1
|
Duvivier L, Gerard L, Diaz A, Gillet JP. Linking ABC transporters to the hallmarks of cancer. Trends Cancer 2024; 10:124-134. [PMID: 37884430 DOI: 10.1016/j.trecan.2023.09.013] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/28/2023]
Abstract
Human ATP-binding cassette (ABC) transporters are ubiquitously expressed and transport a broad range of endogenous and xenobiotic substrates across extra- and intracellular membranes. Mutations in ABC genes cause 21 monogenic diseases, and polymorphisms in these genes are associated with susceptibility to complex diseases. ABC transporters also play a major role in drug bioavailability, and they mediate multidrug resistance in cancer. At least 13 ABC transporters were shown to be involved in drug resistance in vitro. In the past decade, efforts have been made to elucidate their roles in tumor biology. Herein, we explore their involvement in tumorigenesis, focusing on the hallmarks of cells as they make their way from normalcy to neoplastic growth states.
Collapse
Affiliation(s)
- Laurent Duvivier
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Louise Gerard
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Adriana Diaz
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium
| | - Jean-Pierre Gillet
- Laboratory of Molecular Cancer Biology, URPhyM, NARILIS, University of Namur, Namur, Belgium.
| |
Collapse
|
2
|
Mokra D, Mokry J, Barosova R, Hanusrichterova J. Advances in the Use of N-Acetylcysteine in Chronic Respiratory Diseases. Antioxidants (Basel) 2023; 12:1713. [PMID: 37760016 PMCID: PMC10526097 DOI: 10.3390/antiox12091713] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Revised: 08/23/2023] [Accepted: 08/30/2023] [Indexed: 09/29/2023] Open
Abstract
N-acetylcysteine (NAC) is widely used because of its mucolytic effects, taking part in the therapeutic protocols of cystic fibrosis. NAC is also administered as an antidote in acetaminophen (paracetamol) overdosing. Thanks to its wide antioxidative and anti-inflammatory effects, NAC may also be of benefit in other chronic inflammatory and fibrotizing respiratory diseases, such as chronic obstructive pulmonary disease, bronchial asthma, idiopathic lung fibrosis, or lung silicosis. In addition, NAC exerts low toxicity and rare adverse effects even in combination with other treatments, and it is cheap and easily accessible. This article brings a review of information on the mechanisms of inflammation and oxidative stress in selected chronic respiratory diseases and discusses the use of NAC in these disorders.
Collapse
Affiliation(s)
- Daniela Mokra
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juraj Mokry
- Department of Pharmacology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia;
| | - Romana Barosova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| | - Juliana Hanusrichterova
- Department of Physiology, Jessenius Faculty of Medicine in Martin, Comenius University in Bratislava, SK-03601 Martin, Slovakia; (R.B.); (J.H.)
| |
Collapse
|
3
|
Gao X, Zhang X, Jiang L, Xu J, Liu W, Qian Y, Jiang Y, Jin Q, Hong H, Chen M, Jin Z, Wei Z, Yang Z, Zhang H. Forsythin inhibits β-hydroxybutyrate-induced oxidative stress in bovine macrophages by regulating p38/ERK, PI3K/Akt, and Nrf2/HO-1 signaling pathways. Res Vet Sci 2023; 154:59-65. [PMID: 36463586 DOI: 10.1016/j.rvsc.2022.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 10/03/2022] [Accepted: 11/19/2022] [Indexed: 11/23/2022]
Abstract
Ketosis is a metabolic disease of dairy cows in the perinatal period, β-hydroxybutyrate (β-HB) is the main component of ketosis. High levels of β-HB can trigger oxidative stress and inflammatory response in dairy cows, leading to decreased milk yield and multiple postpartum diseases. Forsythin (FOR), the major constituent of the herbal medicine Forsythia, has anti-inflammatory, anti-oxidant, and antiviral effects. FOR was demonstrated to have an antioxidant effect on PC12 cells. However, the effects of FOR on β-HB-stimulated bovine macrophages (BMs) has not been reported. Thus, the aim of the present study was to investigate the effects of FOR on β-HB-stimulated BMs. Firstly, the CCK8 test confirmed that FOR (50, 100, 200 μg/mL) has no effect on BMs activity, and we selected these concentrations for subsequent experiments. Secondly, through detecting the oxidation indexes ROS, MDA and antioxidant indexes CAT and SOD, we confirmed the antioxidant effect of FOR on BMs. Next, qRT-PCR confirmed that FOR dramatically reduced the mRNA levels of IL-1β and IL-6. Furthermore, the western blotting confirmed that FOR observably down-regulated β-HB-stimulated phosphorylation of p38, ERK and Akt and up-regulated expression of Nrf2, and HO-1. Above results suggested that FOR plays antioxidant effects on β-HB-induced BMs through p38, ERK and PI3K/Akt, Nrf2 and HO-1 signaling pathways. Therefore, we speculated that FOR may be a potential medicine to alleviate β-HB-induced inflammatory response and provide a preliminary reference for the research and development of FOR.
Collapse
Affiliation(s)
- Xinxin Gao
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Xu Zhang
- State Key Laboratory of Polymer Physics and Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, PR China
| | - Liqiang Jiang
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Jingnan Xu
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Wei Liu
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yuxiao Qian
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Yuqian Jiang
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Qinqin Jin
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Hongrong Hong
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Meiyi Chen
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zha Jin
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhengkai Wei
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China
| | - Zhengtao Yang
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China.
| | - Haoji Zhang
- College of Life Sciences and Engineering, Foshan University, Foshan 528225, Guangdong Province, PR China.
| |
Collapse
|
4
|
Hassel C, Couchet M, Jacquemot N, Blavignac C, Loï C, Moinard C, Cia D. Citrulline protects human retinal pigment epithelium from hydrogen peroxide and iron/ascorbate induced damages. J Cell Mol Med 2022; 26:2808-2818. [PMID: 35460170 PMCID: PMC9097847 DOI: 10.1111/jcmm.17294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 03/08/2022] [Accepted: 03/10/2022] [Indexed: 12/18/2022] Open
Abstract
Oxidative stress plays an important role in the ageing of the retina and in the pathogenesis of retinal diseases such as age‐related macular degeneration (ARMD). Hydrogen peroxide is a reactive oxygen species generated by the photo‐excited lipofuscin that accumulates during ageing in the retinal pigment epithelium (RPE), and the age‐related accumulation of lipofuscin is associated with ARMD. Iron also accumulates with age in the RPE that may contribute to ARMD as an important source of oxidative stress. The aim of this work was to investigate the effects of L‐Citrulline (CIT), a naturally occurring amino acid with known antioxidant properties, on oxidative stressed cultured RPE cells. Human RPE (ARPE‐19) cells were exposed to hydrogen peroxide (H2O2) or iron/ascorbate (I/A) for 4 h, either in the presence of CIT or after 24 h of pretreatment. Here, we show that supplementation with CIT protects ARPE‐19 cells against H2O2 and I/A. CIT improves cell metabolic activity, decreases ROS production, limits lipid peroxidation, reduces cell death and attenuates IL‐8 secretion. Our study evidences that CIT is able to protect human RPE cells from oxidative damage and suggests potential protective effect for the treatment of retinal diseases associated with oxidative stress.
Collapse
Affiliation(s)
- Chervin Hassel
- Université Clermont Auvergne, INSERM U1107 NEURO-DOL, Laboratoire de Biophysique Neurosensorielle, Clermont-Ferrand, France
| | - Morgane Couchet
- Université Grenoble-Alpes, INSERM U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, Grenoble, France
| | - Nathalie Jacquemot
- Université Clermont Auvergne, INSERM U1107 NEURO-DOL, Laboratoire de Biophysique Neurosensorielle, Clermont-Ferrand, France
| | - Christelle Blavignac
- Université Clermont Auvergne, Centre Imagerie Cellulaire Santé, Clermont-Ferrand, France
| | | | - Christophe Moinard
- Université Grenoble-Alpes, INSERM U1055, Laboratoire de Bioénergétique Fondamentale et Appliquée, Grenoble, France
| | - David Cia
- Université Clermont Auvergne, INSERM U1107 NEURO-DOL, Laboratoire de Biophysique Neurosensorielle, Clermont-Ferrand, France
| |
Collapse
|
5
|
黎 仙, 李 景, 张 媛, 张 罗. [The role of IL-8 in the chronic airway inflammation and its research progress]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2021; 35:1144-1148. [PMID: 34886635 PMCID: PMC10127644 DOI: 10.13201/j.issn.2096-7993.2021.12.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Indexed: 04/30/2023]
Abstract
Chronic rhinosinusitis(CRS), asthma and chronic obstructive pulmonary disease(COPD) are common chronic airway inflammatory diseases, which seriously affect patients' quality of life and bring heavy economic and social burden. Interleukin(IL) -8 is an important chemokine of neutrophils and other inflammatory cells, which plays an important role in the development of chronic airway inflammation. In this review, the inflammatory mechanisms involved in regulating IL-8 gene expression and the role of IL-8 in different airway chronic inflammation were reviewed.
Collapse
Affiliation(s)
- 仙 黎
- 首都医科大学附属北京同仁医院耳鼻咽喉头颈外科 教育部耳鼻咽喉头颈外科重点实验室 北京市耳鼻咽喉科研究所(北京,100730)
- 首都医科大学附属北京同仁医院(鼻)过敏科
| | - 景云 李
- 首都医科大学附属北京同仁医院耳鼻咽喉头颈外科 教育部耳鼻咽喉头颈外科重点实验室 北京市耳鼻咽喉科研究所(北京,100730)
| | - 媛 张
- 首都医科大学附属北京同仁医院耳鼻咽喉头颈外科 教育部耳鼻咽喉头颈外科重点实验室 北京市耳鼻咽喉科研究所(北京,100730)
- 首都医科大学附属北京同仁医院(鼻)过敏科
| | - 罗 张
- 首都医科大学附属北京同仁医院耳鼻咽喉头颈外科 教育部耳鼻咽喉头颈外科重点实验室 北京市耳鼻咽喉科研究所(北京,100730)
- 首都医科大学附属北京同仁医院(鼻)过敏科
| |
Collapse
|
6
|
García-Giménez JL, Garcés C, Romá-Mateo C, Pallardó FV. Oxidative stress-mediated alterations in histone post-translational modifications. Free Radic Biol Med 2021; 170:6-18. [PMID: 33689846 DOI: 10.1016/j.freeradbiomed.2021.02.027] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/16/2021] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Epigenetic regulation of gene expression provides a finely tuned response capacity for cells when undergoing environmental changes. However, in the context of human physiology or disease, any cellular imbalance that modulates homeostasis has the potential to trigger molecular changes that result either in physiological adaptation to a new situation or pathological conditions. These effects are partly due to alterations in the functionality of epigenetic regulators, which cause long-term and often heritable changes in cell lineages. As such, free radicals resulting from unbalanced/extended oxidative stress have been proved to act as modulators of epigenetic agents, resulting in alterations of the epigenetic landscape. In the present review we will focus on the particular effect that oxidative stress and free radicals produce in histone post-translational modifications that contribute to altering the histone code and, consequently, gene expression. The pathological consequences of the changes in this epigenetic layer of regulation of gene expression are thoroughly evidenced by data gathered in many physiological adaptive processes and in human diseases that range from age-related neurodegenerative pathologies to cancer, and that include respiratory syndromes, infertility, and systemic inflammatory conditions like sepsis.
Collapse
Affiliation(s)
- José-Luis García-Giménez
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia- INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Concepción Garcés
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia- INCLIVA, Valencia, 46010, Spain
| | - Carlos Romá-Mateo
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia- INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain
| | - Federico V Pallardó
- Department of Physiology, Faculty of Medicine and Dentistry. University of Valencia- INCLIVA, Valencia, 46010, Spain; Associated Unit for Rare Diseases INCLIVA-CIPF, Valencia, Spain; CIBER de Enfermedades Raras (CIBERER), Valencia, Spain.
| |
Collapse
|
7
|
Bekeschus S, Clemen R, Haralambiev L, Niessner F, Grabarczyk P, Weltmann KD, Menz J, Stope M, von Woedtke T, Gandhirajan R, Schmidt A. The Plasma-Induced Leukemia Cell Death is Dictated by the ROS Chemistry and the HO-1/CXCL8 Axis. IEEE TRANSACTIONS ON RADIATION AND PLASMA MEDICAL SCIENCES 2021. [DOI: 10.1109/trpms.2020.3020686] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
8
|
Wu CL, Yin R, Wang SN, Ying R. A Review of CXCL1 in Cardiac Fibrosis. Front Cardiovasc Med 2021; 8:674498. [PMID: 33996954 PMCID: PMC8113392 DOI: 10.3389/fcvm.2021.674498] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Accepted: 04/01/2021] [Indexed: 12/31/2022] Open
Abstract
Chemokine C-X-C motif ligand-1 (CXCL1), principally expressed in neutrophils, macrophages and epithelial cells, is a valid pro-inflammatory factor which performs an important role in mediating the infiltration of neutrophils and monocytes/macrophages. Elevated serum level of CXCL1 is considered a pro-inflammatory reaction by the organism. CXCL1 is also related to diverse organs fibrosis according to relevant studies. A growing body of evidence suggests that CXCL1 promotes the process of cardiac remodeling and fibrosis. Here, we review structure and physiological functions of CXCL1 and recent progress on the effects and mechanisms of CXCL1 in cardiac fibrosis. In addition, we explore the role of CXCL1 in the fibrosis of other organs. Besides, we probe the possibility that CXCL1 can be a therapeutic target for the treatment of cardiac fibrosis in cardiovascular diseases.
Collapse
Affiliation(s)
- Cheng-Long Wu
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ran Yin
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Su-Nan Wang
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ru Ying
- Department of Cardiology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|
9
|
Briottet M, Shum M, Urbach V. The Role of Specialized Pro-Resolving Mediators in Cystic Fibrosis Airways Disease. Front Pharmacol 2020; 11:1290. [PMID: 32982730 PMCID: PMC7493015 DOI: 10.3389/fphar.2020.01290] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 08/04/2020] [Indexed: 12/26/2022] Open
Abstract
Cystic Fibrosis (CF) is a recessive genetic disease due to mutations of the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) gene encoding the CFTR chloride channel. The ion transport abnormalities related to CFTR mutation generate a dehydrated airway surface liquid (ASL) layer, which is responsible for an altered mucociliary clearance, favors infections and persistent inflammation that lead to progressive lung destruction and respiratory failure. The inflammatory response is normally followed by an active resolution phase to return to tissue homeostasis, which involves specialized pro-resolving mediators (SPMs). SPMs promote resolution of inflammation, clearance of microbes, tissue regeneration and reduce pain, but do not evoke unwanted immunosuppression. The airways of CF patients showed a decreased production of SPMs even in the absence of pathogens. SPMs levels in the airway correlated with CF patients' lung function. The prognosis for CF has greatly improved but there remains a critical need for more effective treatments that prevent excessive inflammation, lung damage, and declining pulmonary function for all CF patients. This review aims to highlight the recent understanding of CF airway inflammation and the possible impact of SPMs on functions that are altered in CF airways.
Collapse
Affiliation(s)
| | | | - Valerie Urbach
- Institut national de la santé et de la recherche médicale (Inserm) U955, Institut Mondor de Recherche Biomédicale (IMRB), Créteil, France
| |
Collapse
|
10
|
Khalaf M, Scott-Ward T, Causer A, Saynor Z, Shepherd A, Górecki D, Lewis A, Laight D, Shute J. Cystic Fibrosis Transmembrane Conductance Regulator (CFTR) in Human Lung Microvascular Endothelial Cells Controls Oxidative Stress, Reactive Oxygen-Mediated Cell Signaling and Inflammatory Responses. Front Physiol 2020; 11:879. [PMID: 32848840 PMCID: PMC7403513 DOI: 10.3389/fphys.2020.00879] [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: 07/30/2019] [Accepted: 06/29/2020] [Indexed: 12/19/2022] Open
Abstract
Background Perturbation of endothelial function in people with cystic fibrosis (CF) has been reported, which may be associated with endothelial cell expression of the cystic fibrosis transmembrane conductance regulator (CFTR). Previous reports indicate that CFTR activity upregulates endothelial barrier function, endothelial nitric oxide synthase (eNOS) expression and NO release, while limiting interleukin-8 (IL-8) release, in human umbilical vein endothelial cells (HUVECs) in cell culture. In view of reported microvascular dysfunction in people with CF we investigated the role of CFTR expression and activity in the regulation of oxidative stress, cell signaling and inflammation in human lung microvascular endothelial cells (HLMVECs) in cell culture. Methods HLMVECs were cultured in the absence and presence of the CFTR inhibitor GlyH-101 and CFTR siRNA. CFTR expression was analyzed using qRT-PCR, immunocytochemistry (IHC) and western blot, and function by membrane potential assay. IL-8 expression was analyzed using qRT-PCR and ELISA. Nrf2 expression, and NF-κB and AP-1 activation were determined using IHC and western blot. The role of the epidermal growth factor receptor (EGFR) in CFTR signaling was investigated using the EGFR tyrosine kinase inhibitor AG1478. Oxidative stress was measured as intracellular ROS and hydrogen peroxide (H2O2) concentration. VEGF and SOD-2 were measured in culture supernatants by ELISA. Results HLMVECs express low levels of CFTR that increase following inhibition of CFTR activity. Inhibition of CFTR, significantly increased intracellular ROS and H2O2 levels over 30 min and significantly decreased Nrf2 expression by 70% while increasing SOD-2 expression over 24 h. CFTR siRNA significantly increased constitutive expression of IL-8 by HLMVECs. CFTR inhibition activated the AP-1 pathway and increased IL-8 expression, without effect on NF-κB activity. Conversely, TNF-α activated the NF-κB pathway and increased IL-8 expression. The effects of TNF-α and GlyH-101 on IL-8 expression were additive and inhibited by AG1478. Inhibition of both CFTR and EGFR in HLMVECs significantly increased VEGF expression. The antioxidant N-acetyl cysteine significantly reduced ROS production and the increase in IL-8 and VEGF expression following CFTR inhibition. Conclusion Functional endothelial CFTR limits oxidative stress and contributes to the normal anti-inflammatory state of HLMVECs. Therapeutic strategies to restore endothelial CFTR function in CF are warranted.
Collapse
Affiliation(s)
- Maha Khalaf
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Toby Scott-Ward
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Adam Causer
- Department of Sport and Exercise Science, University of Portsmouth, Portsmouth, United Kingdom
| | - Zoe Saynor
- Department of Sport and Exercise Science, University of Portsmouth, Portsmouth, United Kingdom
| | - Anthony Shepherd
- Department of Sport and Exercise Science, University of Portsmouth, Portsmouth, United Kingdom
| | - Dariusz Górecki
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Anthony Lewis
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - David Laight
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| | - Janis Shute
- School of Pharmacy and Biomedical Sciences, Institute of Biological and Biomedical Sciences, University of Portsmouth, Portsmouth, United Kingdom
| |
Collapse
|
11
|
Yang J, Gong Y, Cai J, Zheng Y, Liu H, Zhang Z. Chlorpyrifos induces redox imbalance-dependent inflammation in common carp lymphocyte through dysfunction of T-cell receptor γ. JOURNAL OF FISH DISEASES 2020; 43:423-430. [PMID: 32048311 DOI: 10.1111/jfd.13138] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/07/2020] [Accepted: 01/07/2020] [Indexed: 06/10/2023]
Abstract
Chlorpyrifos is a poisonous pesticide that is highly toxic to fish and aquatic organisms. However, there are fewer reports about how chlorpyrifos influences the redox balance of immune cells. Herein, the head kidney tissue treated with chlorpyrifos to do transcriptome analysis and TCR γ was screened out. Subsequently, we established TCR γ knockdown and overexpression carp head kidney lymphocyte models, respectively, by using RNA interference and pcDNA3.1. Real-time PCR, fluorescent staining, oxidation and antioxidant kit were used to detect the related factors. We found that TCR γ knockdown significantly increased the mRNA expression of HSP70 and HSP90 and decreased the mRNA expression of SOD and CAT. Meanwhile, TCR γ knockdown reduced the activities of GSH, GSG-PX, T-AOC, CAT and SOD and increased the content of MDA and H2 O2 and activities of iNOS. Adverse results were obtained in TCR γ overexpression group. Additionally, TCR γ knockdown significantly increased the mRNA expression of IFN-γ, IL-1β, IL-8, IL-10, Nrf2 and NF-κB, but relieved TCR γ overexpression, in which the process of inflammation was activated. Our results reported here indicated that chlorpyrifos induces redox imbalance-dependent inflammation in common carp lymphocyte through dysfunction of T-cell receptor γ, and HSPs play potential protective role in entire process.
Collapse
Affiliation(s)
- Jie Yang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yafan Gong
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Jingzeng Cai
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Yingying Zheng
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| | - Honggui Liu
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- College of Animal Science and Technology, Northeast Agricultural University, Harbin, China
| | - Ziwei Zhang
- College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
- Key Laboratory of the Provincial Education Department of Heilongjiang for Common Animal Disease Prevention and Treatment, College of Veterinary Medicine, Northeast Agricultural University, Harbin, China
| |
Collapse
|
12
|
Murata M, Noda K, Yoshida S, Saito M, Fujiya A, Kanda A, Ishida S. Unsaturated Aldehyde Acrolein Promotes Retinal Glial Cell Migration. Invest Ophthalmol Vis Sci 2020; 60:4425-4435. [PMID: 31652327 DOI: 10.1167/iovs.19-27346] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose To investigate the effect of the unsaturated aldehyde acrolein on retinal glial cell migration. Methods Müller glial cell markers expression in TR-MUL5 were confirmed by RT-PCR and immunostaining. Cell viability and migration rate of TR-MUL5 cells were assessed after the stimulation with acrolein. DNA microarray analysis was performed to analyze changes in the expression levels of migration-related genes in Müller glial cells stimulated with acrolein. Real-time PCR and ELISA were performed to validate DNA microarray analysis results. Inhibitors of C-X-C motif chemokine ligand 1 (CXCL1), one of the genes highly upregulated after the exposure to acrolein, and blockers of its receptor, CXCR2, were used to investigate the role of the CXCL1-CXCR2 axis on glial cell migration. CXCL1 concentration was measured in vitreous fluid samples obtained from proliferative diabetic retinopathy (PDR) and nondiabetic control eyes. CXCL1 and CXCR2 expression in glial cells of fibrovascular tissues obtained from PDR patients was examined by immunostaining. Results At a high concentration, acrolein (100 μM) significantly decreased cell viability. However, in moderate, sublethal concentrations (25-50 μM), acrolein induced cell migration and substantially increased the production of CXCL1 in TR-MUL5 cells. CXCL1 concentration was significantly elevated in vitreous fluids of PDR patients, and CXCL1 and CXCR2 were present in glial cells in fibrovascular tissues of PDR patients. CXCL1 stimulation increased glial cell migration in a dose-dependent manner, which was abrogated by the neutralization of the CXCL1-CXCR2 axis. Conclusions Our data demonstrate that acrolein promotes retinal Müller glial cell migration by enhancing CXCL1 production.
Collapse
Affiliation(s)
- Miyuki Murata
- Laboratory of Ocular Cell Biology and Visual Science, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Kousuke Noda
- Laboratory of Ocular Cell Biology and Visual Science, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Shiho Yoshida
- Laboratory of Ocular Cell Biology and Visual Science, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Michiyuki Saito
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akio Fujiya
- Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Atsuhiro Kanda
- Laboratory of Ocular Cell Biology and Visual Science, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Susumu Ishida
- Laboratory of Ocular Cell Biology and Visual Science, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan.,Department of Ophthalmology, Faculty of Medicine and Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| |
Collapse
|
13
|
Li G, Liao Y, Hu J, Lu L, Zhang Y, Li B, An T. Activation of NF-κB pathways mediating the inflammation and pulmonary diseases associated with atmospheric methylamine exposure. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2019; 252:1216-1224. [PMID: 31252119 DOI: 10.1016/j.envpol.2019.06.059] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 06/13/2019] [Accepted: 06/13/2019] [Indexed: 06/09/2023]
Abstract
The effects of methylamine on human health have been debated for several years, but the exact adverse outcomes and definite signaling cascades have not been elucidated yet. Herein, a NF-κB signal pathway, a positive regulator of inflammation was identified as the main pathway of methylamine exposure induced adverse effects in bronchial airway cells (16HBE) for the first time. The results indicated that methylamine could stimulate the overproduction of reactive oxygen species (ROS) in cytoplasm and mitochondria of 16HBE cells. Moreover, ROS accelerate the translocation and phosphorylation of NF-κB in nucleic and promote the expression of inflammatory, such as IL-8 and IL-6. As a result, methylamine was found to be increased ROS-mediated NF-κB activation in cells, leading to the production of inflammatory cytokine. Furthermore, the results also showed that methylamine could affect the expression of cytokines related genes, p53, STAT3, Bcl2, c-myc, Cyclin D, Hes1, Mcl-1, TGF-β2. The breakdown of those cell proliferation and apoptosis related genes were leading to a common toxic mechanism of cell death. In summary, our work uncovers a mechanism by which methylamine can induce the formation of inflammation response and demonstrates potential inflammation and carcinogenesis in human airway cell upon the methylamine inhaled.
Collapse
Affiliation(s)
- Guiying Li
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yi Liao
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China
| | - Junjie Hu
- School of Environment and Civil Engineering, Dongguan University of Technology, Dongguan, 523808, China
| | - Lirong Lu
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China
| | - Yanan Zhang
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China
| | - Bing Li
- Experimental Medical Research Centre, Guangzhou Medical University, Guangzhou, 510182, China
| | - Taicheng An
- Guangzhou Key Laboratory of Environmental Catalysis and Pollution Control, Guangdong Key Laboratory of Environmental Catalysis and Health Risk Control, School of Environmental Science and Engineering, Institute of Environmental Health and Pollution Control, Guangdong University of Technology, Guangzhou, 510006, China.
| |
Collapse
|
14
|
Worgall S. Stranger in a Strange Land: IL-8 in the Mouse Lung? Am J Respir Cell Mol Biol 2019; 59:525-526. [PMID: 30095972 DOI: 10.1165/rcmb.2018-0206ed] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Stefan Worgall
- 1 Department of Pediatrics Weill Cornell Medicine New York, New York
| |
Collapse
|
15
|
Hao S, Roesch EA, Perez A, Weiner RL, Henderson LC, Cummings L, Consiglio P, Pajka J, Eisenberg A, Yeh L, Cotton CU, Drumm ML. Inactivation of CFTR by CRISPR/Cas9 alters transcriptional regulation of inflammatory pathways and other networks. J Cyst Fibros 2019; 19:34-39. [PMID: 31126900 DOI: 10.1016/j.jcf.2019.05.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 04/24/2019] [Accepted: 05/02/2019] [Indexed: 12/31/2022]
Abstract
BACKGROUND Individuals with cystic fibrosis (CF) experience elevated inflammation in multiple organs, but whether this reflects an inherent feature of CF cells or is a consequence of a pro-inflammatory environment is not clear. METHOD Using CRISPR/Cas9-mediated mutagenesis of CFTR, 17 subclonal cell lines were generated from Caco-2 cells. Clonal lines with functional CFTR (CFTR+) were compared to those without (CFTR-) to directly address the role of CFTR in inflammatory gene regulation. RESULTS All lines maintained CFTR mRNA production and formation of tight junctions. CFTR+ lines displayed short circuit currents in response to forskolin, while the CFTR- lines did not. Baseline expression of cytokines IL6 and CXCL8 (IL8) was not different between the lines regardless of CFTR genotype. All lines responded to TNFα and IL1β by increasing IL6 and CXCL8 mRNA levels, but the CFTR- lines produced more CXCL8 mRNA than the CFTR+ lines. Transcriptomes of 6 CFTR- and 6 CFTR+ lines, before and after stimulation by TNFα, were compared for differential expression as a function of CFTR genotype. While some genes appeared to be differentially expressed simply because of CFTR's absence, others required stimulation for differences to be apparent. CONCLUSION Together, these data suggest cells respond to CFTR's absence by modulating transcriptional networks, some of which are only apparent when cells are exposed to different environmental contexts, such as inflammation. With regards to inflammation, these data suggest a model in which CFTR's absence leads to a poised, pro-inflammatory state of cells that is only revealed by stimulation.
Collapse
Affiliation(s)
- Shuyu Hao
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Erica A Roesch
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Aura Perez
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Rebecca L Weiner
- Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Leigh C Henderson
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Linda Cummings
- Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Paul Consiglio
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Joseph Pajka
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Amy Eisenberg
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Lauren Yeh
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Calvin U Cotton
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America
| | - Mitchell L Drumm
- Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44106, United States of America; Department of Pediatrics, Case Western Reserve University, Cleveland, OH 44106, United States of America.
| |
Collapse
|
16
|
Larson ED, Magno JPM, Steritz MJ, Llanes EGDV, Cardwell J, Pedro M, Roberts TB, Einarsdottir E, Rosanes RAQ, Greenlee C, Santos RAP, Yousaf A, Streubel SO, Santos ATR, Ruiz AG, Lagrana-Villagracia SM, Ray D, Yarza TKL, Scholes MA, Anderson CB, Acharya A, Gubbels SP, Bamshad MJ, Cass SP, Lee NR, Shaikh RS, Nickerson DA, Mohlke KL, Prager JD, Cruz TLG, Yoon PJ, Abes GT, Schwartz DA, Chan AL, Wine TM, Cutiongco-de la Paz EM, Friedman N, Kechris K, Kere J, Leal SM, Yang IV, Patel JA, Tantoco MLC, Riazuddin S, Chan KH, Mattila PS, Reyes-Quintos MRT, Ahmed ZM, Jenkins HA, Chonmaitree T, Hafrén L, Chiong CM, Santos-Cortez RLP. A2ML1 and otitis media: novel variants, differential expression, and relevant pathways. Hum Mutat 2019; 40:1156-1171. [PMID: 31009165 DOI: 10.1002/humu.23769] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Revised: 02/26/2019] [Accepted: 04/18/2019] [Indexed: 12/16/2022]
Abstract
A genetic basis for otitis media is established, however, the role of rare variants in disease etiology is largely unknown. Previously a duplication variant within A2ML1 was identified as a significant risk factor for otitis media in an indigenous Filipino population and in US children. In this report exome and Sanger sequencing was performed using DNA samples from the indigenous Filipino population, Filipino cochlear implantees, US probands, Finnish, and Pakistani families with otitis media. Sixteen novel, damaging A2ML1 variants identified in otitis media patients were rare or low-frequency in population-matched controls. In the indigenous population, both gingivitis and A2ML1 variants including the known duplication variant and the novel splice variant c.4061 + 1 G>C were independently associated with otitis media. Sequencing of salivary RNA samples from indigenous Filipinos demonstrated lower A2ML1 expression according to the carriage of A2ML1 variants. Sequencing of additional salivary RNA samples from US patients with otitis media revealed differentially expressed genes that are highly correlated with A2ML1 expression levels. In particular, RND3 is upregulated in both A2ML1 variant carriers and high-A2ML1 expressors. These findings support a role for A2ML1 in keratinocyte differentiation within the middle ear as part of otitis media pathology and the potential application of ROCK inhibition in otitis media.
Collapse
Affiliation(s)
- Eric D Larson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Jose Pedrito M Magno
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines
| | - Matthew J Steritz
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Erasmo Gonzalo D V Llanes
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Jonathan Cardwell
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Melquiadesa Pedro
- Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Tori Bootpetch Roberts
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Elisabet Einarsdottir
- Folkhälsan Institute of Genetics and Molecular Neurology Research Program, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Rose Anne Q Rosanes
- Department of Community Dentistry, College of Dentistry, University of the Philippines Manila, Manila, Philippines
| | - Christopher Greenlee
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | | | - Ayesha Yousaf
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Punjab, Pakistan
| | - Sven-Olrik Streubel
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | | | - Amanda G Ruiz
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Sheryl Mae Lagrana-Villagracia
- Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Dylan Ray
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Talitha Karisse L Yarza
- Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Newborn Hearing Screening Reference Center, University of the Philippines Manila - National Institutes of Health (NIH), Manila, Philippines
| | - Melissa A Scholes
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Catherine B Anderson
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Anushree Acharya
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | | | - Samuel P Gubbels
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Michael J Bamshad
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Stephen P Cass
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Nanette R Lee
- USC-Office of Population Studies Foundation, Inc. and Department of Anthropology, Sociology and History, University of San Carlos, Cebu, Philippines
| | - Rehan S Shaikh
- Institute of Molecular Biology and Biotechnology, Bahauddin Zakariya University, Multan, Punjab, Pakistan
| | - Deborah A Nickerson
- Department of Genome Sciences, University of Washington, Seattle, Washington
| | - Karen L Mohlke
- Department of Genetics, University of North Carolina, Chapel Hill, North Carolina
| | - Jeremy D Prager
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Teresa Luisa G Cruz
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Patricia J Yoon
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Generoso T Abes
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - David A Schwartz
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Abner L Chan
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Todd M Wine
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Eva Maria Cutiongco-de la Paz
- Philippine Genome Center, University of the Philippines, Quezon City, Philippines.,University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Norman Friedman
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Katerina Kechris
- Department of Biostatistics and Bioinformatics, Colorado School of Public Health, Aurora, Colorado
| | - Juha Kere
- Folkhälsan Institute of Genetics and Molecular Neurology Research Program, University of Helsinki, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Huddinge, Sweden
| | - Suzanne M Leal
- Center for Statistical Genetics, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Ivana V Yang
- Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado
| | - Janak A Patel
- Division of Infectious Diseases, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Ma Leah C Tantoco
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Saima Riazuddin
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Kenny H Chan
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Department of Pediatric Otolaryngology, Children's Hospital Colorado, Aurora, Colorado
| | - Petri S Mattila
- Department of Otorhinolaryngology, Head & Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Maria Rina T Reyes-Quintos
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Newborn Hearing Screening Reference Center, University of the Philippines Manila - National Institutes of Health (NIH), Manila, Philippines.,University of the Philippines Manila - National Institutes of Health, Manila, Philippines
| | - Zubair M Ahmed
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Maryland School of Medicine, Baltimore, Maryland
| | - Herman A Jenkins
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado
| | - Tasnee Chonmaitree
- Division of Infectious Diseases, Department of Pediatrics, University of Texas Medical Branch, Galveston, Texas
| | - Lena Hafrén
- Department of Otorhinolaryngology, Head & Neck Surgery, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Charlotte M Chiong
- Department of Otorhinolaryngology, University of the Philippines Manila College of Medicine - Philippine General Hospital, Manila, Philippines.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Newborn Hearing Screening Reference Center, University of the Philippines Manila - National Institutes of Health (NIH), Manila, Philippines
| | - Regie Lyn P Santos-Cortez
- Department of Otolaryngology, University of Colorado School of Medicine, Aurora, Colorado.,Philippine National Ear Institute, University of the Philippines Manila - National Institutes of Health, Manila, Philippines.,Center for Children's Surgery, Children's Hospital Colorado, Aurora, Colorado
| |
Collapse
|
17
|
Mi X, Tang W, Chen X, Liu F, Tang X. Mitofusin 2 attenuates the histone acetylation at collagen IV promoter in diabetic nephropathy. J Mol Endocrinol 2016; 57:233-249. [PMID: 27997345 DOI: 10.1530/jme-16-0031] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2016] [Accepted: 10/03/2016] [Indexed: 12/15/2022]
Abstract
Extracellular matrix (ECM) increase in diabetic nephropathy (DN) is closely related to mitochondrial dysfunction. The mechanism of protective function of mitofusin 2 (Mfn2) for mitochondria remains largely unknown. In this study, the molecular mechanisms for the effect of Mfn2 on mitochondria and subsequent collagen IV expression in DN were investigated. Ras-binding-deficient mitofusin 2 (Mfn2-Ras(Δ)) were overexpressed in rat glomerular mesangial cells, and then the cells were detected for mitochondrial morphology, cellular reactive oxygen species (ROS), mRNA and protein expression of collagen IV with advanced glycation end-product (AGE) stimulation. Preliminary results reveal that the mitochondrial dysfunction and the increased synthesis of collagen IV after AGE stimulation were reverted by Mfn2-Ras(Δ) overexpression. Bioinformatical computations were performed to search transcriptional factor motifs in the promoter region of collagen IV. Three specific regions for TFAP2A binding were identified, followed by validation with chromatin immunoprecipitation experiments. Knocking down TFAP2A significantly decreased the TF binding in the first two regions and the gene expression of collagen IV. Furthermore, results reveal that Mfn2-Ras(Δ) overexpression significantly mitigated TFAP2A binding and also reverted the histone acetylation at Regions 1 and 2 after AGE stimulation. In streptozotocin-induced diabetic rats, Mfn2-Ras(Δ) overexpression also ameliorated glomerular mesangial lesions with decreased collagen IV expression, accompanied by decreased acetylation and TFAP2A binding at Region 1. In conclusion, this study highlights the pathway by which mitochondria affect the histone acetylation of gene promoter and provides a new potential therapy approach for DN.
Collapse
Affiliation(s)
- Xuhua Mi
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Wanxin Tang
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Xiaolei Chen
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Fei Liu
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| | - Xiaohong Tang
- Division of NephrologyWest China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
18
|
Sharma A, Xu Y, Sung B, Vincent CT, Worgall T, Worgall S. Regulation of the Coxsackie and adenovirus receptor expression is dependent on cystic fibrosis transmembrane regulator in airway epithelial cells. Cell Microbiol 2016; 19. [PMID: 27527752 DOI: 10.1111/cmi.12654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Revised: 08/08/2016] [Accepted: 08/10/2016] [Indexed: 12/21/2022]
Abstract
The coxsackievirus and adenovirus receptor (CAR), in addition to serving as viral receptor, is a component of tight junctions and plays an important role in tissue homeostasis. Defects in the cystic fibrosis transmembrane regulator (CFTR) in lung epithelial cells are linked to inflammation and susceptibility for respiratory tract infections. Here, we demonstrate that CAR expression and infectivity with adenovirus (Ad) are increased in cystic fibrosis airway epithelial cells. Inhibition of CFTR or histone deacetylase (HDAC) enhanced CAR expression while CFTR overexpression or restoration of the diminished HDAC activity in cystic fibrosis cells reduced CAR expression. This connects the CFTR to CAR expression and infectivity with adenovirus through HDAC.
Collapse
Affiliation(s)
- Anurag Sharma
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Yaqin Xu
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA
| | - Biin Sung
- Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| | - C Theresa Vincent
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York, USA.,Department of Pharmacology and Physiology, Karolinska Institute, Stockholm, Sweden
| | - Tilla Worgall
- Department of Pathology, Columbia University, New York, New York, USA
| | - Stefan Worgall
- Department of Pediatrics, Weill Cornell Medicine, New York, New York, USA.,Department of Genetic Medicine, Weill Cornell Medicine, New York, New York, USA
| |
Collapse
|
19
|
Poghosyan A, Patel JK, Clifford RL, Knox AJ. Epigenetic dysregulation of interleukin 8 (CXCL8) hypersecretion in cystic fibrosis airway epithelial cells. Biochem Biophys Res Commun 2016; 476:431-437. [PMID: 27240956 DOI: 10.1016/j.bbrc.2016.05.140] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 05/26/2016] [Indexed: 12/13/2022]
Abstract
Airway epithelial cells in cystic fibrosis (CF) overexpress Interleukin 8 (CXCL8) through poorly defined mechanisms. CXCL8 transcription is dependent on coordinated binding of CCAAT/enhancer binding protein (C/EBP)β, nuclear factor (NF)-κB, and activator protein (AP)-1 to the promoter. Here we show abnormal epigenetic regulation is responsible for CXCL8 overexpression in CF cells. Under basal conditions CF cells had increased bromodomain (Brd)3 and Brd4 recruitment and enhanced NF-κB and C/EBPβ binding to the CXCL8 promoter compared to non-CF cells due to trimethylation of histone H3 at lysine 4 (H3K4me3) and DNA hypomethylation at CpG6. IL-1β increased NF-κB, C/EBPβ and Brd4 binding. Furthermore, inhibitors of bromodomain and extra-terminal domain family (BET) proteins reduced CXCL8 production in CF cells suggesting a therapeutic target for the BET pathway.
Collapse
Affiliation(s)
- Anna Poghosyan
- Division of Respiratory Medicine, Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Nottingham, NG5 1PB, United Kingdom.
| | - Jamie K Patel
- Division of Respiratory Medicine, Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Nottingham, NG5 1PB, United Kingdom
| | - Rachel L Clifford
- Division of Respiratory Medicine, Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Nottingham, NG5 1PB, United Kingdom
| | - Alan J Knox
- Division of Respiratory Medicine, Nottingham Respiratory Research Unit, University of Nottingham, Clinical Sciences Building, City Hospital, Nottingham, NG5 1PB, United Kingdom.
| |
Collapse
|
20
|
Bonfield TL. Macrophage Dysfunction in Cystic Fibrosis: A Therapeutic Target to Enhance Self-Immunity. Am J Respir Crit Care Med 2016; 192:1406-7. [PMID: 26669468 DOI: 10.1164/rccm.201509-1811ed] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Affiliation(s)
- Tracey L Bonfield
- 1 Department of Pediatrics Case Western Reserve University Cleveland, Ohio
| |
Collapse
|
21
|
Abstract
Cystic fibrosis (CF) lung disease is characterized by persistent and unresolved inflammation, with elevated proinflammatory and decreased anti-inflammatory cytokines, and greater numbers of immune cells. Hyperinflammation is recognized as a leading cause of lung tissue destruction in CF. Hyper-inflammation is not solely observed in the lungs of CF patients, since it may contribute to destruction of exocrine pancreas and, likely, to defects in gastrointestinal tract tissue integrity. Paradoxically, despite the robust inflammatory response, and elevated number of immune cells (such as neutrophils and macrophages), CF lungs fail to clear bacteria and are more susceptible to infections. Here, we have summarized the current understanding of immune dysregulation in CF, which may drive hyperinflammation and impaired host defense.
Collapse
Affiliation(s)
- Emanuela M Bruscia
- Section of Respiratory Medicine, Department of Pediatrics, Yale University School of Medicine, 330 Cedar Street, FMP, Room#524, New Haven, CT 06520, USA.
| | - Tracey L Bonfield
- Division of Pulmonology, Allergy and Immunology, Department of Pediatrics, Case Western Reserve University School of Medicine, 0900 Euclid Avenue, Cleveland, OH 44106-4948, USA.
| |
Collapse
|
22
|
Nichols DP, Chmiel JF. Inflammation and its genesis in cystic fibrosis. Pediatr Pulmonol 2015; 50 Suppl 40:S39-56. [PMID: 26335954 DOI: 10.1002/ppul.23242] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/02/2015] [Revised: 06/07/2015] [Accepted: 06/16/2015] [Indexed: 12/17/2022]
Abstract
The host inflammatory response in cystic fibrosis (CF) lung disease has long been recognized as a central pathological feature and an important therapeutic target. Indeed, many believe that bronchiectasis results largely from the oxidative and proteolytic damage comprised within an exuberant airway inflammatory response that is dominated by neutrophils. In this review, we address the longstanding argument of whether or not the inflammatory response is directly attributable to impairment of the cystic fibrosis transmembrane conductance regulator or only secondary to airway obstruction and chronic bacterial infection and challenge the importance of this distinction in the context of therapy. We also review the centrality of neutrophils in CF lung pathophysiology and highlight more recent data that suggest the importance of other cell types and signaling beyond NF-κB activation. We discuss how protease and redox imbalance are critical factors in CF airway inflammation and end by reviewing some of the more promising therapeutic approaches now under development.
Collapse
Affiliation(s)
- David P Nichols
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, Colorado.,Department of Medicine, University of Colorado School of Medicine, Aurora, Colorado.,National Jewish Health, Denver, Colorado
| | - James F Chmiel
- Department of Pediatrics, Case Western Reserve University School of Medicine, Rainbow Babies and Children's Hospital, Cleveland, Ohio
| |
Collapse
|
23
|
Jundi K, Greene CM. Transcription of Interleukin-8: How Altered Regulation Can Affect Cystic Fibrosis Lung Disease. Biomolecules 2015; 5:1386-98. [PMID: 26140537 PMCID: PMC4598756 DOI: 10.3390/biom5031386] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2015] [Revised: 06/19/2015] [Accepted: 06/20/2015] [Indexed: 02/07/2023] Open
Abstract
Interleukin-8 (IL-8) is a neutrophil chemokine that is encoded on the CXCL8 gene. Normally CXCL8 expression is repressed due to histone deacetylation, octamer-1 binding to the promoter and the inhibitory effect of nuclear factor-κB repressing factor (NRF). However, in response to a suitable stimulus, the human CXCL8 gene undergoes transcription due to its inducible promoter that is regulated by the transcription factors nuclear factor-κB (NF-κB), activating protein (AP-1), CAAT/enhancer-binding protein β (C/EBPβ, also known as NF-IL-6), C/EBP homologous protein (CHOP) and cAMP response element binding protein (CREB). CXCL8 mRNA is then stabilised by the activity of p38 mitogen-activated protein kinase (p38 MAPK). Cystic fibrosis (CF) lung disease is characterised by a neutrophil-dominated airway inflammatory response. A major factor contributing to the large number of neutrophils is the higher than normal levels of IL-8 that are present within the CF lung. Infection and inflammation, together with intrinsic alterations in CF airway cells are responsible for the abnormally high intrapulmonary levels of IL-8. Strategies to inhibit aberrantly high CXCL8 expression hold therapeutic potential for CF lung disease.
Collapse
Affiliation(s)
- Karim Jundi
- Department of Medicine, Respiratory Research Division, Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland.
| | - Catherine M Greene
- Department of Medicine, Respiratory Research Division, Royal College of Surgeons in Ireland Education and Research Centre, Beaumont Hospital, Dublin 9, Ireland.
| |
Collapse
|
24
|
Afanas'ev I. Mechanisms of superoxide signaling in epigenetic processes: relation to aging and cancer. Aging Dis 2015; 6:216-27. [PMID: 26029480 DOI: 10.14336/ad.2014.0924] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/24/2014] [Indexed: 01/09/2023] Open
Abstract
Superoxide is a precursor of many free radicals and reactive oxygen species (ROS) in biological systems. It has been shown that superoxide regulates major epigenetic processes of DNA methylation, histone methylation, and histone acetylation. We suggested that superoxide, being a radical anion and a strong nucleophile, could participate in DNA methylation and histone methylation and acetylation through mechanism of nucleophilic substitution and free radical abstraction. In nucleophilic reactions superoxide is able to neutralize positive charges of methyl donors S-adenosyl-L-methionine (SAM) and acetyl-coenzyme A (AcCoA) enhancing their nucleophilic capacity or to deprotonate cytosine. In the reversed free radical reactions of demethylation and deacetylation superoxide is formed catalytically by the (Tet) family of dioxygenates and converted into the iron form of hydroxyl radical with subsequent oxidation and final eradication of methyl substituents. Double role of superoxide in these epigenetic processes might be of importance for understanding of ROS effects under physiological and pathological conditions including cancer and aging.
Collapse
Affiliation(s)
- Igor Afanas'ev
- Vitamin Research Institute, Moscow, Russia, Porto, Portugal
| |
Collapse
|
25
|
Clifford RL, Patel JK, John AE, Tatler AL, Mazengarb L, Brightling CE, Knox AJ. CXCL8 histone H3 acetylation is dysfunctional in airway smooth muscle in asthma: regulation by BET. Am J Physiol Lung Cell Mol Physiol 2015; 308:L962-72. [PMID: 25713319 PMCID: PMC4421784 DOI: 10.1152/ajplung.00021.2015] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Accepted: 02/13/2015] [Indexed: 01/03/2023] Open
Abstract
Asthma is characterized by airway inflammation and remodeling and CXCL8 is a CXC chemokine that drives steroid-resistant neutrophilic airway inflammation. We have shown that airway smooth muscle (ASM) cells isolated from asthmatic individuals secrete more CXCL8 than cells from nonasthmatic individuals. Here we investigated chromatin modifications at the CXCL8 promoter in ASM cells from nonasthmatic and asthmatic donors to further understand how CXCL8 is dysregulated in asthma. ASM cells from asthmatic donors had increased histone H3 acetylation, specifically histone H3K18 acetylation, and increased binding of histone acetyltransferase p300 compared with nonasthmatic donors but no differences in CXCL8 DNA methylation. The acetylation reader proteins Brd3 and Brd4 were bound to the CXCL8 promoter and Brd inhibitors inhibited CXCL8 secretion from ASM cells by disrupting Brd4 and RNA polymerase II binding to the CXCL8 promoter. Our results show a novel dysregulation of CXCL8 transcriptional regulation in asthma characterized by a promoter complex that is abnormal in ASM cells isolated from asthmatic donors and can be modulated by Brd inhibitors. Brd inhibitors may provide a new therapeutic strategy for steroid-resistant inflammation.
Collapse
Affiliation(s)
- Rachel L Clifford
- Department of Respiratory Medicine and Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom; and
| | - Jamie K Patel
- Department of Respiratory Medicine and Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom; and
| | - Alison E John
- Department of Respiratory Medicine and Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom; and
| | - Amanda L Tatler
- Department of Respiratory Medicine and Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom; and
| | - Lisa Mazengarb
- Department of Respiratory Medicine and Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom; and
| | - Christopher E Brightling
- Institute for Lung Health, Department of Infection, Immunity and Inflammation, University of Leicester, Leicester, United Kingdom
| | - Alan J Knox
- Department of Respiratory Medicine and Nottingham Respiratory Research Unit, University of Nottingham, Nottingham, United Kingdom; and
| |
Collapse
|
26
|
Bartling TR, Subbaram S, Clark RR, Chandrasekaran A, Kar S, Melendez JA. Redox-sensitive gene-regulatory events controlling aberrant matrix metalloproteinase-1 expression. Free Radic Biol Med 2014; 74:99-107. [PMID: 24973648 PMCID: PMC4146650 DOI: 10.1016/j.freeradbiomed.2014.06.017] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 05/27/2014] [Accepted: 06/19/2014] [Indexed: 12/20/2022]
Abstract
Aberrant matrix metalloproteinase-1 (MMP-1) expression contributes to the pathogenesis of many degenerative disease processes that are associated with increased oxidative damage or stress. We and others have established that shifts in steady-state H2O2 production resulting from enforced antioxidant gene expression, senescence, or UV irradiation control MMP-1 expression. Here we establish that histone deacetylase-2 (HDAC2) protein levels and its occupancy of the MMP-1 promoter are decreased in response to enforced manganese superoxide dismutase (Sod2) expression. Inhibition of HDAC activity further accentuates the redox-dependent expression of MMP-1. Sod2-dependent decreases in HDAC2 are associated with increases in a proteasome-sensitive pool of ubiquitinylated HDAC2 and MMP-1-specific histone H3 acetylation. Sod2 overexpression also enhanced recruitment of Ets-1, c-Jun, c-Fos, and the histone acetyltransferase PCAF to the distal and proximal regions of the MMP-1 promoter. Furthermore, the Sod2-dependent expression of MMP-1 can be reversed by silencing the transcriptional activator c-Jun. All of the above Sod2-dependent alterations are largely reversed by catalase coexpression, indicating that the redox control of MMP-1 is H2O2-dependent. These findings identify a novel redox regulation of MMP-1 transcription that involves site-specific promoter recruitment of both activating factors and chromatin-modifying enzymes, which converge to maximally drive MMP-1 gene expression.
Collapse
Affiliation(s)
- Toni R Bartling
- College of Nanoscale Science and Engineering, State University of New York, Albany, NY 12203, USA
| | - Sita Subbaram
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | - Ryan R Clark
- College of Nanoscale Science and Engineering, State University of New York, Albany, NY 12203, USA
| | - Akshaya Chandrasekaran
- College of Nanoscale Science and Engineering, State University of New York, Albany, NY 12203, USA
| | - Supriya Kar
- Pediatrics, Albany Medical College, Albany, NY 12208, USA
| | - J Andres Melendez
- College of Nanoscale Science and Engineering, State University of New York, Albany, NY 12203, USA.
| |
Collapse
|
27
|
Disruption of interleukin-1β autocrine signaling rescues complex I activity and improves ROS levels in immortalized epithelial cells with impaired cystic fibrosis transmembrane conductance regulator (CFTR) function. PLoS One 2014; 9:e99257. [PMID: 24901709 PMCID: PMC4047112 DOI: 10.1371/journal.pone.0099257] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2013] [Accepted: 05/13/2014] [Indexed: 12/22/2022] Open
Abstract
Patients with cystic fibrosis (CF) have elevated concentration of cytokines in sputum and a general inflammatory condition. In addition, CF cells in culture produce diverse cytokines in excess, including IL-1β. We have previously shown that IL-1β, at low doses (∼30 pM), can stimulate the expression of CFTR in T84 colon carcinoma cells, through NF-κB signaling. However, at higher doses (>2.5 ng/ml, ∼150 pM), IL-1β inhibit CFTR mRNA expression. On the other hand, by using differential display, we found two genes with reduced expression in CF cells, corresponding to the mitochondrial proteins CISD1 and MTND4. The last is a key subunit for the activity of mitochondrial Complex I (mCx-I); accordingly, we later found a reduced mCx-I activity in CF cells. Here we found that IB3-1 cells (CF cells), cultured in serum-free media, secrete 323±5 pg/ml of IL-1β in 24 h vs 127±3 pg/ml for S9 cells (CFTR-corrected IB3-1 cells). Externally added IL-1β (5 ng/ml) reduces the mCx-I activity and increases the mitochondrial (MitoSOX probe) and cellular (DCFH-DA probe) ROS levels of S9 (CFTR-corrected IB3-1 CF cells) or Caco-2/pRSctrl cells (shRNA control cells) to values comparable to those of IB3-1 or Caco-2/pRS26 cells (shRNA specific for CFTR). Treatments of IB3-1 or Caco-2/pRS26 cells with either IL-1β blocking antibody, IL-1 receptor antagonist, IKK inhibitor III (NF-κB pathway) or SB203580 (p38 MAPK pathway), restored the mCx-I activity. In addition, in IB3-1 or Caco-2/pRS26 cells, IL-1β blocking antibody, IKK inhibitor III or SB203580 reduced the mitochondrial ROS levels by ∼50% and the cellular ROS levels near to basal values. The AP-1 inhibitors U0126 (MEK1/2) or SP600125 (JNK1/2/3 inhibitor) had no effects. The results suggest that in these cells IL-1β, through an autocrine effect, acts as a bridge connecting the CFTR with the mCx-I activity and the ROS levels.
Collapse
|
28
|
Umunakwe OC, Seegmiller AC. Abnormal n-6 fatty acid metabolism in cystic fibrosis is caused by activation of AMP-activated protein kinase. J Lipid Res 2014; 55:1489-97. [PMID: 24859760 DOI: 10.1194/jlr.m050369] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Indexed: 12/14/2022] Open
Abstract
Cystic fibrosis (CF) patients and model systems exhibit consistent abnormalities in PUFA metabolism, including increased metabolism of linoleate to arachidonate. Recent studies have connected these abnormalities to increased expression and activity of the Δ6- and Δ5-desaturase enzymes. However, the mechanism connecting these changes to the CF transmembrane conductance regulator (CFTR) mutations responsible for CF is unknown. This study tests the hypothesis that increased activity of AMP-activated protein kinase (AMPK), previously described in CF bronchial epithelial cells, causes these changes in fatty acid metabolism by driving desaturase expression. Using CF bronchial epithelial cell culture models, we confirm elevated activity of AMPK in CF cells and show that it is due to increased phosphorylation of AMPK by Ca(2+)/calmodulin-dependent protein kinase kinase β (CaMKKβ). We also show that inhibition of AMPK or CaMKKβ reduces desaturase expression and reverses the metabolic alterations seen in CF cells. These results signify a novel AMPK-dependent mechanism linking the genetic defect in CF to alterations in PUFA metabolism.
Collapse
Affiliation(s)
- Obi C Umunakwe
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| | - Adam C Seegmiller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, TN
| |
Collapse
|
29
|
Khan YM, Kirkham P, Barnes PJ, Adcock IM. Brd4 is essential for IL-1β-induced inflammation in human airway epithelial cells. PLoS One 2014; 9:e95051. [PMID: 24759736 PMCID: PMC3997389 DOI: 10.1371/journal.pone.0095051] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2013] [Accepted: 03/23/2014] [Indexed: 11/20/2022] Open
Abstract
BACKGROUND Chronic inflammation and oxidative stress are key features of chronic obstructive pulmonary disease (COPD). Oxidative stress enhances COPD inflammation under the control of the pro-inflammatory redox-sensitive transcription factor nuclear factor-kappaB (NF-κB). Histone acetylation plays a critical role in chronic inflammation and bromodomain and extra terminal (BET) proteins act as "readers" of acetylated histones. Therefore, we examined the role of BET proteins in particular Brd2 and Brd4 and their inhibitors (JQ1 and PFI-1) in oxidative stress- enhanced inflammation in human bronchial epithelial cells. METHODS Human primary epithelial (NHBE) cells and BEAS-2B cell lines were stimulated with IL-1β (inflammatory stimulus) in the presence or absence of H2O2 (oxidative stress) and the effect of pre-treatment with bromodomain inhibitors (JQ1 and PFI-1) was investigated. Pro-inflammatory mediators (CXCL8 and IL-6) were measured by ELISA and transcripts by RT-PCR. H3 and H4 acetylation and recruitment of p65 and Brd4 to the native IL-8 and IL-6 promoters was investigated using chromatin immunoprecipitation (ChIP). The impact of Brd2 and Brd4 siRNA knockdown on inflammatory mediators was also investigated. RESULT H2O2 enhanced IL1β-induced IL-6 and CXCL8 expression in NHBE and BEAS-2B cells whereas H2O2 alone did not have any affect. H3 acetylation at the IL-6 and IL-8 promoters was associated with recruitment of p65 and Brd4 proteins. Although p65 acetylation was increased this was not directly targeted by Brd4. The BET inhibitors JQ1 and PFI-1 significantly reduced IL-6 and CXCL8 expression whereas no effect was seen with the inactive enantiomer JQ1(-). Brd4, but not Brd2, knockdown markedly reduced IL-6 and CXCL8 release. JQ1 also inhibited p65 and Brd4 recruitment to the IL-6 and IL-8 promoters. CONCLUSION Oxidative stress enhanced IL1β-induced IL-6 and CXCL8 expression was significantly reduced by Brd4 inhibition. Brd4 plays an important role in the regulation of inflammatory genes and provides a potential novel anti-inflammatory target.
Collapse
Affiliation(s)
- Younis M. Khan
- Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Paul Kirkham
- School of Applied Sciences, University of Wolverhampton, Wolverhampton, United Kingdom
| | - Peter J. Barnes
- Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| | - Ian M. Adcock
- Airways Disease Section, National Heart & Lung Institute, Imperial College London, London, United Kingdom
| |
Collapse
|
30
|
Afanas’ev I. New nucleophilic mechanisms of ros-dependent epigenetic modifications: comparison of aging and cancer. Aging Dis 2014; 5:52-62. [PMID: 24490117 PMCID: PMC3901614 DOI: 10.14336/ad.2014.050052] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2013] [Revised: 10/07/2013] [Accepted: 10/16/2013] [Indexed: 12/31/2022] Open
Abstract
It has been shown that ROS (reactive oxygen species, superoxide and hydrogen peroxide) regulate major epigenetic processes, DNA methylation and histone acetylation, although the mechanism of ROS action (ROS signaling) is still unknown. Both DNA methylation and histone acetylation are nucleophilic processes and therefore ROS signaling through typical free radical processes, for example hydrogen atom abstraction is impossible. However, being "super-nucleophile" superoxide can participate in these reactions. Now we propose new nucleophilic mechanisms of DNA methylation and histone modification. During DNA methylation superoxide can deprotonate the cytosine molecule at C-5 position and by this accelerate the reaction of DNA with the positive-charged intermediate S-adenosyl-L-methionine (SAM). Superoxide can also deprotonate histone N-terminal tail lysines and accelerate the formation of their complexes with acetyl-coenzyme A (AcCoA), the supplier of acetyl groups. In cancer cells ROS enhance DNA methylation causing the silencing of tumor suppressor and antioxidant genes and enhancing the proliferation of cancer cells under condition of oxidative stress. ROS signaling in senescent cells probably causes DNA hypomethylation although there are insufficient data for such proposal.
Collapse
Affiliation(s)
- Igor Afanas’ev
- Correspondence should be addressed to: Dr. Igor Afanas’ev, Rua Vitorino Nemesio 48, 6.1, 2050-638, Porto, Portugal.
| |
Collapse
|
31
|
Nod-like receptor X-1 is required for rhinovirus-induced barrier dysfunction in airway epithelial cells. J Virol 2014; 88:3705-18. [PMID: 24429360 DOI: 10.1128/jvi.03039-13] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
UNLABELLED Barrier dysfunction of airway epithelium may increase the risk for acquiring secondary infections or allergen sensitization. Both rhinovirus (RV) and polyinosinic-polycytidilic acid [poly(I·C)], a double-stranded RNA (dsRNA) mimetic, cause airway epithelial barrier dysfunction, which is reactive oxygen species (ROS) dependent, implying that dsRNA generated during RV replication is sufficient for disrupting barrier function. We also demonstrated that RV or poly(I·C)-stimulated NADPH oxidase 1 (NOX-1) partially accounts for RV-induced ROS generation. In this study, we identified a dsRNA receptor(s) contributing to RV-induced maximal ROS generation and thus barrier disruption. We demonstrate that genetic silencing of the newly discovered dsRNA receptor Nod-like receptor X-1 (NLRX-1), but not other previously described dsRNA receptors, abrogated RV-induced ROS generation and reduction of transepithelial resistance (R(T)) in polarized airway epithelial cells. In addition, both RV and poly(I·C) stimulated mitochondrial ROS, the generation of which was dependent on NLRX-1. Treatment with Mito-Tempo, an antioxidant targeted to mitochondria, abolished RV-induced mitochondrial ROS generation, reduction in R(T), and bacterial transmigration. Furthermore, RV infection increased NLRX-1 localization to the mitochondria. Additionally, NLRX-1 interacts with RV RNA and poly(I·C) in polarized airway epithelial cells. Finally, we show that NLRX-1 is also required for RV-stimulated NOX-1 expression. These findings suggest a novel mechanism by which RV stimulates generation of ROS, which is required for disruption of airway epithelial barrier function. IMPORTANCE Rhinovirus (RV), a virus responsible for a majority of common colds, disrupts the barrier function of the airway epithelium by increasing reactive oxygen species (ROS). Poly(I·C), a double-stranded RNA (dsRNA) mimetic, also causes ROS-dependent barrier disruption, implying that the dsRNA intermediate generated during RV replication is sufficient for this process. Here, we demonstrate that both RV RNA and poly(I·C) interact with NLRX-1 (a newly discovered dsRNA receptor) and stimulate mitochondrial ROS. We show for the first time that NLRX-1 is primarily expressed in the cytoplasm and at the apical surface rather than in the mitochondria and that NLRX-1 translocates to mitochondria following RV infection. Together, our results suggest a novel mechanism for RV-induced barrier disruption involving NLRX-1 and mitochondrial ROS. Although ROS is necessary for optimal viral clearance, if not neutralized efficiently, it may increase susceptibility to secondary infections and alter innate immune responses to subsequently inhaled pathogens, allergens, and other environmental factors.
Collapse
|
32
|
Zhang PX, Murray TS, Villella VR, Ferrari E, Esposito S, D'Souza A, Raia V, Maiuri L, Krause DS, Egan ME, Bruscia EM. Reduced caveolin-1 promotes hyperinflammation due to abnormal heme oxygenase-1 localization in lipopolysaccharide-challenged macrophages with dysfunctional cystic fibrosis transmembrane conductance regulator. THE JOURNAL OF IMMUNOLOGY 2013; 190:5196-206. [PMID: 23606537 DOI: 10.4049/jimmunol.1201607] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
We have previously reported that TLR4 signaling is increased in LPS-stimulated cystic fibrosis (CF) macrophages (MΦs), contributing to the robust production of proinflammatory cytokines. The heme oxygenase-1 (HO-1)/CO pathway modulates cellular redox status, inflammatory responses, and cell survival. The HO-1 enzyme, together with the scaffold protein caveolin 1 (CAV-1), also acts as a negative regulator of TLR4 signaling in MΦs. In this study, we demonstrate that in LPS-challenged CF MΦs, HO-1 does not compartmentalize normally to the cell surface and instead accumulates intracellularly. The abnormal HO-1 localization in CF MΦs in response to LPS is due to decreased CAV-1 expression, which is controlled by the cellular oxidative state, and is required for HO-1 delivery to the cell surface. Overexpression of HO-1 or stimulating the pathway with CO-releasing molecules enhances CAV-1 expression in CF MΦs, suggesting a positive-feed forward loop between HO-1/CO induction and CAV-1 expression. These manipulations re-established HO-1 and CAV-1 cell surface localization in CF MΦs. Consistent with restoration of HO-1/CAV-1-negative regulation of TLR4 signaling, genetic or pharmacological (CO-releasing molecule 2) induced enhancement of this pathway decreased the inflammatory response of CF MΦs and CF mice treated with LPS. In conclusion, our results demonstrate that the counterregulatory HO-1/CO pathway, which is critical in balancing and limiting the inflammatory response, is defective in CF MΦs through a CAV-1-dependent mechanism, exacerbating the CF MΦ response to LPS. This pathway could be a potential target for therapeutic intervention for CF lung disease.
Collapse
Affiliation(s)
- Ping-Xia Zhang
- Department of Pediatrics, Yale University School of Medicine, New Haven, CT 06509, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Valdivieso AG, Santa-Coloma TA. CFTR activity and mitochondrial function. Redox Biol 2013; 1:190-202. [PMID: 24024153 PMCID: PMC3757715 DOI: 10.1016/j.redox.2012.11.007] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2012] [Accepted: 11/12/2012] [Indexed: 12/21/2022] Open
Abstract
Cystic Fibrosis (CF) is a frequent and lethal autosomal recessive disease, caused by mutations in the gene encoding the Cystic Fibrosis Transmembrane Conductance Regulator (CFTR). Before the discovery of the CFTR gene, several hypotheses attempted to explain the etiology of this disease, including the possible role of a chloride channel, diverse alterations in mitochondrial functions, the overexpression of the lysosomal enzyme α-glucosidase and a deficiency in the cytosolic enzyme glucose 6-phosphate dehydrogenase. Because of the diverse mitochondrial changes found, some authors proposed that the affected gene should codify for a mitochondrial protein. Later, the CFTR cloning and the demonstration of its chloride channel activity turned the mitochondrial, lysosomal and cytosolic hypotheses obsolete. However, in recent years, using new approaches, several investigators reported similar or new alterations of mitochondrial functions in Cystic Fibrosis, thus rediscovering a possible role of mitochondria in this disease. Here, we review these CFTR-driven mitochondrial defects, including differential gene expression, alterations in oxidative phosphorylation, calcium homeostasis, oxidative stress, apoptosis and innate immune response, which might explain some characteristics of the complex CF phenotype and reveals potential new targets for therapy.
Collapse
Affiliation(s)
- Angel Gabriel Valdivieso
- Institute for Biomedical Research (BIOMED CONICET-UCA), Laboratory of Cellular and Molecular Biology, School of Medical Sciences, Pontifical Catholic University of Argentina (UCA), Buenos Aires, Argentina
| | | |
Collapse
|
34
|
Ghio AJ, Roggli VL, Soukup JM, Richards JH, Randell SH, Muhlebach MS. Iron accumulates in the lavage and explanted lungs of cystic fibrosis patients. J Cyst Fibros 2012. [PMID: 23176785 DOI: 10.1016/j.jcf.2012.10.010] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Oxidative stress participates in the pathophysiology of cystic fibrosis (CF). An underlying disruption in iron homeostasis can frequently be demonstrated in injuries and diseases associated with an oxidative stress. We tested the hypothesis that iron accumulation and altered expression of iron-related proteins could be demonstrated in both the bronchoalveolar lavage (BAL) fluid and explanted lungs of patients with cystic fibrosis. BAL fluid collected from 10 children with CF showed elevated concentrations of protein, iron, ferritin, transferrin, heme, and hemoglobin relative to that obtained from 20 healthy volunteers. Using Perl's Prussian blue staining, explanted lung from CF patients revealed increased iron in alveolar and interstitial macrophages. Similarly, there was an increased expression of ferritin, the iron importer DMT1, and the exporter ferroportin 1 in lung tissue from CF patients. We conclude that iron homeostasis is disrupted in CF patients with an accumulation of this metal and altered expression of iron-related proteins being evident in the lungs.
Collapse
Affiliation(s)
- Andrew J Ghio
- National Health and Environmental Effects Research Laboratory, US EPA, Chapel Hill, NC 27599-7315, USA.
| | | | | | | | | | | |
Collapse
|
35
|
Ion channels/transporters as epigenetic regulators? -a microRNA perspective. SCIENCE CHINA-LIFE SCIENCES 2012; 55:753-60. [PMID: 23015123 DOI: 10.1007/s11427-012-4369-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/20/2012] [Accepted: 07/30/2012] [Indexed: 10/27/2022]
Abstract
MicroRNA (miRNA) alterations in response to changes in an extracellular microenvironment have been observed and considered as one of the major mechanisms for epigenetic modifications of the cell. While enormous efforts have been made in the understanding of the role of miRNAs in regulating cellular responses to the microenvironment, the mechanistic insight into how extracellular signals can be transduced into miRNA alterations in cells is still lacking. Interestingly, recent studies have shown that ion channels/transporters, which are known to conduct or transport ions across the cell membrane, also exhibit changes in levels of expression and activities in response to changes of extracellular microenvironment. More importantly, alterations in expression and function of ion channels/transporters have been shown to result in changes in miRNAs that are known to change in response to alteration of the microenvironment. In this review, we aim to summarize the recent data demonstrating the ability of ion channels/transporters to transduce extracellular signals into miRNA changes and propose a potential link between cells and their microenvironment through ion channels/transporters. At the same time, we hope to provide new insights into epigenetic regulatory mechanisms underlying a number of physiological and pathological processes, including embryo development and cancer metastasis.
Collapse
|
36
|
Sato A, Xu Y, Whitsett JA, Ikegami M. CCAAT/enhancer binding protein-α regulates the protease/antiprotease balance required for bronchiolar epithelium regeneration. Am J Respir Cell Mol Biol 2012; 47:454-63. [PMID: 22652201 DOI: 10.1165/rcmb.2011-0239oc] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Many transcription factors that regulate lung morphogenesis during development are reactivated to mediate repairs of the injured adult lung. We hypothesized that CCAAT/enhancer binding protein-α (C/EBPα), a transcription factor critical for perinatal lung maturation, regulates genes required for the normal repair of the bronchiolar epithelium after injury. Transgenic Cebpα(Δ/Δ) mice, in which Cebpa was conditionally deleted from Clara cells and Type II cells after birth, were used in this study. Airway injury was induced in mice by the intraperitoneal administration of naphthalene to ablate bronchiolar epithelial cells. Although the deletion of C/EBPα did not influence lung structure and function under unstressed conditions, C/EBPα was required for the normal repair of terminal bronchiolar epithelium after naphthalene injury. To identify cellular processes that are influenced by C/EBPα during repair, mRNA microarray was performed on terminal bronchiolar epithelial cells isolated by laser-capture microdissection. Normal repair of the terminal bronchiolar epithelium was highly associated with the mRNAs regulating antiprotease activities, and their induction required C/EBPα. The defective deposition of fibronectin in Cebpα(Δ/Δ) mice was associated with increased protease activity and delayed differentiation of FoxJ1-expressing ciliated cells. The fibronectin and ciliated cells were restored by the intratracheal treatment of Cebpα(Δ/Δ) mice with the serine protease inhibitor. In conclusion, C/EBPα regulates the expression of serine protease inhibitors that are required for the normal increase of fibronectin and the restoration of ciliated cells after injury. Treatment with serine protease inhibitor may aid in the recovery of injured bronchiolar epithelial cells, and prevent common chronic lung diseases.
Collapse
Affiliation(s)
- Atsuyasu Sato
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229-3039, USA
| | | | | | | |
Collapse
|
37
|
Abstract
Cystic fibrosis transmembrane conductance regulator (CFTR) functions as a channel that regulates the transport of ions and the movement of water across the epithelial barrier. Mutations in CFTR, which form the basis for the clinical manifestations of cystic fibrosis, affect the epithelial innate immune function in the lung, resulting in exaggerated and ineffective airway inflammation that fails to eradicate pulmonary pathogens. Compounding the effects of excessive neutrophil recruitment, the mutant CFTR channel does not transport antioxidants to counteract neutrophil-associated oxidative stress. Whereas mutant CFTR expression in leukocytes outside of the lung does not markedly impair their function, the expected regulation of inflammation in the airways is clearly deficient in cystic fibrosis. The resulting bacterial infections, which are caused by organisms that have substantial genetic and metabolic flexibility, can resist multiple classes of antibiotics and evade phagocytic clearance. The development of animal models that approximate the human pulmonary phenotypes-airway inflammation and spontaneous infection-may provide the much-needed tools to establish how CFTR regulates mucosal immunity and to test directly the effect of pharmacologic potentiation and correction of mutant CFTR function on bacterial clearance.
Collapse
|
38
|
Saadane A, Eastman J, Berger M, Bonfield TL. Parthenolide inhibits ERK and AP-1 which are dysregulated and contribute to excessive IL-8 expression and secretion in cystic fibrosis cells. JOURNAL OF INFLAMMATION-LONDON 2011; 8:26. [PMID: 21992677 PMCID: PMC3226551 DOI: 10.1186/1476-9255-8-26] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2010] [Accepted: 10/12/2011] [Indexed: 11/17/2022]
Abstract
Background Excessive secretion of IL-8 characterizes cystic fibrosis (CF). This has been attributed to excessive activation of epithelial cell I-κB Kinase and/or NFκB. Maximum IL-8 production requires 3 cooperative mechanisms: 1) release of the promoter from repression; 2) activation of transcription by NFκB and AP-1; 3) stabilization of mRNA by p38-MAPK. Little is known about regulation of IL-8 by MAPKs or AP-1 in CF. Methods We studied our hypothesis in vitro using 3-cellular models. Two of these models are transformed cell lines with defective versus normal cystic fibrosis transmembrane conductance regulator (CFTR) expression: an antisense/sense transfected cell line and the patient derived IB3-1/S9. In the third series of studies, we studied primary necropsy human tracheal epithelial cells treated with an inhibitor of CFTR function. All cell lines were pretreated with parthenolide and then stimulated with TNFα and/or IL-1β. Results In response to stimulation with TNFα and/or IL-1β, IL-8 production and mRNA expression was greater in CF-type cells than in non-CF controls. This was associated with enhanced phosphorylation of p38, ERK1/2 and JNK and increased activation of AP-1. Since we previously showed that parthenolide inhibits excessive IL-8 production by CF cells, we evaluated its effects on MAPK and AP-1 activation and showed that parthenolide inhibited ERK and AP-1 activation. Using a luciferase promoter assay, our studies showed that parthenolide decreased activation of the IL-8 promoter in CF cells stimulated with TNFα/IL-1β. Conclusions In addition to NFκB MAPKs ERK, JNK and p38 and the transcription factor AP-1 are also dysregulated in CF epithelial cells. Parthenolide inhibited both NFκB and MAPK/AP-1 pathways contributing to the inhibition of IL-8 production.
Collapse
Affiliation(s)
- Aicha Saadane
- Department of Pediatrics, Case Western Reserve University, 11100 Euclid Avenue, BRB-822 Cleveland Ohio 44106, OH 44106, USA
| | - Jean Eastman
- Department of Pediatrics, Case Western Reserve University, 11100 Euclid Avenue, BRB-822 Cleveland Ohio 44106, OH 44106, USA
| | - Melvin Berger
- Department of Pediatrics, Case Western Reserve University, 11100 Euclid Avenue, BRB-822 Cleveland Ohio 44106, OH 44106, USA
| | - Tracey L Bonfield
- Department of Pediatrics, Case Western Reserve University, 11100 Euclid Avenue, BRB-822 Cleveland Ohio 44106, OH 44106, USA
| |
Collapse
|
39
|
Hamdaoui N, Baudoin-Legros M, Kelly M, Aissat A, Moriceau S, Vieu DL, Colas J, Fritsch J, Edelman A, Planelles G. Resveratrol rescues cAMP-dependent anionic transport in the cystic fibrosis pancreatic cell line CFPAC1. Br J Pharmacol 2011; 163:876-86. [PMID: 21366549 DOI: 10.1111/j.1476-5381.2011.01289.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE The cystic fibrosis transmembrane conductance regulator (CFTR) is a cAMP-dependent chloride channel in the plasma membrane of epithelia whose mutation is the cause of the genetic disease cystic fibrosis (CF). The most frequent CFTR mutation is deletion of Phe(508) and this mutant protein (delF508CFTR) does not readily translocate to the plasma membrane and is rapidly degraded within the cell. We hypothesized that treating epithelial cells with resveratrol, a natural polyphenolic, phyto-ooestrogenic compound from grapes, could modulate both the expression and localization of CFTR. EXPERIMENTAL APPROACH Cells endogenously expressing CFTR (MDCK1 and CAPAN1 cells) or delF508CFTR (CFPAC1 and airway epithelial cells, deriving from human bronchial biopsies) were treated with resveratrol for 2 or 18 h. The effect of this treatment on CFTR and delF508CFTR expression and localization was evaluated using RT-PCR, Western blot and immunocytochemistry. Halide efflux was measured with a fluorescent dye and with halide-sensitive electrodes. Production of interleukin-8 by these cells was assayed by ELISA. KEY RESULTS Resveratrol treatment increased CFTR expression or maturation in immunoblotting experiments in MDCK1 cells or in CFPAC1 cells. Indirect immunofluorescence experiments showed a shift of delF508CFTR localization towards the (peri)-membrane area in CFPAC1 cells and in human airway epithelial cells. A cAMP-dependent increase in membrane permeability to halide was detected in resveratrol-treated CFPAC1 cells, and was inhibited by a selective inhibitor of CFTR. CONCLUSION AND IMPLICATIONS These results show that resveratrol modulated CFTR expression and localization and could rescue cAMP-dependent chloride transport in delF508CFTR cells.
Collapse
|
40
|
Kim J, Natarajan S, Vaickus LJ, Bouchard JC, Beal D, Cruikshank WW, Remick DG. Diesel exhaust particulates exacerbate asthma-like inflammation by increasing CXC chemokines. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 179:2730-9. [PMID: 21967814 DOI: 10.1016/j.ajpath.2011.08.008] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 04/23/2011] [Revised: 07/11/2011] [Accepted: 08/10/2011] [Indexed: 11/17/2022]
Abstract
Particulate matter heavily pollutes the urban atmosphere, and several studies show a link between increased ambient particulate air pollution and exacerbation of pre-existing pulmonary diseases, including asthma. We investigated how diesel exhaust particulates (DEPs) aggravate asthma-like pulmonary inflammation in a mouse model of asthma induced by a house dust extract (HDE) containing cockroach allergens and endotoxin. BALB/c mice were exposed to three pulmonary challenges via hypopharyngeal administration of an HDE collected from the home of an asthmatic child. One hour before each pulmonary challenge, mice were exposed to DEP or PBS. Pulmonary inflammation was assessed by histological features, oxidative stress, respiratory physiological features, inflammatory cell recruitment, and local CXC chemokine production. To prove the role of CXC chemokines in the augmented inflammation, CXC chemokine-specific antibodies were delivered to the lungs before DEP exposure. DEP exacerbated HDE-induced airway inflammation, with increased airway mucus production, oxidative stress, inflammatory cell infiltration, bronchoalveolar lavage concentrations of CXC chemokines, and airway hyperreactivity. Neutralization of airway keratinocyte-derived chemokine and macrophage inflammatory protein-2 significantly improves the respiratory function in addition to decreasing the infiltration of neutrophils and eosinophils. Blocking the chemokines also decreased airway mucus production. These results demonstrate that DEP exacerbates airway inflammation induced by allergen through increased pulmonary expression of the CXC chemokines (keratinocyte-derived chemokine and macrophage inflammatory protein-2).
Collapse
Affiliation(s)
- Jiyoun Kim
- Department of Pathology and Laboratory Medicine, Boston University School of Medicine, Boston, Massachusetts 02118, USA.
| | | | | | | | | | | | | |
Collapse
|
41
|
Pseudomonas aeruginosa suppresses interferon response to rhinovirus infection in cystic fibrosis but not in normal bronchial epithelial cells. Infect Immun 2011; 79:4131-45. [PMID: 21825067 DOI: 10.1128/iai.05120-11] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Despite increased morbidity associated with secondary respiratory viral infections in cystic fibrosis (CF) patients with chronic Pseudomonas aeruginosa infection, the underlying mechanisms are not well understood. Here, we investigated the effect of P. aeruginosa infection on the innate immune responses of bronchial epithelial cells to rhinovirus (RV) infection. CF cells sequentially infected with mucoid P. aeruginosa (MPA) and RV showed lower levels of interferons (IFNs) and higher viral loads than those of RV-infected cells. Unlike results for CF cells, normal bronchial epithelial cells coinfected with MPA/RV showed higher IFN expression than RV-infected cells. In both CF and normal cells, the RV-stimulated IFN response requires phosphorylation of Akt and interferon response factor 3 (IRF3). Preinfection with MPA inhibited RV-stimulated Akt phosphorylation and decreased IRF3 phosphorylation in CF cells but not in normal cells. Compared to normal, unstimulated CF cells or normal cells treated with CFTR inhibitor showed increased reactive oxygen species (ROS) production. Treatment of CF cells with antioxidants prior to MPA infection partially reversed the suppressive effect of MPA on the RV-stimulated IFN response. Together, these results suggest that MPA preinfection inhibits viral clearance by suppressing the antiviral response particularly in CF cells but not in normal cells. Further, increased oxidative stress in CF cells appears to modulate the innate immune responses to coinfection.
Collapse
|
42
|
Kelly-Aubert M, Trudel S, Fritsch J, Nguyen-Khoa T, Baudouin-Legros M, Moriceau S, Jeanson L, Djouadi F, Matar C, Conti M, Ollero M, Brouillard F, Edelman A. GSH monoethyl ester rescues mitochondrial defects in cystic fibrosis models. Hum Mol Genet 2011; 20:2745-59. [DOI: 10.1093/hmg/ddr173] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
43
|
|
44
|
Marwick JA, Adcock IM, Chung KF. Overcoming reduced glucocorticoid sensitivity in airway disease: molecular mechanisms and therapeutic approaches. Drugs 2010; 70:929-48. [PMID: 20481652 DOI: 10.2165/10898520-000000000-00000] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
There is a considerable and growing unmet medical need in respiratory disease concerning effective anti-inflammatory therapies for conditions such as severe asthma, chronic obstructive pulmonary disease and cystic fibrosis. These diseases share a predominant characteristic of an enhanced and uncontrolled inflammatory response in the lungs, which contributes to disease progression, hospitalization and mortality. These diseases are poorly controlled by current anti-inflammatory therapies including glucocorticoids, which are otherwise effective in many other inflammatory conditions or in milder disease such as asthma. The exact cause of this apparent impairment of glucocorticoid function remains largely unclear; however, recent studies have now implicated a number of possible mechanisms. Central among these is an elevation of the oxidant burden in the lungs and the resulting reduction in the activity of histone deacetylase (HDAC)-2. This contributes to both the enhancement of proinflammatory mediator expression and the impaired ability of the glucocorticoid receptor (GR)-alpha to repress proinflammatory gene expression. The oxidant-mediated reduction in HDAC-2 activity is, in part, a result of an elevation in the phosphoinositol 3-kinase (PI3K) delta/Akt signalling pathway. Blockade of the PI3Kdelta pathway restores glucocortiocoid function in both in vitro and in vivo models, and in primary cells from disease. In addition, inhibition of the PI3Kdelta and PI3Kgamma isoforms is anti-inflammatory in both innate and adaptive immune responses. Consequently, selective inhibition of this pathway may provide a therapeutic strategy both as a novel anti-inflammatory and in combination therapy with glucocorticoids to restore their function. However, a number of other oxidant-related and -unrelated mechanisms, including altered kinase signalling and expression of the dominant negative GRbeta, may also play a role in the development of glucocorticoid insensitivity. Further elucidation of these mechanisms and pathways will enable novel therapeutic targeting for alternative anti-inflammatory drugs or combination therapies providing restoration for the anti-inflammatory action of glucocorticoids.
Collapse
Affiliation(s)
- John A Marwick
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh Medical School, Edinburgh, UK.
| | | | | |
Collapse
|
45
|
Kelly M, Trudel S, Brouillard F, Bouillaud F, Colas J, Nguyen-Khoa T, Ollero M, Edelman A, Fritsch J. Cystic fibrosis transmembrane regulator inhibitors CFTR(inh)-172 and GlyH-101 target mitochondrial functions, independently of chloride channel inhibition. J Pharmacol Exp Ther 2010; 333:60-9. [PMID: 20051483 DOI: 10.1124/jpet.109.162032] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Two highly potent and selective cystic fibrosis (CF) transmembrane regulator (CFTR) inhibitors have been identified by high-throughput screening: the thiazolidinone CFTR(inh)-172 [3-[(3-trifluoromethyl)phenyl]-5-[(4-carboxyphenyl)methylene]- 2-thioxo-4-thiazolidinone] and the glycine hydrazide GlyH-101 [N-(2-naphthalenyl)-((3,5-dibromo-2,4-dihydroxyphenyl)methylene)glycine hydrazide]. Inhibition of the CFTR chloride channel by these compounds has been suggested to be of pharmacological interest in the treatment of secretory diarrheas and polycystic kidney disease. In addition, functional inhibition of CFTR by CFTR(inh)-172 has been proposed to be sufficient to mimic the CF inflammatory profile. In the present study, we investigated the effects of the two compounds on reactive oxygen species (ROS) production and mitochondrial membrane potential in several cell lines: the CFTR-deficient human lung epithelial IB3-1 (expressing the heterozygous F508del/W1282X mutation), the isogenic CFTR-corrected C38, and HeLa and A549 as non-CFTR-expressing controls. Both inhibitors were able to induce a rapid increase in ROS levels and depolarize mitochondria in the four cell types, suggesting that these effects are independent of CFTR inhibition. In HeLa cells, these events were associated with a decrease in the rate of oxygen consumption, with GlyH-101 demonstrating a higher potency than CFTR(inh)-172. The impact of CFTR inhibitors on inflammatory parameters was also tested in HeLa cells. CFTR(inh)-172, but not GlyH-101, induced nuclear translocation of nuclear factor-kappaB (NF-kappaB). CFTR(inh)-172 slightly decreased interleukin-8 secretion, whereas GlyH-101 induced a slight increase. These results support the conclusion that CFTR inhibitors may exert nonspecific effects regarding ROS production, mitochondrial failure, and activation of the NF-kappaB signaling pathway, independently of CFTR inhibition.
Collapse
Affiliation(s)
- Mairead Kelly
- Institut National de la Santé et de la Recherche Médicale, U845, Centre de Recherche Croissance and Signalization, 156 Rue de Vaugirard, Paris, France
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Current World Literature. Curr Opin Pulm Med 2009; 15:638-44. [DOI: 10.1097/mcp.0b013e3283328a80] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
|
47
|
Modulation of expression of IL-8 gene in bronchial epithelial cells by 5-methoxypsoralen. Int Immunopharmacol 2009; 9:1411-22. [PMID: 19720161 DOI: 10.1016/j.intimp.2009.08.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2009] [Revised: 07/29/2009] [Accepted: 08/19/2009] [Indexed: 12/13/2022]
Abstract
Persistent recruitment of neutrophils in the bronchi of cystic fibrosis patients contributes to airway tissue damage, suggesting the importance of intervening on the expression of the neutrophil chemokine IL-8. Extracts from plants have been investigated to select components able to reduce IL-8 expression in bronchial epithelial cells challenged with Pseudomonas aeruginosa. Extracts and purified components have been added to cells 24 h before pro-inflammatory challenge with P. aeruginosa and IL-8 transcription was quantified in the IB3-1 CF cells in vitro. P. aeruginosa-dependent IL-8 mRNA induction was increased by Argemone mexicana and Vernonia anthelmintica whereas no significant modification of transcription was observed with Aphanamixis polystachya, Lagerstroemia speciosa and Hemidesmus indicus. Finally, inhibition of IL-8 was observed with Polyalthia longifolia (IC50=200 microg/ml) and Aegle marmelos (IC50=20 microg/ml). Compounds from A. marmelos were isolated and identified by GC-MS. No significant effect was observed with butyl-p-tolyl sulphate, whereas the inhibition obtained with 6-methyl-4-chromanone concentration was accompanied by an anti-proliferative effect. On the contrary, 5-methoxypsoralen resulted in IL-8 inhibition at 10 microM concentration, without effects on cell proliferation. In synthesis, 5-methoxypsoralen can be taken into consideration to investigate mechanisms of neutrophil chemotactic signalling and for its potential application in modulating the excessive CF lung inflammation.
Collapse
|
48
|
Watson JA, Watson CJ, McCrohan AM, Woodfine K, Tosetto M, McDaid J, Gallagher E, Betts D, Baugh J, O'Sullivan J, Murrell A, Watson RWG, McCann A. Generation of an epigenetic signature by chronic hypoxia in prostate cells. Hum Mol Genet 2009; 18:3594-604. [PMID: 19584087 DOI: 10.1093/hmg/ddp307] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Increasing levels of tissue hypoxia have been reported as a natural feature of the aging prostate gland and may be a risk factor for the development of prostate cancer. In this study, we have used PwR-1E benign prostate epithelial cells and an equivalently aged hypoxia-adapted PwR-1E sub-line to identify phenotypic and epigenetic consequences of chronic hypoxia in prostate cells. We have identified a significantly altered cellular phenotype in response to chronic hypoxia as characterized by increased receptor-mediated apoptotic resistance, the induction of cellular senescence, increased invasion and the increased secretion of IL-1 beta, IL6, IL8 and TNFalpha cytokines. In association with these phenotypic changes and the absence of HIF-1 alpha protein expression, we have demonstrated significant increases in global levels of DNA methylation and H3K9 histone acetylation in these cells, concomitant with the increased expression of DNA methyltransferase DMNT3b and gene-specific changes in DNA methylation at key imprinting loci. In conclusion, we have demonstrated a genome-wide adjustment of DNA methylation and histone acetylation under chronic hypoxic conditions in the prostate. These epigenetic signatures may represent an additional mechanism to promote and maintain a hypoxic-adapted cellular phenotype with a potential role in tumour development.
Collapse
Affiliation(s)
- Jenny A Watson
- The UCD School of Medicine and Medical Science and The UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Lawless MW, O'Byrne KJ, Gray SG. Oxidative stress induced lung cancer and COPD: opportunities for epigenetic therapy. J Cell Mol Med 2009; 13:2800-21. [PMID: 19602054 PMCID: PMC4498937 DOI: 10.1111/j.1582-4934.2009.00845.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Reactive oxygen species (ROS) form as a natural by-product of the normal metabolism of oxygen and play important roles within the cell. Under normal circumstances the cell is able to maintain an adequate homeostasis between the formation of ROS and its removal through particular enzymatic pathways or via antioxidants. If however, this balance is disturbed a situation called oxidative stress occurs. Critically, oxidative stress plays important roles in the pathogenesis of many diseases, including cancer. Epigenetics is a process where gene expression is regulated by heritable mechanisms that do not cause any direct changes to the DNA sequence itself, and disruption of epigenetic mechanisms has important implications in disease. Evidence is emerging that histone deacetylases (HDACs) play decisive roles in regulating important cellular oxidative stress pathways including those involved with sensing oxidative stress and those involved with regulating the cellular response to oxidative stress. In particular aberrant regulation of these pathways by HDACs may play critical roles in cancer progression. In this review we discuss the current evidence linking epigenetics and oxidative stress and cancer, using chronic obstructive pulmonary disease and non-small cell lung cancer to illustrate the importance of epigenetics on these pathways within these disease settings.
Collapse
Affiliation(s)
- Matthew W Lawless
- Centre for Liver Disease, School of Medicine and Medical Science, Mater Misericordiae University Hospital, University College Dublin, Dublin, Ireland
| | | | | |
Collapse
|
50
|
Herfs M, Hubert P, Delvenne P. Epithelial metaplasia: adult stem cell reprogramming and (pre)neoplastic transformation mediated by inflammation? Trends Mol Med 2009; 15:245-53. [PMID: 19457719 DOI: 10.1016/j.molmed.2009.04.002] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2009] [Revised: 04/03/2009] [Accepted: 04/03/2009] [Indexed: 12/17/2022]
Abstract
Throughout adult life, new developmental commitment of adult stem cells causes metaplastic conversions to occur frequently in some organs. These reversible epithelial replacements are almost always observed in association with chronic inflammation and persistent irritation. Although metaplasia is not synonymous with dysplasia, clinical surveillance has demonstrated that these adaptive processes have an increased susceptibility to evolve into cancer. We propose that cytokines and other soluble factors released by both epithelial and inflammatory cells might alter the transcription-factor expression profile of stem cells and lead to the development of metaplasia. Furthermore, inflammatory mediators might also promote the malignant transformation of epithelial metaplasia by inducing genetic and epigenetic changes and by preventing the immune system from mounting an efficient anti-tumour immune response. A better understanding of the molecular mechanisms leading to metaplasia might help in the design of new therapies for neoplastic and degenerative diseases.
Collapse
Affiliation(s)
- Michael Herfs
- Laboratory of Experimental Pathology, GIGA-Cancer (Centre for Experimental Cancer Research), University of Liege, Liege, Belgium
| | | | | |
Collapse
|