics, redox signaling, mitophagy, and interaction with the endoplasmic reticulum are involved in chronic lung diseases and highlight strategies focused on mitochondrial therapy (mito-therapy) that could be tested as a potential therapeutic target for lung diseases.
Collapse
Affiliation(s)
- Dayene de Assis Fernandes Caldeira
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Daniel J Weiss
- Department of Medicine, College of Medicine, University of Vermont, Burlington, VT, United States
| | - Patricia Rieken Macêdo Rocco
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Pedro Leme Silva
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| | - Fernanda Ferreira Cruz
- Laboratory of Pulmonary Investigation, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,National Institute of Science and Technology for Regenerative Medicine, Rio de Janeiro, Brazil.,Rio de Janeiro Innovation Network in Nanosystems for Health-NanoSAÚDE/FAPERJ, Rio de Janeiro, Brazil
| |
Collapse
|
62
|
Blunting p38 MAPKα and ERK1/2 activities by empagliflozin enhances the antifibrotic effect of metformin and augments its AMPK-induced NF-κB inactivation in mice intoxicated with carbon tetrachloride. Life Sci 2021; 286:120070. [PMID: 34688695 DOI: 10.1016/j.lfs.2021.120070] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/09/2021] [Accepted: 10/17/2021] [Indexed: 12/11/2022]
Abstract
AIM Metformin and empagliflozin combined therapy may have complementary effects that go beyond the well-recognized targets of their monotherapy through AMPK activation. Therefore, the current study was designed to investigate for the first time the hepatoprotective effects of such combination therapy in the carbon tetrachloride (CCl4)-induced hepatic fibrosis model in mice. MATERIALS AND METHODS Determination of liver enzymes and the liver content of oxidative stress parameters, and hydroxyproline were performed biochemically. ELISA was performed to measure PDGF-BB, TNF-α, TGF-β, TIMP-1, AMPK, p-mTOR, NF-κB P65 binding activity, p38 MAPKα, JNK1/2 and ERK1/2. Real-time qPCR was conducted to determine Col1a1 and α-SMA. In addition, histopathological examination using H&E and Masson's trichrome stain were performed for determination of histopathological changes. KEY FINDINGS Empagliflozin inhibited the activation of p38 MAPK and ERK1/2 and exhibited a weak AMPKα stimulation. On the other hand, metformin exerted a more robust stimulatory action on the AMPKα that was accompanied by a notable decrease in the NF-κB nuclear binding activity and a decline in the p-mTOR levels. Nevertheless, the effect of metformin on MAPK kinases was insignificant. Our results revealed that blunting p38 MAPKα and ERK1/2 activities by empagliflozin enhanced the antifibrotic effect of metformin and augmented its AMPK-induced NF-κB inactivation. SIGNIFICANCE As diabetes is one of the most common risk factors for liver fibrosis, the use of antidiabetic drugs is expected to improve therapeutic outcome. Therefore, metformin/empagliflozin combined therapy could be promising in preventing hepatic inflammation and fibrosis via exhibiting complementary effects particularly in diabetic patients.
Collapse
|
63
|
Schiffers C, Reynaert NL, Wouters EFM, van der Vliet A. Redox Dysregulation in Aging and COPD: Role of NOX Enzymes and Implications for Antioxidant Strategies. Antioxidants (Basel) 2021; 10:antiox10111799. [PMID: 34829671 PMCID: PMC8615131 DOI: 10.3390/antiox10111799] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2021] [Revised: 11/04/2021] [Accepted: 11/06/2021] [Indexed: 12/23/2022] Open
Abstract
With a rapidly growing elderly human population, the incidence of age-related lung diseases such as chronic obstructive pulmonary disease (COPD) continues to rise. It is widely believed that reactive oxygen species (ROS) play an important role in ageing and in age-related disease, and approaches of antioxidant supplementation have been touted as useful strategies to mitigate age-related disease progression, although success of such strategies has been very limited to date. Involvement of ROS in ageing is largely attributed to mitochondrial dysfunction and impaired adaptive antioxidant responses. NADPH oxidase (NOX) enzymes represent an important enzyme family that generates ROS in a regulated fashion for purposes of oxidative host defense and redox-based signalling, however, the associations of NOX enzymes with lung ageing or age-related lung disease have to date only been minimally addressed. The present review will focus on our current understanding of the impact of ageing on NOX biology and its consequences for age-related lung disease, particularly COPD, and will also discuss the implications of altered NOX biology for current and future antioxidant-based strategies aimed at treating these diseases.
Collapse
Affiliation(s)
- Caspar Schiffers
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT 05405, USA; (C.S.); (E.F.M.W.)
- Ludwig Boltzmann Institute for Lung Health, 1140 Vienna, Austria
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, The Netherlands;
| | - Niki L. Reynaert
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, The Netherlands;
| | - Emiel F. M. Wouters
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT 05405, USA; (C.S.); (E.F.M.W.)
- Ludwig Boltzmann Institute for Lung Health, 1140 Vienna, Austria
- Department of Respiratory Medicine, NUTRIM School of Nutrition and Translational Research in Metabolism, Maastricht University Medical Center, 6211 LK Maastricht, The Netherlands;
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT 05405, USA; (C.S.); (E.F.M.W.)
- Correspondence:
| |
Collapse
|
64
|
Fois SS, Canu S, Fois AG. The Role of Oxidative Stress in Sarcoidosis. Int J Mol Sci 2021; 22:ijms222111712. [PMID: 34769145 PMCID: PMC8584035 DOI: 10.3390/ijms222111712] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2021] [Revised: 10/24/2021] [Accepted: 10/26/2021] [Indexed: 01/15/2023] Open
Abstract
Sarcoidosis is a rare, systemic inflammatory disease whose diagnosis and management can pose a challenge for clinicians and specialists. Scientific knowledge on the molecular pathways that drive its development is still lacking, with no standardized therapies available and insufficient strategies to predict patient outcome. In recent years, oxidative stress has been highlighted as an important factor in the pathogenesis of sarcoidosis, involving several enzymes and molecules in the mechanism of the disease. This review presents current data on the role of oxidative stress in sarcoidosis and its interaction with inflammation, as well as the application of antioxidative therapy in the disease.
Collapse
Affiliation(s)
- Sara Solveig Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy;
- Correspondence:
| | - Sara Canu
- Respiratory Diseases Operative Unit, University Hospital of Sassari, 07100 Sassari, Italy;
| | - Alessandro Giuseppe Fois
- Department of Medical, Surgical and Experimental Sciences, University of Sassari, Viale San Pietro 43, 07100 Sassari, Italy;
| |
Collapse
|
65
|
Michaeloudes C, Abubakar-Waziri H, Lakhdar R, Raby K, Dixey P, Adcock IM, Mumby S, Bhavsar PK, Chung KF. Molecular mechanisms of oxidative stress in asthma. Mol Aspects Med 2021; 85:101026. [PMID: 34625291 DOI: 10.1016/j.mam.2021.101026] [Citation(s) in RCA: 151] [Impact Index Per Article: 37.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 09/15/2021] [Indexed: 01/18/2023]
Abstract
The lungs are exposed to reactive oxygen species oxygen (ROS) produced as a result of inhalation of oxygen, as well as smoke and other air pollutants. Cell metabolism and the NADPH oxidases (Nox) generate low levels of intracellular ROS that act as signal transduction mediators by inducing oxidative modifications of histones, enzymes and transcription factors. Redox signalling is also regulated by localised production and sensing of ROS in mitochondria, the endoplasmic reticulum (ER) and inside the nucleus. Intracellular ROS are maintained at low levels through the action of a battery of enzymatic and non-enzymatic antioxidants. Asthma is a heterogeneous airway inflammatory disease with different immune endotypes; these include atopic or non-atopic Th2 type immune response associated with eosinophilia, or a non-Th2 response associated with neutrophilia. Airway remodelling and hyperresponsiveness accompany the inflammatory response in asthma. Over-production of ROS resulting from infiltrating immune cells, particularly eosinophils and neutrophils, and a concomitant impairment of antioxidant responses lead to development of oxidative stress in asthma. Oxidative stress is augmented in severe asthma and during exacerbations, as well as by air pollution and obesity, and causes oxidative damage of tissues promoting airway inflammation and hyperresponsiveness. Furthermore, deregulated Nox activity, mitochondrial dysfunction, ER stress and/or oxidative DNA damage, resulting from exposure to irritants, inflammatory mediators or obesity, may lead to redox-dependent changes in cell signalling. ROS play a central role in airway epithelium-mediated sensing, development of innate and adaptive immune responses, and airway remodelling and hyperresponsiveness. Nonetheless, antioxidant compounds have proven clinically ineffective as therapeutic agents for asthma, partly due to issues with stability and in vivo metabolism of these compounds. The compartmentalised nature of ROS production and sensing, and the role of ROS in homeostatic responses and in the action of corticosteroids and β2-adrenergic receptor agonists, adds another layer of complexity to antioxidant therapy development. Nox inhibitors and mitochondrial-targeted antioxidants are in clinical development for a number of diseases but they have not yet been investigated in asthma. A better understanding of the complex role of ROS in the pathogenesis of asthma will highlight new opportunities for more targeted and effective redox therapies.
Collapse
Affiliation(s)
- Charalambos Michaeloudes
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom.
| | - Hisham Abubakar-Waziri
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Ramzi Lakhdar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Katie Raby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Piers Dixey
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Ian M Adcock
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Sharon Mumby
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Pankaj K Bhavsar
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom
| | - Kian Fan Chung
- National Heart and Lung Institute, Imperial College London, London, United Kingdom; NIHR Imperial Biomedical Research Centre, United Kingdom; Royal Brompton & Harefield NHS Trust, London, UK
| |
Collapse
|
66
|
Mustafa Rizvi SH, Shao D, Tsukahara Y, Pimentel DR, Weisbrod RM, Hamburg NM, McComb ME, Matsui R, Bachschmid MM. Oxidized GAPDH transfers S-glutathionylation to a nuclear protein Sirtuin-1 leading to apoptosis. Free Radic Biol Med 2021; 174:73-83. [PMID: 34332079 PMCID: PMC8432375 DOI: 10.1016/j.freeradbiomed.2021.07.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/16/2021] [Accepted: 07/25/2021] [Indexed: 11/17/2022]
Abstract
AIMS S-glutathionylation is a reversible oxidative modification of protein cysteines that plays a critical role in redox signaling. Glutaredoxin-1 (Glrx), a glutathione-specific thioltransferase, removes protein S-glutathionylation. Glrx, though a cytosolic protein, can activate a nuclear protein Sirtuin-1 (SirT1) by removing its S-glutathionylation. Glrx ablation causes metabolic abnormalities and promotes controlled cell death and fibrosis in mice. Glyceraldehyde 3-phosphate dehydrogenase (GAPDH), a key enzyme of glycolysis, is sensitive to oxidative modifications and involved in apoptotic signaling via the SirT1/p53 pathway in the nucleus. We aimed to elucidate the extent to which S-glutathionylation of GAPDH and glutaredoxin-1 contribute to GAPDH/SirT1/p53 apoptosis pathway. RESULTS Exposure of HEK 293T cells to hydrogen peroxide (H2O2) caused rapid S-glutathionylation and nuclear translocation of GAPDH. Nuclear GAPDH peaked 10-15 min after the addition of H2O2. Overexpression of Glrx or redox dead mutant GAPDH inhibited S-glutathionylation and nuclear translocation. Nuclear GAPDH formed a protein complex with SirT1 and exchanged S-glutathionylation to SirT1 and inhibited its deacetylase activity. Inactivated SirT1 remained stably bound to acetylated-p53 and initiated apoptotic signaling resulting in cleavage of caspase-3. We observed similar effects in human primary aortic endothelial cells suggesting the GAPDH/SirT1/p53 pathway as a common apoptotic mechanism. CONCLUSIONS Abundant GAPDH with its highly reactive-cysteine thiolate may function as a cytoplasmic rheostat to sense oxidative stress. S-glutathionylation of GAPDH may relay the signal to the nucleus where GAPDH trans-glutathionylates nuclear proteins such as SirT1 to initiate apoptosis. Glrx reverses GAPDH S-glutathionylation and prevents its nuclear translocation and cytoplasmic-nuclear redox signaling leading to apoptosis. Our data suggest that trans-glutathionylation is a critical step in apoptotic signaling and a potential mechanism that cytosolic Glrx controls nuclear transcription factors.
Collapse
Affiliation(s)
- Syed Husain Mustafa Rizvi
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA; Cardiology, Whitaker Cardiovascular Institute, And Boston University School of Medicine, Boston, MA, USA
| | - Di Shao
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
| | - Yuko Tsukahara
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA
| | - David Richard Pimentel
- Cardiology, Whitaker Cardiovascular Institute, And Boston University School of Medicine, Boston, MA, USA
| | - Robert M Weisbrod
- Cardiology, Whitaker Cardiovascular Institute, And Boston University School of Medicine, Boston, MA, USA
| | - Naomi M Hamburg
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA; Cardiology, Whitaker Cardiovascular Institute, And Boston University School of Medicine, Boston, MA, USA
| | - Mark E McComb
- Cardiovascular Proteomics Center, Boston University School of Medicine, Boston, MA, USA
| | - Reiko Matsui
- Vascular Biology Section, Boston University School of Medicine, Boston, MA, USA.
| | | |
Collapse
|
67
|
Pérez-Peiró M, Martín-Ontiyuelo C, Rodó-Pi A, Piccari L, Admetlló M, Durán X, Rodríguez-Chiaradía DA, Barreiro E. Iron Replacement and Redox Balance in Non-Anemic and Mildly Anemic Iron Deficiency COPD Patients: Insights from a Clinical Trial. Biomedicines 2021; 9:1191. [PMID: 34572377 PMCID: PMC8470868 DOI: 10.3390/biomedicines9091191] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/18/2022] Open
Abstract
In COPD patients, non-anemic iron deficiency (NAID) is a common systemic manifestation. We hypothesized that in COPD patients with NAID, iron therapy may improve systemic oxidative stress. The FACE (Ferinject assessment in patients with COPD and iron deficiency to improve exercise tolerance) study was a single-blind, unicentric, parallel-group, placebo-controlled clinical trial (trial registry: 2016-001238-89). Sixty-six patients were enrolled (randomization 2:1): iron arm, n = 44 and placebo arm, n = 22, with similar clinical characteristics. Serum levels of 3-nitrotyrosine, MDA-protein adducts, and reactive carbonyls, catalase, superoxide dismutase (SOD), glutathione, Trolox equivalent antioxidant capacity (TEAC), and iron metabolism biomarkers were quantified in both groups. In the iron-treated patients compared to placebo, MDA-protein adducts and 3-nitrotyrosine serum levels significantly declined, while those of GSH increased and iron metabolism parameters significantly improved. Hepcidin was associated with iron status parameters. This randomized clinical trial evidenced that iron replacement elicited a decline in serum oxidative stress markers along with an improvement in GSH levels in patients with stable severe COPD. Hepcidin may be a surrogate biomarker of iron status and metabolism in patients with chronic respiratory diseases. These findings have potential clinical implications in the management of patients with severe COPD.
Collapse
Affiliation(s)
- Maria Pérez-Peiró
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Clara Martín-Ontiyuelo
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
| | - Anna Rodó-Pi
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
| | - Lucilla Piccari
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
| | - Mireia Admetlló
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
| | - Xavier Durán
- Scientific and Technical Department, Hospital del Mar-IMIM, 08003 Barcelona, Spain;
| | - Diego A. Rodríguez-Chiaradía
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| | - Esther Barreiro
- Pulmonology Department-Muscle Wasting and Cachexia in Chronic Respiratory Diseases and Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Health and Experimental Sciences Department (CEXS), Universitat Pompeu Fabra (UPF), Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Spain; (M.P.-P.); (C.M.-O.); (A.R.-P.); (L.P.); (M.A.); (D.A.R.-C.)
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 08003 Barcelona, Spain
| |
Collapse
|
68
|
Song J, Yao L, Shi J, Li J, Xu C. Protective effects of N-acetylcysteine on a chemical-induced murine model of asthma. J Asthma 2021; 58:1208-1215. [PMID: 32546031 DOI: 10.1080/02770903.2020.1781166] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 05/24/2020] [Accepted: 06/07/2020] [Indexed: 12/26/2022]
Abstract
INTRODUCTION Oxidative stress is involved in the pathophysiology of inflammatory airway diseases, including asthma. In this study, we elucidated the possible protective effects of the antioxidant N-acetylcysteine (NAC) on a toluene diisocyanate (TDI)-induced murine asthma model. METHODS Male BALB/c mice were sensitized and challenged with TDI to generate a chemical-induced asthma model. NAC was given intraperitoneally to mice immediately after each TDI challenge. Airway reactivity to methacholine and bronchoalveolar lavage fluid was analyzed. Lungs were examined by histology. RESULTS NAC treatment dramatically reduced the increased airway hyperresponsiveness, inflammatory infiltration, and goblet cell metaplasia in TDI-exposed mice. Numbers of total cells, neutrophils, and eosinophils in the bronchoalveolar lavage fluid of TDI-challenged mice were significantly higher than vehicle control, but the administration of NAC decreased these inflammatory cell counts. TDI exposure led to significantly increased levels of interleukin 4 (IL-4) and IL-5, which were also suppressed by NAC. In addition, diminished lung reduced oxidized glutathione ratio and superoxide dismutase activity were observed after TDI challenge, and these changes were attenuated by NAC. CONCLUSION NAC treatment has beneficial effects in TDI-induced asthma.
Collapse
Affiliation(s)
- Jiafu Song
- Department of Respiratory and Critical Care Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Lianyungang, China
| | - Lihong Yao
- State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, China
| | - Jiaxin Shi
- Department of Respiratory and Critical Care Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Lianyungang, China
| | - Jiashu Li
- Department of Respiratory and Critical Care Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Lianyungang, China
| | - Caiyun Xu
- Department of Critical Care Medicine, Lianyungang First People's Hospital, Affiliated Hospital of Xuzhou Medical College, Lianyungang, China
| |
Collapse
|
69
|
Cottrill KA, Giacalone VD, Margaroli C, Bridges RJ, Koval M, Tirouvanziam R, McCarty NA. Mechanistic analysis and significance of sphingomyelinase-mediated decreases in transepithelial CFTR currents in nHBEs. Physiol Rep 2021; 9:e15023. [PMID: 34514718 PMCID: PMC8436056 DOI: 10.14814/phy2.15023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2021] [Accepted: 07/02/2021] [Indexed: 12/15/2022] Open
Abstract
Loss of function of the cystic fibrosis transmembrane conductance regulator (CFTR) causes cystic fibrosis (CF). In the lungs, this manifests as immune cell infiltration and bacterial infections, leading to tissue destruction. Previous work has determined that acute bacterial sphingomyelinase (SMase) decreases CFTR function in bronchial epithelial cells from individuals without CF (nHBEs) and with CF (cfHBEs, homozygous ΔF508-CFTR mutation). This study focuses on exploring the mechanisms underlying this effect. SMase increased the abundance of dihydroceramides, a result mimicked by blockade of ceramidase enzyme using ceranib-1, which also decreased CFTR function. The SMase-mediated inhibitory mechanism did not involve the reduction of cellular CFTR abundance or removal of CFTR from the apical surface, nor did it involve the activation of 5' adenosine monophosphate-activated protein kinase. In order to determine the pathological relevance of these sphingolipid imbalances, we evaluated the sphingolipid profiles of cfHBEs and cfHNEs (nasal) as compared to non-CF controls. Sphingomyelins, ceramides, and dihydroceramides were largely increased in CF cells. Correction of ΔF508-CFTR trafficking with VX445 + VX661 decreased some sphingomyelins and all ceramides, but exacerbated increases in dihydroceramides. Additional treatment with the CFTR potentiator VX770 did not affect these changes, suggesting rescue of misfolded CFTR was sufficient. We furthermore determined that cfHBEs express more acid-SMase protein than nHBEs. Lastly, we determined that airway-like neutrophils, which are increased in the CF lung, secrete acid-SMase. Identifying the mechanism of SMase-mediated inhibition of CFTR will be important, given the imbalance of sphingolipids in CF cells and the secretion of acid-SMase from cell types relevant to CF.
Collapse
Affiliation(s)
- Kirsten A. Cottrill
- Molecular and Systems Pharmacology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
| | - Vincent D. Giacalone
- Immunology and Molecular Pathogenesis PhD ProgramEmory UniversityAtlantaGeorgiaUSA
| | - Camilla Margaroli
- Department of MedicineDivision of PulmonaryAllergy & Critical Care MedicineUniversity of Alabama at BirminghamBirminghamAlabamaUSA
- Program in Protease/Matrix BiologyUniversity of Alabama at BirminghamBirminghamAlabamaUSA
| | - Robert J. Bridges
- Department of Physiology and BiophysicsCenter for Genetic DiseasesChicago Medical SchoolNorth ChicagoIllinoisUSA
| | - Michael Koval
- Department of MedicineDivision of Pulmonary, Allergy, Critical Care and Sleep Medicine and Department of Cell BiologyEmory UniversityAtlantaGeorgiaUSA
| | - Rabindra Tirouvanziam
- Department of Pediatrics and Children’s Healthcare of AtlantaCenter for Cystic Fibrosis and Airways Disease ResearchEmory University School of MedicineAtlantaGeorgiaUSA
| | - Nael A. McCarty
- Molecular and Systems Pharmacology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
- Department of Pediatrics and Children’s Healthcare of AtlantaCenter for Cystic Fibrosis and Airways Disease ResearchEmory University School of MedicineAtlantaGeorgiaUSA
| |
Collapse
|
70
|
Wang C, Jiang S, Zhang S, Ouyang Z, Wang G, Wang F. Research Progress of Metabolomics in Asthma. Metabolites 2021; 11:567. [PMID: 34564383 PMCID: PMC8466166 DOI: 10.3390/metabo11090567] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Revised: 08/10/2021] [Accepted: 08/19/2021] [Indexed: 12/25/2022] Open
Abstract
Asthma is a highly heterogeneous disease, but the pathogenesis of asthma is still unclear. It is well known that the airway inflammatory immune response is the pathological basis of asthma. Metabolomics is a systems biology method to analyze the difference of low molecular weight metabolites (<1.5 kDa) and explore the relationship between metabolic small molecules and pathophysiological changes of the organisms. The functional interdependence between immune response and metabolic regulation is one of the cores of the body's steady-state regulation, and its dysfunction will lead to a series of metabolic disorders. The signal transduction effect of specific metabolites may affect the occurrence of the airway inflammatory immune response, which may be closely related to the pathogenesis of asthma. Emerging metabolomic analysis may provide insights into the pathogenesis and diagnosis of asthma. The review aims to analyze the changes of metabolites in blood/serum/plasma, urine, lung tissue, and exhaled breath condensate (EBC) samples, and further reveals the potential pathogenesis of asthma according to the disordered metabolic pathways.
Collapse
Affiliation(s)
- Chao Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Shengyu Jiang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Siyu Zhang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Zhuoer Ouyang
- Department of Cellular Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China;
| | - Guoqiang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| | - Fang Wang
- Department of Pathogen Biology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (C.W.); (S.J.); (S.Z.)
| |
Collapse
|
71
|
Mucha P, Skoczyńska A, Małecka M, Hikisz P, Budzisz E. Overview of the Antioxidant and Anti-Inflammatory Activities of Selected Plant Compounds and Their Metal Ions Complexes. Molecules 2021; 26:4886. [PMID: 34443474 PMCID: PMC8398118 DOI: 10.3390/molecules26164886] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 08/09/2021] [Accepted: 08/10/2021] [Indexed: 12/12/2022] Open
Abstract
Numerous plant compounds and their metal-ion complexes exert antioxidative, anti-inflammatory, anticancer, and other beneficial effects. This review highlights the different bioactivities of flavonoids, chromones, and coumarins and their metal-ions complexes due to different structural characteristics. In addition to insight into the most studied antioxidative properties of these compounds, the first part of the review provides a comprehensive overview of exogenous and endogenous sources of reactive oxygen and nitrogen species, oxidative stress-mediated damages of lipids and proteins, and on protective roles of antioxidant defense systems, including plant-derived antioxidants. Additionally, the review covers the anti-inflammatory and antimicrobial activities of flavonoids, chromones, coumarins and their metal-ion complexes which support its application in medicine, pharmacy, and cosmetology.
Collapse
Affiliation(s)
- Paulina Mucha
- Department of the Chemistry of Cosmetic Raw Materials, Faculty of Pharmacy, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland
| | - Anna Skoczyńska
- Department of Pharmacology, School of Pharmacy with the Division of Laboratory Medicine in Sosnowiec, Medical University of Silesia, Poniatowskiego 15, 41-200 Sosnowiec, Poland;
| | - Magdalena Małecka
- Department of Physical Chemistry, Faculty of Chemistry, University of Lodz, Pomorska 163/165, 90-236 Łódź, Poland;
| | - Paweł Hikisz
- Department of Molecular Biophysics, Faculty of Biology and Environmental Protection, University of Lodz, Pomorska 141/143, 90-236 Łódź, Poland;
| | - Elzbieta Budzisz
- Department of the Chemistry of Cosmetic Raw Materials, Faculty of Pharmacy, Medical University of Łódź, Muszyńskiego 1, 90-151 Łódź, Poland
| |
Collapse
|
72
|
Cottrill KA, Peterson RJ, Lewallen CF, Koval M, Bridges RJ, McCarty NA. Sphingomyelinase decreases transepithelial anion secretion in airway epithelial cells in part by inhibiting CFTR-mediated apical conductance. Physiol Rep 2021; 9:e14928. [PMID: 34382377 PMCID: PMC8358481 DOI: 10.14814/phy2.14928] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2021] [Accepted: 05/21/2021] [Indexed: 12/11/2022] Open
Abstract
The cystic fibrosis transmembrane conductance regulator (CFTR) is an anion channel whose dysfunction causes cystic fibrosis (CF). The loss of CFTR function in pulmonary epithelial cells causes surface dehydration, mucus build-up, inflammation, and bacterial infections that lead to lung failure. Little has been done to evaluate the effects of lipid perturbation on CFTR activity, despite CFTR residing in the plasma membrane. This work focuses on the acute effects of sphingomyelinase (SMase), a bacterial virulence factor secreted by CF relevant airway bacteria which degrades sphingomyelin into ceramide and phosphocholine, on the electrical circuitry of pulmonary epithelial monolayers. We report that basolateral SMase decreases CFTR-mediated transepithelial anion secretion in both primary bronchial and tracheal epithelial cells from explant tissue, with current CFTR modulators unable to rescue this effect. Focusing on primary cells, we took a holistic ion homeostasis approach to determine a cause for reduced anion secretion following SMase treatment. Using impedance analysis, we determined that basolateral SMase inhibits apical and basolateral conductance in non-CF primary cells without affecting paracellular permeability. In CF primary airway cells, correction with clinically relevant CFTR modulators did not prevent SMase-mediated inhibition of CFTR currents. Furthermore, SMase was found to inhibit only apical conductance in these cells. Future work should determine the mechanism for SMase-mediated inhibition of CFTR currents, and further explore the clinical relevance of SMase and sphingolipid imbalances.
Collapse
Affiliation(s)
- Kirsten A. Cottrill
- Molecular and Systems Pharmacology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
| | - Raven J. Peterson
- Biochemistry, Cell, and Developmental Biology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
| | - Colby F. Lewallen
- Georgia Institute of TechnologyG.W. Woodruff School of Mechanical EngineeringAtlantaGeorgiaUSA
| | - Michael Koval
- Division of Pulmonary, Allergy, Critical Care and Sleep MedicineDepartment of MedicineEmory UniversityAtlantaGeorgiaUSA
- Department of Cell BiologyEmory UniversityAtlantaGeorgiaUSA
| | - Robert J. Bridges
- Department of Physiology and BiophysicsCenter for Genetic DiseasesChicago Medical SchoolNorth Chicago, IllinoisUSA
| | - Nael A. McCarty
- Molecular and Systems Pharmacology PhD ProgramEmory UniversityAtlantaGeorgiaUSA
- Department of Pediatrics and Children’s Healthcare of AtlantaCenter for Cystic Fibrosis and Airways Disease ResearchEmory University School of MedicineAtlantaGeorgiaUSA
| |
Collapse
|
73
|
Assayag M, Goldstein S, Samuni A, Berkman N. 3-Carbamoyl-proxyl nitroxide radicals attenuate bleomycin-induced pulmonary fibrosis in mice. Free Radic Biol Med 2021; 171:135-142. [PMID: 33989760 DOI: 10.1016/j.freeradbiomed.2021.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 05/05/2021] [Accepted: 05/06/2021] [Indexed: 12/31/2022]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a fatal interstitial lung disease with a poor prognosis and limited treatment options. Oxidative and nitrosative stress is implicated as one of the main pathogenic pathways in IPF. The rationale for the use of antioxidants to treat lung fibrosis is appealing, however to date a consistent beneficial effect for such an approach has not been observed. We have recently demonstrated that nitroxides, particularly 3-carbamoyl-proxyl (3-CP), markedly reduce airway inflammation, airway hyper-responsiveness, and protein nitration of the lung tissue in a mouse model of ovalbumin-induced acute asthma, thus prompting its use for the treatment of IPF. The present study investigates the effect of 3-CP on the development of lung fibrosis using the murine intratracheal bleomycin model. 3-CP was administered either intranasally or orally during the entire experiment or starting 7 days after induction of the lung injury. 3-CP was found to be both a preventive and a therapeutic drug reducing the lung fibrosis (histological score), the increase in collagen content, protein nitration, TGF-β levels, the degree of weight loss as well as inhibiting the impairment of lung function. Nitroxides are catalytic antioxidants that preferentially detoxify radicals, and therefore the effect of 3-CP on the severity of the disease supports the involvement of reactive oxygen and nitrogen species in the disease pathology.
Collapse
Affiliation(s)
- Miri Assayag
- Institute of Pulmonary Medicine, Hadassah Medical Center and Faculty of Medicine, Israel
| | - Sara Goldstein
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, 91904, Israel.
| | - Amram Samuni
- Institute of Medical Research, Israel-Canada Medical School, The Hebrew University of Jerusalem, Jerusalem, 91120, Israel
| | - Neville Berkman
- Institute of Pulmonary Medicine, Hadassah Medical Center and Faculty of Medicine, Israel
| |
Collapse
|
74
|
Das SK, Ali M, Shetake NG, Dumpala RMR, Pandey BN, Kumar A. Mechanism of thorium-nitrate and thorium-dioxide induced cytotoxicity in normal human lung epithelial cells (WI26): Role of oxidative stress, HSPs and DNA damage. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 281:116969. [PMID: 33845224 DOI: 10.1016/j.envpol.2021.116969] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 03/01/2021] [Accepted: 03/16/2021] [Indexed: 06/12/2023]
Abstract
Inhalation represents the most prevalent route of exposure with Thorium-232 compounds (Th-nitrate/Th-dioxide)/Th-containing dust in real occupational scenario. The present study investigated the mechanism of Th response in normal human alveolar epithelial cells (WI26), exposed to Th-nitrate or colloidal Th-dioxide (1-100 μg/ml, 24-72 h). Assessment in terms of changes in cell morphology, cell proliferation (cell count), plasma membrane integrity (lactate dehydrogenase leakage) and mitochondrial metabolic activity (MTT reduction) showed that Th-dioxide was quantitatively more deleterious than Th-nitrate to WI26 cells. TEM and immunofluorescence analysis suggested that Th-dioxide followed a clathrin/caveolin-mediated endocytosis, however, membrane perforation/non-endocytosis seemed to be the mode of Th internalization in cells exposed to Th-nitrate. Th-estimation by ICP-MS showed significantly higher uptake of Th in cells treated with Th-dioxide than with Th-nitrate at a given concentration. Both Th-dioxide and nitrate were found to increase the level of reactive oxygen species, which seemed to be responsible for lipid peroxidation, alteration in mitochondrial membrane potential and DNA-damage. Amongst HSPs, the protein levels of HSP70 and HSP90 were affected differentially by Th-nitrate/dioxide. Specific inhibitors of ATM (KU55933) or HSP90 (17AAG) were found to increase the Th- cytotoxicity suggesting prosurvival role of these signaling molecules in rescuing the cells from Th-toxicity.
Collapse
Affiliation(s)
- Sourav Kumar Das
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - Manjoor Ali
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - Neena G Shetake
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - Rama Mohan R Dumpala
- Radiochemistry Division, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India
| | - Badri N Pandey
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400 094, India
| | - Amit Kumar
- Radiation Biology & Health Sciences Division, Bhabha Atomic Research Centre, Trombay, Mumbai, 400 085, India; Homi Bhabha National Institute, Anushakti Nagar, Mumbai, 400 094, India.
| |
Collapse
|
75
|
Tang G, Xu Y, Zhang C, Wang N, Li H, Feng Y. Green Tea and Epigallocatechin Gallate (EGCG) for the Management of Nonalcoholic Fatty Liver Diseases (NAFLD): Insights into the Role of Oxidative Stress and Antioxidant Mechanism. Antioxidants (Basel) 2021; 10:1076. [PMID: 34356308 PMCID: PMC8301033 DOI: 10.3390/antiox10071076] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 06/27/2021] [Accepted: 07/01/2021] [Indexed: 02/07/2023] Open
Abstract
Nonalcoholic fatty liver diseases (NAFLD) represent a set of liver disorders progressing from steatosis to steatohepatitis, fibrosis, cirrhosis, and hepatocellular carcinoma, which induce huge burden to human health. Many pathophysiological factors are considered to influence NAFLD in a parallel pattern, involving insulin resistance, oxidative stress, lipotoxicity, mitochondrial dysfunction, endoplasmic reticulum stress, inflammatory cascades, fibrogenic reaction, etc. However, the underlying mechanisms, including those that induce NAFLD development, have not been fully understood. Specifically, oxidative stress, mainly mediated by excessive accumulation of reactive oxygen species, has participated in the multiple NAFLD-related signaling by serving as an accelerator. Ameliorating oxidative stress and maintaining redox homeostasis may be a promising approach for the management of NAFLD. Green tea is one of the most important dietary resources of natural antioxidants, above which epigallocatechin gallate (EGCG) notably contributes to its antioxidative action. Accumulative evidence from randomized clinical trials, systematic reviews, and meta-analysis has revealed the beneficial functions of green tea and EGCG in preventing and managing NAFLD, with acceptable safety in the patients. Abundant animal and cellular studies have demonstrated that green tea and EGCG may protect against NAFLD initiation and development by alleviating oxidative stress and the related metabolism dysfunction, inflammation, fibrosis, and tumorigenesis. The targeted signaling pathways may include, but are not limited to, NRF2, AMPK, SIRT1, NF-κB, TLR4/MYD88, TGF-β/SMAD, and PI3K/Akt/FoxO1, etc. In this review, we thoroughly discuss the oxidative stress-related mechanisms involved in NAFLD development, as well as summarize the protective effects and underlying mechanisms of green tea and EGCG against NAFLD.
Collapse
Affiliation(s)
- Guoyi Tang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; (G.T.); (Y.X.); (C.Z.); (N.W.)
| | - Yu Xu
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; (G.T.); (Y.X.); (C.Z.); (N.W.)
| | - Cheng Zhang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; (G.T.); (Y.X.); (C.Z.); (N.W.)
| | - Ning Wang
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; (G.T.); (Y.X.); (C.Z.); (N.W.)
| | - Huabin Li
- School of Public Health, Sun Yat-sen University, Guangzhou 510080, China;
| | - Yibin Feng
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, 10 Sassoon Road, Pokfulam, Hong Kong, China; (G.T.); (Y.X.); (C.Z.); (N.W.)
| |
Collapse
|
76
|
Camarinho R, Garcia PV, Choi H, Rodrigues AS. Pulmonary oxidative stress and apoptosis in mice chronically exposed to hydrothermal volcanic emissions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:35709-35716. [PMID: 33675493 DOI: 10.1007/s11356-021-13043-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 02/15/2021] [Indexed: 06/12/2023]
Abstract
Recent studies have shown that exposure to hydrothermal emissions has a negative impact on the respiratory system. Still, volcanogenic air pollution studies are still outnumbered when compared to anthropogenic studies which can result in an unknown risk to the human populations living near volcanically active areas. This study was carried out in São Miguel Island, with noneruptive volcanically active environments, such as the Furnas volcano caldera. Its noneruptive volcanism presents itself as hydrothermal emissions, mainly by the release of nearly 1000 T d-1 of CO2 along with H2S, and the radioactive gas radon; metals [e.g., mercury (Hg), cadmium (Cd), copper (Cu), and zinc (Zn)] and particulate matter are also released in a daily basis. We test the hypothesis whether chronic exposure to hydrothermal emissions causes pulmonary oxidative stress, using Mus musculus as a surrogate species. Mus musculus was live-captured in two villages with hydrothermal emissions and one village without any type of volcanic activity. The level of pulmonary oxidative stress was immunohistochemically assessed by using an OxyIHCTM Oxidative stress detection kit, and the detection of terminal deoxynucleotidyl transferase (TdT)-mediated dUTP nick end-labeling (TUNEL) was used to evaluate apoptosis in lung tissues. Mice chronically exposed to hydrothermal emissions presented increased levels of oxidative stress and amount of apoptotic cells. We demonstrate, for the first time, the high oxidative stress potential in the lungs of mice chronically exposed to hydrothermal emissions. This study highlights the usefulness of M. musculus as a bioindicator species and enforces the necessity of regularly biomonitor the inhabitants of hydrothermal areas to prevent respiratory pathologies.
Collapse
Affiliation(s)
- Ricardo Camarinho
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, Rua da Mãe de Deus, Apartado 1422, 9501-801, Ponta Delgada, Açores, Portugal.
- IVAR - Instituto de Vulcanologia e Avaliação de Riscos, University of the Azores, 9501-801, Ponta Delgada, Portugal.
| | - Patrícia Ventura Garcia
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, Rua da Mãe de Deus, Apartado 1422, 9501-801, Ponta Delgada, Açores, Portugal
- CE3C - cE3c, Centre for Ecology, Evolution and Environmental Changes/Azorean Biodiversity Group, University of the Azores, 9501-801, Ponta Delgada, Azores, Portugal
| | - Hyunok Choi
- College of Health, Lehigh University, STEPS Building, Room 264, 1 West Packer Avenue, Bethlehem, PA, 18015-3001, USA
| | - Armindo Santos Rodrigues
- Faculdade de Ciências e Tecnologia, Universidade dos Açores, Rua da Mãe de Deus, Apartado 1422, 9501-801, Ponta Delgada, Açores, Portugal
- IVAR - Instituto de Vulcanologia e Avaliação de Riscos, University of the Azores, 9501-801, Ponta Delgada, Portugal
| |
Collapse
|
77
|
Zhao X, Abulikemu A, Lv S, Qi Y, Duan J, Zhang J, Chen R, Guo C, Li Y, Sun Z. Oxidative stress- and mitochondrial dysfunction-mediated cytotoxicity by silica nanoparticle in lung epithelial cells from metabolomic perspective. CHEMOSPHERE 2021; 275:129969. [PMID: 33662726 DOI: 10.1016/j.chemosphere.2021.129969] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 02/07/2021] [Accepted: 02/08/2021] [Indexed: 06/12/2023]
Abstract
Quantities of researches have demonstrated silica nanoparticles (SiNPs) exposure inevitably induced damage to respiratory system, nonetheless, knowledge of its toxicological behavior and metabolic interactions with the cellular machinery that determines the potentially deleterious outcomes are limited and poorly elucidated. Here, the metabolic responses of lung bronchial epithelial cells (BEAS-2B) under SiNPs exposure were investigated using ultra performance liquid chromatography-mass spectrum (UPLC-MS)-based metabolomics research. Results revealed that even with low cytotoxicity, SiNPs disturbed global metabolism. Five metabolic pathways were significantly perturbed, in particular, oxidative stress- and mitochondrial dysfunction-related GSH metabolism and pantothenate and coenzyme A (CoA) biosynthesis, where the identified metabolites glutathione (GSH), glycine, beta-alanine, cysteine, cysteinyl-glycine and pantothenic acid were included. In support of the metabolomics profiling, SiNPs caused abnormality in mitochondrial structure and mitochondrial dysfunction, as evidenced by the inhibition of cellular respiration and ATP production. Moreover, SiNPs triggered oxidative stress as confirmed by the dose-dependent ROS generation, down-regulated nuclear factor erythroid 2-related factor 2 (NRF2) signaling, together with GSH depletion in SiNPs-treated BEAS-2B cells. Oxidative DNA damage and cell membrane dis-integrity were also detected in response to SiNPs exposure, which was correspondingly in agreed with the elevated 8-hydroxyguanosine (8-OHdG) and decreased phospholipids screened through metabolic analysis. Thereby, we successfully used the metabolomics approaches to manifest SiNPs-elicited toxicity through oxidative stress, mitochondrial dysfunction, DNA damage and rupture of membrane integrity in BEAS-2B cells. Overall, our study provided novel insights into the mechanism underlying SiNPs-induced pulmonary toxicity.
Collapse
Affiliation(s)
- Xinying Zhao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Alimire Abulikemu
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Songqing Lv
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Yi Qi
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Junchao Duan
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China
| | - Jie Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, 361102, Xiamen, China
| | - Rui Chen
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| | - Caixia Guo
- Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China; Department of Occupational Health and Environmental Health, School of Public Health, Capital Medical University, Beijing, 100069, China.
| | - Yanbo Li
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China.
| | - Zhiwei Sun
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing, 100069, China; Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing, 100069, China
| |
Collapse
|
78
|
Keum H, Kim D, Kim J, Kim TW, Whang CH, Jung W, Jon S. A bilirubin-derived nanomedicine attenuates the pathological cascade of pulmonary fibrosis. Biomaterials 2021; 275:120986. [PMID: 34175563 PMCID: PMC8218594 DOI: 10.1016/j.biomaterials.2021.120986] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 06/10/2021] [Accepted: 06/20/2021] [Indexed: 12/20/2022]
Abstract
Pulmonary fibrosis is an irreparable and life-threatening disease with only limited therapeutic options. The recent outbreak of COVID-19 has caused a sharp rise in the incidence of pulmonary fibrosis owing to SARS-CoV-2 infection-mediated acute respiratory distress syndrome (ARDS). The considerable oxidative damage caused by locally infiltrated immune cells plays a crucial role in ARDS, suggesting the potential use of antioxidative therapeutics. Here, we report the therapeutic potential of nanoparticles derived from the endogenous antioxidant and anti-inflammatory bile acid, bilirubin (BRNPs), in treating pulmonary fibrosis in a bleomycin-induced mouse model of the disease. Our results demonstrate that BRNPs can effectively reduce clinical signs in mice, as shown by histological, disease index evaluations, and detection of biomarkers. Our findings suggest that BRNPs, with their potent antioxidant and anti-inflammatory effects, long blood circulation half-life, and preferential accumulation at the inflamed site, are potentially a viable clinical option for preventing Covid-19 infection-associated pulmonary fibrosis.
Collapse
Affiliation(s)
- Hyeongseop Keum
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Dohyeon Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Jinjoo Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Tae Woo Kim
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Chang-Hee Whang
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Wonsik Jung
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| | - Sangyong Jon
- Department of Biological Sciences, KAIST Institute for the BioCentury, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea; Center for Precision Bio-Nanomedicine, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Daejeon, 34141, Republic of Korea.
| |
Collapse
|
79
|
Jiao B, Guo S, Yang X, Sun L, Sai L, Yu G, Bo C, Zhang Y, Peng C, Jia Q, Dai Y. The role of HMGB1 on TDI-induced NLPR3 inflammasome activation via ROS/NF-κB pathway in HBE cells. Int Immunopharmacol 2021; 98:107859. [PMID: 34153664 DOI: 10.1016/j.intimp.2021.107859] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 06/04/2021] [Accepted: 06/04/2021] [Indexed: 01/03/2023]
Abstract
To explore the potential role of HMGB1 on TDI-induced NLRP3 inflammasome activation, HBE cells were treated with TDI-HSA conjugate to observe the changes of HMGB1, TLR4, NF-κB, Nrf2 and NLRP3 inflammasome related proteins expressions, ROS release and MMP. NAC, TPCA-1 and Resatorvid pre-treatments were applied to explore the effects of ROS, NF-κB and TLR4 on TDI-induced NLRP3 inflammasome activation. The CRISPR/Cas9 system was used to construct HMGB1 gene knockout HBE cell line and then to explore the role of HMGB1 on TDI-HSA induced NLRP3 inflammasome activation. GL pre-treatment was applied to further confirm the role of HMGB1. Results showed that TDI increased HMGB1, TLR4, P-p65, Nrf2 proteins expressions and ROS release, decreased MMP level and activated NLRP3 inflammasome in HBE cells in a dose dependent manner. NAC, TPCA-1 and Resatorvid pre-treatments decreased the expression of P-p65 and inhibited NLRP3 inflammasome activation. Inhibition of HMGB1 decreased Nrf2 expression and ROS release, improved MMP level and reduced NLRP3 inflammasome activation. GL ameliorated NLRP3 inflammasome activation via inhibiting HMGB1 regulated ROS/NF-κB pathway. These results indicated that HMGB1 was involved in TDI-induced NLRP3 inflammasome activation as a positive regulatory mechanism. The study provided a potential target for early prevention and treatment of TDI-OA.
Collapse
Affiliation(s)
- Bo Jiao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Sumei Guo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Xiaohan Yang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Lei Sun
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Linlin Sai
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Cunxiang Bo
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Yu Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China
| | - Cheng Peng
- Queensland Alliance for Environmental Health Sciences (QAEHS), University of Queensland, Brisbane, Queensland 4029, Australia
| | - Qiang Jia
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250062, China.
| | - Yufei Dai
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention (CDC), Beijing 100050, China.
| |
Collapse
|
80
|
Peng L, Wen L, Shi Q, Gao F, Huang B, Wang C. Chelerythrine Ameliorates Pulmonary Fibrosis via Activating the Nrf2/ARE Signaling Pathway. Cell Biochem Biophys 2021; 79:337-347. [PMID: 33580396 DOI: 10.1007/s12013-021-00967-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2021] [Indexed: 12/12/2022]
Abstract
Chelerythrine (CHE) is a natural benzophenanthridine alkaloid, which has shown its anti-fibrosis activity in kidney and liver, while the impact of CHE in pulmonary fibrosis is still unclear. This study is developed to explore the impact and mechanism of CHE in pulmonary fibrosis. Pulmonary fibrosis mouse models were established through intratracheal injection of bleomycin (BLM), after which the mice were intraperitoneally injected with CHE (0.375 or 0.75 mg/kg/d) every other day. The mice were sacrificed at the 28th day to collect blood serum, bronchoalveolar lavage fluid (BALF), and pulmonary tissues. Then, the severity of pulmonary fibrosis and the expression of nuclear factor erythroid 2 [NF-E2]-related factor 2 (Nrf2) in the pulmonary tissues were detected. Western blot analysis quantified the expressions of fibronectin and alpha-smooth muscle actin (α-SMA). The levels of 4-hydroxynonenal (4-HNE), glutathione (GSH), superoxide dismutase (SOD), TGF-β and hydroxyproline (HP) in the BALF, and pulmonary tissues were measured. The expression levels of Nrf2 and its downstream genes, hemeoxygenase-1 (HO-1) and NAD (P) H: quinone oxidoreductase (NQO1) were examined. CHE at the concentration of 0.375 or 0.75 mg/kg/d could attenuate pulmonary fibrosis. CHE injection reduced the expression levels of fibronectin, α-SMA, and TGF-β, upregulated the levels of SOD and GSH and decreased the levels of 4-HNE and HP. Also, CHE increased the expressions of Nrf2, HO-1, and NQO1. Treatment of Nrf2/antioxidant response element (ARE) inhibitor could block the Nrf2/ARE signaling pathway, thus perturbing the inhibition of CHE on BLM-stimulated pulmonary fibrosis in mice. CHE alleviates BLM-induced pulmonary fibrosis in mice through activating the Nrf2/ARE pathway to increase the activity of antioxidant enzymes.
Collapse
Affiliation(s)
- Ling Peng
- Department of Nephrology, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan, China
| | - Li Wen
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541000, Guangxi, China
| | - Qingfeng Shi
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541000, Guangxi, China
| | - Feng Gao
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541000, Guangxi, China
| | - Bin Huang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541000, Guangxi, China
| | - Changming Wang
- Department of Respiratory Medicine, The Fifth Affiliated Hospital of Guilin Medical University, Guilin People's Hospital, Guilin, 541000, Guangxi, China.
| |
Collapse
|
81
|
Dwivedi S, Kushalan S, Paithankar JG, D'Souza LC, Hegde S, Sharma A. Environmental toxicants, oxidative stress and health adversities: interventions of phytochemicals. J Pharm Pharmacol 2021; 74:516-536. [PMID: 33822130 DOI: 10.1093/jpp/rgab044] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/17/2021] [Indexed: 12/17/2022]
Abstract
OBJECTIVES Oxidative stress is the most common factor mediating environmental chemical-induced health adversities. Recently, an exponential rise in the use of phytochemicals as an alternative therapeutics against oxidative stress-mediated diseases has been documented. Due to their free radical quenching property, plant-derived natural products have gained substantial attention as a therapeutic agent in environmental toxicology. The present review aimed to describe the therapeutic role of phytochemicals in mitigating environmental toxicant-mediated sub-cellular and organ toxicities via controlling cellular antioxidant response. METHODS The present review has covered the recently related studies, mainly focussing on the free radical scavenging role of phytochemicals in environmental toxicology. KEY FINDINGS In vitro and in vivo studies have reported that supplementation of antioxidant-rich compounds can ameliorate the toxicant-induced oxidative stress, thereby improving the health conditions. Improving the cellular antioxidant pool has been considered as a mode of action of phytochemicals. However, the other cellular targets of phytochemicals remain uncertain. CONCLUSIONS Knowing the therapeutic value of phytochemicals to mitigate the chemical-induced toxicity is an initial stage; mechanistic understanding needs to decipher for development as therapeutics. Moreover, examining the efficacy of phytochemicals against mixer toxicity and identifying the bioactive molecule are major challenges in the field.
Collapse
Affiliation(s)
- Shiwangi Dwivedi
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Sharanya Kushalan
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Deralakatte, Mangaluru, India
| | - Jagdish Gopal Paithankar
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Leonard Clinton D'Souza
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| | - Smitha Hegde
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Bioresource and Biotechnology, Deralakatte, Mangaluru, India
| | - Anurag Sharma
- Nitte (Deemed to be University), Nitte University Centre for Science Education and Research (NUCSER), Division of Environmental Health and Toxicology, Deralakatte, Mangaluru, India
| |
Collapse
|
82
|
Qi C, Wang H, Liu Z, Yang H. Oxidative Stress and Trace Elements in Pulmonary Tuberculosis Patients During 6 Months Anti-tuberculosis Treatment. Biol Trace Elem Res 2021; 199:1259-1267. [PMID: 32583224 DOI: 10.1007/s12011-020-02254-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Accepted: 06/15/2020] [Indexed: 12/27/2022]
Abstract
Pulmonary tuberculosis (TB) is a well-known cause of imbalance in oxidative stress (OS) status and trace element levels. However, little information is available for targeting the correlation between OS and trace elements in pulmonary TB patients. The aim of our study was to analyze the OS status and its correlation with trace elements in patients initially and during 6 months anti-TB treatment. Eighty-six newly diagnosed pulmonary TB patients were consecutively recruited, and 112 age- and sex-matched healthy controls participated in the study. Serum markers of OS and trace elements levels were tested and analyzed in all subjects during 6 months anti-TB treatment. Compared with healthy controls, significantly increased level of malondialdehyde (MDA), decreased glutathione (GSH) level, superoxide dismutase (SOD), and catalase (CAT) activities were found in TB patients. The activities of SOD and CAT and GSH level recovered till normal range at treatment final. Zinc (Zn), selenium (Se), and copper (Cu) concentrations were significantly lower in TB patients in comparison with healthy controls, whereas Zn, Cu, and Se concentrations rise during 6 months anti-TB treatment. Zn was positively correlated with Cu, Se, and GSH, while MDA was negatively correlated with Zn, Se, SOD, and CAT, and SOD was positively correlated with Cu, Zn, and CAT. Our findings indicate that anti-TB treatment could reduce the status of OS and increase the levels of trace elements. The routine assessment of OS markers and element traces may guarantee improved monitoring the anti-TB treatment.
Collapse
Affiliation(s)
- Chaoqun Qi
- Department of Clinical Laboratory, Linyi People's Hospital, Fenghuang Street 233, Hedong District, Linyi, 276000, China
| | - Hongjun Wang
- Department of Occupational Disease, Linyi People's Hospital, Fenghuang Street 233, Hedong District, Linyi, 276000, China
| | - Zhaoying Liu
- Department of Occupational Disease, Linyi People's Hospital, Fenghuang Street 233, Hedong District, Linyi, 276000, China
| | - Haibo Yang
- Department of Occupational Disease, Linyi People's Hospital, Fenghuang Street 233, Hedong District, Linyi, 276000, China.
| |
Collapse
|
83
|
NOX4-Derived ROS Promotes Collagen I Deposition in Bronchial Smooth Muscle Cells by Activating Noncanonical p38MAPK/Akt-Mediated TGF- β Signaling. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6668971. [PMID: 33824697 PMCID: PMC8007363 DOI: 10.1155/2021/6668971] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/10/2021] [Accepted: 02/18/2021] [Indexed: 01/17/2023]
Abstract
Background Airway smooth muscle (ASM) remodeling is a hallmark in chronic obstructive pulmonary disease (COPD). NADPH oxidase 4- (NOX4-) mediated reactive oxygen species (ROS) production plays a crucial role in cell differentiation and extracellular matrix (ECM) synthesis in ASM remodeling. However, the precise mechanisms underpinning its pathogenic roles remain elusive. Methods The expression of NOX4 and TGF-β1 in the airway of the lung was measured in COPD patients and the control group. Cigarette smoke- (CS-) induced emphysema mice were generated, and the alteration of α-SMA, NOX4, TGF-β1, and collagen I was accessed. The changes of the expression of ECM markers, NOX4, components of TGF-β/Smad, and MAPK/Akt signaling in human bronchial smooth muscle cells (HBSMCs) were ascertained for delineating mechanisms of NOX4-mediated ROS production on cell differentiation and remodeling in human ASM cells. Results An increased abundance of NOX4 and TGF-β1 proteins in the epithelial cells and ASM of lung was observed in COPD patients compared with the control group. Additionally, an increased abundance expression of NOX4 and α-SMA was observed in the lungs of the CS-induced emphysema mouse model. TGF-β1 displayed abilities to increase the oxidative burden and collagen I production, along with enhanced phosphorylation of ERK, p38MAPK, and p-Akt473 in HBSMCs. These effects of TGF-β1 could be inhibited by the ROS scavenger N-acetylcysteine (NAC), siRNA-mediated knockdown of Smad3 and NOX4, and pharmacological inhibitors SB203580 (p38MAPK inhibitor) and LY294002 (Akt inhibitor). Conclusions NOX4-mediated ROS production alters TGF-β1-induced cell differentiation and collagen I protein synthesis in HBSMCs in part through the p38MAPK/Akt signaling pathway in a Smad-dependent manner.
Collapse
|
84
|
Sharma A, Tewari D, Nabavi SF, Nabavi SM, Habtemariam S. Reactive oxygen species modulators in pulmonary medicine. Curr Opin Pharmacol 2021; 57:157-164. [PMID: 33743400 DOI: 10.1016/j.coph.2021.02.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 11/23/2020] [Accepted: 02/08/2021] [Indexed: 12/23/2022]
Abstract
Adapted to effectively capture oxygen from inhaled air and deliver it to all other parts of the body, the lungs constitute the organ with the largest surface area. This makes the lungs more susceptible to airborne pathogens and pollutants that mediate pathologies through generation of reactive oxygen species (ROS). One pathological consequence of excessive levels of ROS production is pulmonary diseases that account for a large number of mortality and morbidity in the world. Of the various mechanisms involved in pulmonary disease pathogenesis, mitochondrial dysfunction takes prominent importance. Herein, we briefly describe the significance of oxidative stress caused by ROS in pulmonary diseases and some possible therapeutic strategies.
Collapse
Affiliation(s)
- Ankush Sharma
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Devesh Tewari
- Department of Pharmacognosy, School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, 144411, India
| | - Seyed Fazel Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, 1435916471, Iran
| | - Seyed Mohammad Nabavi
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, 1435916471, Iran
| | - Solomon Habtemariam
- Pharmacognosy Research Laboratories and Herbal Analysis Services, School of Science, University of Greenwich, Central Avenue, Chatham-Maritime, Kent, ME4 4TB, United Kingdom.
| |
Collapse
|
85
|
Rinaldi L, Pafundi PC, Galiero R, Caturano A, Morone MV, Silvestri C, Giordano M, Salvatore T, Sasso FC. Mechanisms of Non-Alcoholic Fatty Liver Disease in the Metabolic Syndrome. A Narrative Review. Antioxidants (Basel) 2021; 10:270. [PMID: 33578702 PMCID: PMC7916383 DOI: 10.3390/antiox10020270] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/04/2021] [Accepted: 02/08/2021] [Indexed: 02/07/2023] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) and metabolic syndrome (MS) are two different entities sharing common clinical and physio-pathological features, with insulin resistance (IR) as the most relevant. Large evidence leads to consider it as a risk factor for cardiovascular disease, regardless of age, sex, smoking habit, cholesterolemia, and other elements of MS. Therapeutic strategies remain still unclear, but lifestyle modifications (diet, physical exercise, and weight loss) determine an improvement in IR, MS, and both clinical and histologic liver picture. NAFLD and IR are bidirectionally correlated and, consequently, the development of pre-diabetes and diabetes is the most direct consequence at the extrahepatic level. In turn, type 2 diabetes is a well-known risk factor for multiorgan damage, including an involvement of cardiovascular system, kidney and peripheral nervous system. The increased MS incidence worldwide, above all due to changes in diet and lifestyle, is associated with an equally significant increase in NAFLD, with a subsequent rise in both morbidity and mortality due to both metabolic, hepatic and cardiovascular diseases. Therefore, the slowdown in the increase of the "bad company" constituted by MS and NAFLD, with all the consequent direct and indirect costs, represents one of the main challenges for the National Health Systems.
Collapse
Affiliation(s)
- Luca Rinaldi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| | - Pia Clara Pafundi
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| | - Raffaele Galiero
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| | - Alfredo Caturano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| | - Maria Vittoria Morone
- Department of Experimental Medicine, Section of Microbiology, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy;
| | - Chiara Silvestri
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| | - Mauro Giordano
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| | - Teresa Salvatore
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Via De Crecchio 7, 80138 Naples, Italy;
| | - Ferdinando Carlo Sasso
- Department of Advanced Medical and Surgical Sciences, University of Campania Luigi Vanvitelli, Piazza Luigi Miraglia 2, 80138 Naples, Italy; (L.R.); (P.C.P.); (R.G.); (A.C.); (C.S.); (M.G.)
| |
Collapse
|
86
|
Asthma and air pollution: recent insights in pathogenesis and clinical implications. Curr Opin Pulm Med 2021; 26:10-19. [PMID: 31724961 DOI: 10.1097/mcp.0000000000000644] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
PURPOSE OF REVIEW Air pollution has adverse effects on the onset and morbidity of respiratory diseases, including asthma. In this review, we discuss recent insights into the effects of air pollution on the incidence and exacerbation of asthma. We focus on epidemiological studies that describe the association between air pollution exposure and development, mortality, persistence and exacerbations of asthma among different age groups. Moreover, we also provide an update on translational studies describing the mechanisms behind this association. RECENT FINDINGS Mechanisms linking air pollutants such as particulate matter, nitrogen dioxide (NO2) and ozone to the development and exacerbation of asthma include the induction of both eosinophilic and neutrophilic inflammation driven by stimulation of airway epithelium and increase of pro-inflammatory cytokine production, oxidative stress and DNA methylation changes. Although exposure during foetal development is often reported as a crucial timeframe, exposure to air pollution is detrimental in people of all ages, thus influencing asthma onset as well as increase in asthma prevalence, mortality, persistence and exacerbation. SUMMARY In conclusion, this review highlights the importance of reducing air pollution levels to avert the progressive increase in asthma incidence and morbidity.
Collapse
|
87
|
Li S, Shao L, Fang J, Zhang J, Chen Y, Yeo AJ, Lavin MF, Yu G, Shao H. Hesperetin attenuates silica-induced lung injury by reducing oxidative damage and inflammatory response. Exp Ther Med 2021; 21:297. [PMID: 33717240 PMCID: PMC7885076 DOI: 10.3892/etm.2021.9728] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Accepted: 12/11/2020] [Indexed: 02/07/2023] Open
Abstract
Oxidative stress and the inflammatory response are two important mechanisms of silica-induced lung injury. Hesperetin (HSP) is a natural flavonoid compound that is found in citrus fruits and has been indicated to exhibit strong antioxidant and anti-inflammatory properties. The current study evaluated the protective effect of HSP on lung injury in rats exposed to silica. The results indicated that the degree of alveolitis and pulmonary fibrosis in the HSP-treated group was significantly decreased compared with the silica model group. The content of hydroxyproline (HYP) was also revealed to decrease overall in the HSP treated group compared with the silica model group, indicating that the degree of pulmonary fibrosis was decreased compared with the silica model group. The present study also demonstrated that HSP reduced oxidation levels of malondialdehyde (MDA) and increased the activities of antioxidant enzymes superoxide dismutase (SOD), catalase (CAT) and glutathione peroxidase (GSH-PX). Total antioxidant capacity (T-AOC) was also increased following HSP treatment, indicating that HSP can alleviate oxidative stress in the lung tissue of silica-exposed rats. In addition, HSP was revealed to inhibit the synthesis and secretion of fibrogenic factor TGF-β1, reduce the production of pro-inflammatory cytokines IL-1β, IL-4, TNF-α and increase the levels of anti-inflammatory factors IFN-γ and IL-10. The current study demonstrated that HSP can effectively attenuate silica-induced lung injury by reducing oxidative damage and the inflammatory response.
Collapse
Affiliation(s)
- Shuxian Li
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Linlin Shao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Jinguo Fang
- Primary Health Department, Linqing Health Bureau, Linqing, Shandong 252600, P.R. China
| | - Juan Zhang
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Yanqin Chen
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Abrey J Yeo
- Centre for Clinical Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Martin F Lavin
- Centre for Clinical Research, The University of Queensland, Brisbane, Queensland 4072, Australia
| | - Gongchang Yu
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| | - Hua Shao
- Shandong Academy of Occupational Health and Occupational Medicine, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250062, P.R. China
| |
Collapse
|
88
|
Gauthier AG, Wu J, Lin M, Sitapara R, Kulkarni A, Thakur GA, Schmidt EE, Perron JC, Ashby CR, Mantell LL. The Positive Allosteric Modulation of alpha7-Nicotinic Cholinergic Receptors by GAT107 Increases Bacterial Lung Clearance in Hyperoxic Mice by Decreasing Oxidative Stress in Macrophages. Antioxidants (Basel) 2021; 10:135. [PMID: 33477969 PMCID: PMC7835977 DOI: 10.3390/antiox10010135] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 01/15/2021] [Indexed: 12/20/2022] Open
Abstract
Supplemental oxygen therapy with supraphysiological concentrations of oxygen (hyperoxia; >21% O2) is a life-saving intervention for patients experiencing respiratory distress. However, prolonged exposure to hyperoxia can compromise bacterial clearance processes, due to oxidative stress-mediated impairment of macrophages, contributing to the increased susceptibility to pulmonary infections. This study reports that the activation of the α7 nicotinic acetylcholine receptor (α7nAChR) with the delete allosteric agonistic-positive allosteric modulator, GAT107, decreases the bacterial burden in mouse lungs by improving hyperoxia-induced lung redox imbalance. The incubation of RAW 264.7 cells with GAT107 (3.3 µM) rescues hyperoxia-compromised phagocytic functions in cultured macrophages, RAW 264.7 cells, and primary bone marrow-derived macrophages. Similarly, GAT107 (3.3 µM) also attenuated oxidative stress in hyperoxia-exposed macrophages, which prevents oxidation and hyper-polymerization of phagosome filamentous actin (F-actin) from oxidation. Furthermore, GAT107 (3.3 µM) increases the (1) activity of superoxide dismutase 1; (2) activation of Nrf2 and (3) the expression of heme oxygenase-1 (HO-1) in macrophages exposed to hyperoxia. Overall, these data suggest that the novel α7nAChR compound, GAT107, could be used to improve host defense functions in patients, such as those with COVID-19, who are exposed to prolonged periods of hyperoxia.
Collapse
Affiliation(s)
- Alex G. Gauthier
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Jiaqi Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Mosi Lin
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Ravikumar Sitapara
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Abhijit Kulkarni
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (A.K.); (G.A.T.)
| | - Ganesh A. Thakur
- Department of Pharmaceutical Sciences, Northeastern University, Boston, MA 02115, USA; (A.K.); (G.A.T.)
| | - Edward E. Schmidt
- Department of Microbiology and Immunology, Montana State University, Bozeman, MT 59717, USA;
| | - Jeanette C. Perron
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Charles R. Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
| | - Lin L. Mantell
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John’s University, Queens, New York, NY 11439, USA; (A.G.G.); (J.W.); (M.L.); (R.S.); (J.C.P.); (C.R.A.J.)
- Feinstein Institute for Medical Research, Northwell Health, Manhasset, NY 11030, USA
| |
Collapse
|
89
|
Diesel Exhaust Particulates Induce Neutrophilic Lung Inflammation by Modulating Endoplasmic Reticulum Stress-Mediated CXCL1/KC Expression in Alveolar Macrophages. Molecules 2020; 25:molecules25246046. [PMID: 33371364 PMCID: PMC7767360 DOI: 10.3390/molecules25246046] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Revised: 12/11/2020] [Accepted: 12/14/2020] [Indexed: 12/25/2022] Open
Abstract
Diesel exhaust particulates (DEP) have adverse effects on the respiratory system. Endoplasmic reticulum (ER) abnormalities contribute to lung inflammation. However, the relationship between DEP exposure and ER stress in the respiratory immune system and especially the alveolar macrophages (AM) is poorly understood. Here, we examined ER stress and inflammatory responses using both in vivo and in vitro study. For in vivo study, mice were intratracheally instilled with 25, 50, and 100 μg DEP and in vitro AM were stimulated with DEP at 1, 2, and 3 mg/mL. DEP increased lung weight and the number of inflammatory cells, especially neutrophils, and inflammatory cytokines in bronchoalveolar lavage fluid of mice. DEP also increased the number of DEP-pigmented AM and ER stress markers including bound immunoglobulin protein (BiP) and CCAAT/enhancer binding protein-homologous protein (CHOP) were upregulated in the lungs of DEP-treated mice. In an in vitro study, DEP caused cell damage, increased intracellular reactive oxygen species, and upregulated inflammatory genes and ER stress-related BiP, CHOP, splicing X-box binding protein 1, and activating transcription factor 4 expressions in AM. Furthermore, DEP released the C-X-C Motif Chemokine Ligand 1 (CXCL1/KC) in AM. In conclusion, DEP may contribute to neutrophilic lung inflammation pathogenesis by modulating ER stress-mediated CXCL1/KC expression in AM.
Collapse
|
90
|
Pinkston R, Zaman H, Hossain E, Penn AL, Noël A. Cell-specific toxicity of short-term JUUL aerosol exposure to human bronchial epithelial cells and murine macrophages exposed at the air-liquid interface. Respir Res 2020; 21:269. [PMID: 33069224 PMCID: PMC7568376 DOI: 10.1186/s12931-020-01539-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/08/2020] [Indexed: 01/09/2023] Open
Abstract
Backgroud JUUL, an electronic nicotine delivery system (ENDS), which first appeared on the US market in 2015, controled more than 75% of the US ENDS sales in 2018. JUUL-type devices are currently the most commonly used form of ENDS among youth in the US. In contrast to free-base nicotine contained in cigarettes and other ENDS, JUUL contains high levels of nicotine salt (35 or 59 mg/mL), whose cellular and molecular effects on lung cells are largely unknown. In the present study, we evaluated the in vitro toxicity of JUUL crème brûlée-flavored aerosols on 2 types of human bronchial epithelial cell lines (BEAS-2B, H292) and a murine macrophage cell line (RAW 264.7). Methods Human lung epithelial cells and murine macrophages were exposed to JUUL crème brûlée-flavored aerosols at the air–liquid interface (ALI) for 1-h followed by a 24-h recovery period. Membrane integrity, cytotoxicity, extracellular release of nitrogen species and reactive oxygen species, cellular morphology and gene expression were assessed. Results Crème brûlée-flavored aerosol contained elevated concentrations of benzoic acid (86.9 μg/puff), a well-established respiratory irritant. In BEAS-2B cells, crème brûlée-flavored aerosol decreased cell viability (≥ 50%) and increased nitric oxide (NO) production (≥ 30%), as well as iNOS gene expression. Crème brûlée-flavored aerosol did not affect the viability of either H292 cells or RAW macrophages, but increased the production of reactive oxygen species (ROS) by ≥ 20% in both cell types. While crème brûlée-flavored aerosol did not alter NO levels in H292 cells, RAW macrophages exposed to crème brûlée-flavored aerosol displayed decreased NO (≥ 50%) and down-regulation of the iNOS gene, possibly due to increased ROS. Additionally, crème brûlée-flavored aerosol dysregulated the expression of several genes related to biotransformation, inflammation and airway remodeling, including CYP1A1, IL-6, and MMP12 in all 3 cell lines. Conclusion Our results indicate that crème brûlée-flavored aerosol causes cell-specific toxicity to lung cells. This study contributes to providing scientific evidence towards regulation of nicotine salt-based products.
Collapse
Affiliation(s)
- Rakeysha Pinkston
- Department of Environmental Toxicology, College of Sciences and Engineering, Southern University and A&M College, Baton Rouge, LA, 70813, USA.,Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Hasan Zaman
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Ekhtear Hossain
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Arthur L Penn
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA
| | - Alexandra Noël
- Department of Comparative Biomedical Sciences, School of Veterinary Medicine, Louisiana State University, 1909 Skip Bertman Drive, Baton Rouge, LA, 70803, USA.
| |
Collapse
|
91
|
Impaired mitochondrial function of alveolar macrophages in carbon nanotube-induced chronic pulmonary granulomatous disease. Toxicology 2020; 445:152598. [PMID: 32976959 DOI: 10.1016/j.tox.2020.152598] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Revised: 09/15/2020] [Accepted: 09/18/2020] [Indexed: 12/18/2022]
Abstract
Human exposure to carbon nanotubes (CNT) has been associated with the development of pulmonary sarcoid-like granulomatous disease. Our previous studies demonstrated that multi-walled carbon nanotubes (MWCNT) induced chronic pulmonary granulomatous inflammation in mice. Granuloma formation was accompanied by decreased peroxisome proliferator-activated receptor gamma (PPARγ) and disrupted intracellular lipid homeostasis in alveolar macrophages. Others have shown that PPARγ activation increases mitochondrial fatty acid oxidation (FAO) to reduce free fatty acid accumulation. Hence, we hypothesized that the disrupted lipid metabolism suppresses mitochondrial FAO. To test our hypothesis, C57BL/6 J mice were instilled by an oropharyngeal route with 100 μg MWCNT freshly suspended in 35 % Infasurf. Control sham mice received vehicle alone. Sixty days following instillation, mitochondrial FAO was measured in permeabilized bronchoalveolar lavage (BAL) cells. MWCNT instillation reduced the mitochondrial oxygen consumption rate of BAL cells in the presence of palmitoyl-carnitine as mitochondrial fuel. MWCNT also reduced mRNA expression of mitochondrial genes regulating FAO, carnitine palmitoyl transferase-1 (CPT1), carnitine palmitoyl transferase-2 (CPT2), hydroxyacyl-CoA dehydrogenase subunit beta (HADHB), and PPARγ coactivator 1 alpha (PPARGC1A). Importantly, both oxidative stress and apoptosis in alveolar macrophages and lung tissues of MWCNT-instilled mice were increased. Because macrophage PPARγ expression has been reported to be controlled by miR-27b which is known to induce oxidative stress and apoptosis, we measured the expression of miR-27b. Results indicated elevated levels in alveolar macrophages from MWCNT-instilled mice compared to controls. Given that inhibition of FAO and apoptosis are linked to M1 and M2 macrophage activation, respectively, the expression of both M1 and M2 key indicator genes were measured. Interestingly, results showed that both M1 and M2 phenotypes of alveolar macrophages were activated in MWCNT-instilled mice. In conclusion, alveolar macrophages of MWCNT-instilled mice had increased miR-27b expression, which may reduce the expression of PPARγ resulting in attenuation of FAO. This reduction in FAO may lead to activation of M1 macrophages. The upregulation of miR-27b may also induce apoptosis, which in turn can cause M2 activation of alveolar macrophages. These observations indicate a possible role of miR-27b in impaired mitochondrial function in the chronic activation of alveolar macrophages by MWCNT and the development of chronic pulmonary granulomatous inflammation.
Collapse
|
92
|
Ma R, Ortiz Serrano TP, Davis J, Prigge AD, Ridge KM. The cGAS-STING pathway: The role of self-DNA sensing in inflammatory lung disease. FASEB J 2020; 34:13156-13170. [PMID: 32860267 PMCID: PMC8121456 DOI: 10.1096/fj.202001607r] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/15/2022]
Abstract
The presence of DNA in the cytosol is usually a sign of microbial infections, which alerts the host innate immune system to mount a defense response. Cyclic GMP-AMP synthase (cGAS) is a critical cytosolic DNA sensor that elicits robust innate immune responses through the production of the second messenger, cyclic GMP-AMP (cGAMP), which binds and activates stimulator of interferon genes (STING). However, cGAS binds to DNA irrespective of DNA sequence, therefore, self-DNA leaked from the nucleus or mitochondria can also serve as a cGAS ligand to activate this pathway and trigger extensive inflammatory responses. Dysregulation of the cGAS-STING pathway is responsible for a broad array of inflammatory and autoimmune diseases. Recently, evidence has shown that self-DNA release and cGAS-STING pathway over-activation can drive lung disease, making this pathway a promising therapeutic target for inflammatory lung disease. Here, we review recent advances on the cGAS-STING pathway governing self-DNA sensing, highlighting its role in pulmonary disease.
Collapse
Affiliation(s)
- Ruihua Ma
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tatiana P Ortiz Serrano
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jennifer Davis
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Andrew D Prigge
- Division of Critical Care Medicine, Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL, USA
| | - Karen M Ridge
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.,Department of Cell and Developmental Biology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
93
|
Koureas M, Kirgou P, Amoutzias G, Hadjichristodoulou C, Gourgoulianis K, Tsakalof A. Target Analysis of Volatile Organic Compounds in Exhaled Breath for Lung Cancer Discrimination from Other Pulmonary Diseases and Healthy Persons. Metabolites 2020; 10:metabo10080317. [PMID: 32756521 PMCID: PMC7464039 DOI: 10.3390/metabo10080317] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/29/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023] Open
Abstract
The aim of the present study was to investigate the ability of breath analysis to distinguish lung cancer (LC) patients from patients with other respiratory diseases and healthy people. The population sample consisted of 51 patients with confirmed LC, 38 patients with pathological computed tomography (CT) findings not diagnosed with LC, and 53 healthy controls. The concentrations of 19 volatile organic compounds (VOCs) were quantified in the exhaled breath of study participants by solid phase microextraction (SPME) of the VOCs and subsequent gas chromatography-mass spectrometry (GC-MS) analysis. Kruskal-Wallis and Mann-Whitney tests were used to identify significant differences between subgroups. Machine learning methods were used to determine the discriminant power of the method. Several compounds were found to differ significantly between LC patients and healthy controls. Strong associations were identified for 2-propanol, 1-propanol, toluene, ethylbenzene, and styrene (p-values < 0.001-0.006). These associations remained significant when ambient air concentrations were subtracted from breath concentrations. VOC levels were found to be affected by ambient air concentrations and a few by smoking status. The random forest machine learning algorithm achieved a correct classification of patients of 88.5% (area under the curve-AUC 0.94). However, none of the methods used achieved adequate discrimination between LC patients and patients with abnormal computed tomography (CT) findings. Biomarker sets, consisting mainly of the exogenous monoaromatic compounds and 1- and 2- propanol, adequately discriminated LC patients from healthy controls. The breath concentrations of these compounds may reflect the alterations in patient's physiological and biochemical status and perhaps can be used as probes for the investigation of these statuses or normalization of patient-related factors in breath analysis.
Collapse
Affiliation(s)
- Michalis Koureas
- Department of Hygiene and Epidemiology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 22 Papakyriazi Street, 41222 Larissa, Greece; (M.K.); (C.H.)
| | - Paraskevi Kirgou
- Respiratory Medicine Department, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece; (P.K.); (K.G.)
| | - Grigoris Amoutzias
- Bioinformatics Laboratory, Department of Biochemistry and Biotechnology, University of Thessaly, 41500 Larissa, Greece;
| | - Christos Hadjichristodoulou
- Department of Hygiene and Epidemiology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 22 Papakyriazi Street, 41222 Larissa, Greece; (M.K.); (C.H.)
| | - Konstantinos Gourgoulianis
- Respiratory Medicine Department, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece; (P.K.); (K.G.)
| | - Andreas Tsakalof
- Department of Hygiene and Epidemiology, University Hospital of Larissa, Faculty of Medicine, University of Thessaly, 22 Papakyriazi Street, 41222 Larissa, Greece; (M.K.); (C.H.)
- Department of Biochemistry, Faculty of Medicine, University of Thessaly, 41500 Larissa, Greece
- Correspondence: ; Tel.: +30-2410685580
| |
Collapse
|
94
|
Ruwanpura SM, Thomas BJ, Bardin PG. Pirfenidone: Molecular Mechanisms and Potential Clinical Applications in Lung Disease. Am J Respir Cell Mol Biol 2020; 62:413-422. [PMID: 31967851 DOI: 10.1165/rcmb.2019-0328tr] [Citation(s) in RCA: 154] [Impact Index Per Article: 30.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Pirfenidone (PFD) is a pharmacological compound with therapeutic efficacy in idiopathic pulmonary fibrosis. It has been chiefly characterized as an antifibrotic agent, although it was initially developed as an antiinflammatory compound because of its ability to diminish the accumulation of inflammatory cells and cytokines. Despite recent studies that have elucidated key mechanisms, the precise molecular activities of PFD remain incompletely understood. PFD modulates fibrogenic growth factors, thereby attenuating fibroblast proliferation, myofibroblast differentiation, collagen and fibronectin synthesis, and deposition of extracellular matrix. This effect is mediated by suppression of TGF-β1 (transforming growth factor-β1) and other growth factors. Here, we appraise the impact of PFD on TGF-β1 production and its downstream pathways. Accumulating evidence indicates that PFD also downregulates inflammatory pathways and therefore has considerable potential as a viable and innovative antiinflammatory compound. We examine the effects of PFD on inflammatory cells and the production of pro- and antiinflammatory cytokines in the lung. In this context, recent evidence that PFD can target inflammasome pathways and ensuing lung inflammation is highlighted. Finally, the antioxidant properties of PFD, such as its ability to inhibit redox reactions and regulate oxidative stress-related genes and enzymes, are detailed. In summary, this narrative review examines molecular mechanisms underpinning PFD and its recognized benefits in lung fibrosis. We highlight preclinical data that demonstrate the potential of PFD as a nonsteroidal antiinflammatory agent and outline areas for future research.
Collapse
Affiliation(s)
- Saleela M Ruwanpura
- Monash Lung and Sleep, Monash Health, Monash Medical Centre, Clayton, Victoria, Australia; and
| | - Belinda J Thomas
- Monash Lung and Sleep, Monash Health, Monash Medical Centre, Clayton, Victoria, Australia; and.,Hudson Institute of Medical Research, Clayton, Victoria, Australia
| | - Philip G Bardin
- Monash Lung and Sleep, Monash Health, Monash Medical Centre, Clayton, Victoria, Australia; and.,Hudson Institute of Medical Research, Clayton, Victoria, Australia
| |
Collapse
|
95
|
Alomari MA, Alzoubi KH, Khabour OF. Differences in oxidative stress profile in adolescents smoking waterpipe versus cigarettes: The Irbid TRY Project. Physiol Rep 2020. [PMCID: PMC7484827 DOI: 10.14814/phy2.14512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Mahmoud A. Alomari
- Department of Physical Education Qatar University Doha Qatar
- Division of Physical Therapy Department of Rehabilitation Sciences Jordan University of Science and Technology Irbid Jordan
| | - Karem H. Alzoubi
- Department of Clinical Pharmacy Jordan University of Science and Technology Irbid Jordan
| | - Omar F. Khabour
- Department of Medical Laboratory Sciences Jordan University of Science and Technology Irbid Jordan
| |
Collapse
|
96
|
Chen Z, Tian R, She Z, Cai J, Li H. Role of oxidative stress in the pathogenesis of nonalcoholic fatty liver disease. Free Radic Biol Med 2020; 152:116-141. [PMID: 32156524 DOI: 10.1016/j.freeradbiomed.2020.02.025] [Citation(s) in RCA: 739] [Impact Index Per Article: 147.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2019] [Revised: 02/20/2020] [Accepted: 02/26/2020] [Indexed: 02/07/2023]
Abstract
Nonalcoholic fatty liver disease (NAFLD) has emerged as the most common chronic liver disease worldwide and is strongly associated with the presence of oxidative stress. Disturbances in lipid metabolism lead to hepatic lipid accumulation, which affects different reactive oxygen species (ROS) generators, including mitochondria, endoplasmic reticulum, and NADPH oxidase. Mitochondrial function adapts to NAFLD mainly through the downregulation of the electron transport chain (ETC) and the preserved or enhanced capacity of mitochondrial fatty acid oxidation, which stimulates ROS overproduction within different ETC components upstream of cytochrome c oxidase. However, non-ETC sources of ROS, in particular, fatty acid β-oxidation, appear to produce more ROS in hepatic metabolic diseases. Endoplasmic reticulum stress and NADPH oxidase alterations are also associated with NAFLD, but the degree of their contribution to oxidative stress in NAFLD remains unclear. Increased ROS generation induces changes in insulin sensitivity and in the expression and activity of key enzymes involved in lipid metabolism. Moreover, the interaction between redox signaling and innate immune signaling forms a complex network that regulates inflammatory responses. Based on the mechanistic view described above, this review summarizes the mechanisms that may account for the excessive production of ROS, the potential mechanistic roles of ROS that drive NAFLD progression, and therapeutic interventions that are related to oxidative stress.
Collapse
Affiliation(s)
- Ze Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Ruifeng Tian
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Zhigang She
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China
| | - Jingjing Cai
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Department of Cardiology, The Third Xiangya Hospital, Central South University, Changsha, 410013, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China
| | - Hongliang Li
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, PR China; Institute of Model Animals of Wuhan University, Wuhan, 430072, PR China; Basic Medical School, Wuhan University, Wuhan, 430071, PR China; Medical Research Institute, School of Medicine, Wuhan University, Wuhan, 430071, PR China.
| |
Collapse
|
97
|
Huguet-Casquero A, Moreno-Sastre M, López-Méndez TB, Gainza E, Pedraz JL. Encapsulation of Oleuropein in Nanostructured Lipid Carriers: Biocompatibility and Antioxidant Efficacy in Lung Epithelial Cells. Pharmaceutics 2020; 12:pharmaceutics12050429. [PMID: 32384817 PMCID: PMC7285197 DOI: 10.3390/pharmaceutics12050429] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 04/27/2020] [Accepted: 05/05/2020] [Indexed: 12/13/2022] Open
Abstract
Oxidative damage has been linked to a number of diseases. Oleuropein (OLE), a natural occurring polyphenol from olive leaves (Olea europaea L.), is known to be a potent antioxidant compound with inherent instability and compromised bioavailability. Therefore, in this work, nanostructured lipid carriers (NLCs) were proposed for OLE encapsulation to protect and improve its antioxidant efficacy. The lipid matrix, composed of olive oil and Precirol, was optimized prior to OLE encapsulation. The characterization of the optimized oleuropein-loaded NLCs (NLC-OLE) showed a mean size of 150 nm, a zeta potential of −21 mV, an encapsulation efficiency of 99.12%, sustained release profile, and improved radical scavenging activity. The cellular in vitro assays demonstrated the biocompatibility of the NLCs, which were found to improve and maintain OLE antioxidant efficacy in the A549 and CuFi-1 lung epithelial cell lines, respectively. Overall, these findings suggest a promising potential of NLC-OLE to further design a pulmonary formulation for OLE delivery in lung epithelia.
Collapse
Affiliation(s)
- Amaia Huguet-Casquero
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.H.-C.); (M.M.-S.); (T.B.L.-M.)
- Biosasun S.A., Iturralde 10, Etxabarri-Ibiña, 01006 Zigoitia, Spain;
| | - Maria Moreno-Sastre
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.H.-C.); (M.M.-S.); (T.B.L.-M.)
| | - Tania Belén López-Méndez
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.H.-C.); (M.M.-S.); (T.B.L.-M.)
| | - Eusebio Gainza
- Biosasun S.A., Iturralde 10, Etxabarri-Ibiña, 01006 Zigoitia, Spain;
| | - Jose Luis Pedraz
- NanoBioCel Group, Laboratory of Pharmaceutics, University of the Basque Country (UPV/EHU), School of Pharmacy, Paseo de la Universidad 7, 01006 Vitoria-Gasteiz, Spain; (A.H.-C.); (M.M.-S.); (T.B.L.-M.)
- Biomedical Research Networking Center in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), 01006 Vitoria-Gasteiz, Spain
- Correspondence:
| |
Collapse
|
98
|
Dustin CM, Heppner DE, Lin MCJ, van der Vliet A. Redox regulation of tyrosine kinase signalling: more than meets the eye. J Biochem 2020; 167:151-163. [PMID: 31599960 DOI: 10.1093/jb/mvz085] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 09/20/2019] [Indexed: 02/06/2023] Open
Abstract
Protein kinases are essential mediators of cellular signal transduction and are often dysregulated in disease. Among these, protein tyrosine kinases (PTKs) have received specific interest due to their common roles in various diseases including cancer, and emerging observations indicating that PTK signalling pathways are susceptible to regulation by reactive oxygen species (ROS), which are also frequently implicated in disease pathology. While it is well recognized that ROS can impact on tyrosine kinase signalling by inhibiting tyrosine phosphatases, more recent studies highlight additional modes of redox-based regulation of tyrosine kinase signalling by direct redox modification of non-catalytic cysteines within tyrosine kinases or other protein components of this signalling pathway. In this review, we will present recent advancements with respect to redox-based mechanisms in regulating PTK signalling, with a specific focus on recent studies demonstrating direct redox regulation of Src-family kinases and epidermal growth factor receptor kinases. Importantly, redox-based modulation of tyrosine kinases may be relevant for many other kinases and has implications for current approaches to develop pharmacological inhibitors for these proteins.
Collapse
Affiliation(s)
- Christopher M Dustin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - David E Heppner
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Miao-Chong J Lin
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, VT, USA
| |
Collapse
|
99
|
Sphingosine Kinase 1/S1P Signaling Contributes to Pulmonary Fibrosis by Activating Hippo/YAP Pathway and Mitochondrial Reactive Oxygen Species in Lung Fibroblasts. Int J Mol Sci 2020; 21:ijms21062064. [PMID: 32192225 PMCID: PMC7139883 DOI: 10.3390/ijms21062064] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 03/12/2020] [Accepted: 03/13/2020] [Indexed: 12/19/2022] Open
Abstract
The sphingosine kinase 1 (SPHK1)/sphingosine–1–phosphate (S1P) signaling axis is emerging as a key player in the development of idiopathic pulmonary fibrosis (IPF) and bleomycin (BLM)-induced lung fibrosis in mice. Recent evidence implicates the involvement of the Hippo/Yes-associated protein (YAP) 1 pathway in lung diseases, including IPF, but its plausible link to the SPHK1/S1P signaling pathway is unclear. Herein, we demonstrate the increased co-localization of YAP1 with the fibroblast marker FSP1 in the lung fibroblasts of BLM-challenged mice, and the genetic deletion of Sphk1 in mouse lung fibroblasts (MLFs) reduced YAP1 localization in fibrotic foci. The PF543 inhibition of SPHK1 activity in mice attenuated YAP1 co-localization with FSP1 in lung fibroblasts. In vitro, TGF-β stimulated YAP1 translocation to the nucleus in primary MLFs, and the deletion of Sphk1 or inhibition with PF543 attenuated TGF-β-mediated YAP1 nuclear localization. Moreover, the PF543 inhibition of SPHK1, or the verteporfin inhibition of YAP1, decreased the TGF-β- or BLM-induced mitochondrial reactive oxygen species (mtROS) in human lung fibroblasts (HLFs) and the expression of fibronectin (FN) and alpha-smooth muscle actin (α-SMA). Furthermore, scavenging mtROS with MitoTEMPO attenuated the TGF-β-induced expression of FN and α-SMA. The addition of the S1P antibody to HLFs reduced TGF-β- or S1P-mediated YAP1 activation, mtROS, and the expression of FN and α-SMA. These results suggest a role for SPHK1/S1P signaling in TGF-β-induced YAP1 activation and mtROS generation, resulting in fibroblast activation, a critical driver of pulmonary fibrosis.
Collapse
|
100
|
Tan KS, Lim RL, Liu J, Ong HH, Tan VJ, Lim HF, Chung KF, Adcock IM, Chow VT, Wang DY. Respiratory Viral Infections in Exacerbation of Chronic Airway Inflammatory Diseases: Novel Mechanisms and Insights From the Upper Airway Epithelium. Front Cell Dev Biol 2020; 8:99. [PMID: 32161756 PMCID: PMC7052386 DOI: 10.3389/fcell.2020.00099] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/07/2020] [Indexed: 12/16/2022] Open
Abstract
Respiratory virus infection is one of the major sources of exacerbation of chronic airway inflammatory diseases. These exacerbations are associated with high morbidity and even mortality worldwide. The current understanding on viral-induced exacerbations is that viral infection increases airway inflammation which aggravates disease symptoms. Recent advances in in vitro air-liquid interface 3D cultures, organoid cultures and the use of novel human and animal challenge models have evoked new understandings as to the mechanisms of viral exacerbations. In this review, we will focus on recent novel findings that elucidate how respiratory viral infections alter the epithelial barrier in the airways, the upper airway microbial environment, epigenetic modifications including miRNA modulation, and other changes in immune responses throughout the upper and lower airways. First, we reviewed the prevalence of different respiratory viral infections in causing exacerbations in chronic airway inflammatory diseases. Subsequently we also summarized how recent models have expanded our appreciation of the mechanisms of viral-induced exacerbations. Further we highlighted the importance of the virome within the airway microbiome environment and its impact on subsequent bacterial infection. This review consolidates the understanding of viral induced exacerbation in chronic airway inflammatory diseases and indicates pathways that may be targeted for more effective management of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Kai Sen Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Rachel Liyu Lim
- Infectious Disease Research and Training Office, National Centre for Infectious Diseases, Singapore, Singapore
| | - Jing Liu
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hsiao Hui Ong
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Vivian Jiayi Tan
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Hui Fang Lim
- Division of Respiratory and Critical Care Medicine, National University Hospital, Singapore, Singapore.,Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kian Fan Chung
- Airway Disease, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Ian M Adcock
- Airway Disease, National Heart and Lung Institute, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Vincent T Chow
- Department of Microbiology and Immunology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - De Yun Wang
- Department of Otolaryngology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|