1
|
Mirra D, Esposito R, Spaziano G, Rafaniello C, Panico F, Squillante A, Falciani M, Abrego-Guandique DM, Caiazzo E, Gallelli L, Cione E, D’Agostino B. miRNA Signatures in Alveolar Macrophages Related to Cigarette Smoke: Assessment and Bioinformatics Analysis. Int J Mol Sci 2025; 26:1277. [PMID: 39941045 PMCID: PMC11818525 DOI: 10.3390/ijms26031277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 01/23/2025] [Accepted: 01/30/2025] [Indexed: 02/16/2025] Open
Abstract
Cigarette smoke (CS) is a driver of many respiratory diseases, including chronic obstructive pulmonary disease (COPD) and non-small cell lung cancer (NSCLC). Tobacco causes oxidative stress, impaired phagocytosis of alveolar macrophages (AMs), and alterations in gene expression in the lungs of smokers. MicroRNAs (miRNAs) are small non-coding RNAs that influence several regulatory pathways. Previously, we monitored the expressions of hsa-miR-223-5p, 16-5p, 20a-5p, -17-5p, 34a-5p, and 106a-5p in AMs derived from the bronchoalveolar lavage (BAL) of subjects with NSCLC, COPD, and smoker and non-smoker control groups. Here, we investigated the capability of CS conditionate media to modulate the abovementioned miRNAs in primary AMs obtained in the same 43 sex-matched subjects. The expressions of has-miR-34a-5p, 17-5p, 16-5p, 106a-5p, 223-5p, and 20a-5p were assessed before and after in vitro CS exposure by RT-PCR. In addition, a comprehensive bioinformatic analysis of miRNAs KEGGS and PPI linked to inflammation was performed. Distinct and common miRNA expression profiles were identified in response to CS, suggesting their possible role in smoking-related diseases. It is worth noting that, following exposure to CS, the expression levels of hsa-miR-34a-5p and 17-5p in both smokers and non-smokers, 106a-5p in non-smokers, and 20a-5p in smokers, shifted towards those found in individuals with COPD, suggesting them as a risk factor in developing this lung condition. Moreover, CS-focused sub-analysis identified miRNA which exhibited CS-dependent pattern and modulated mRNA involved in the immune system or AMs property regulation. In conclusion, our study uncovered miRNA signatures in AMs exposed to CS, indicating that CS might modify epigenetic patterns that contribute to macrophage activation and lung disease onset and progression.
Collapse
Affiliation(s)
- Davida Mirra
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Renata Esposito
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Giuseppe Spaziano
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Concetta Rafaniello
- Campania Regional Centre for Pharmacovigilance and Pharmacoepidemiology, 80138 Naples, Italy;
- Section of Pharmacology “L. Donatelli”, Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy
| | - Francesca Panico
- Science of Health Department, School of Medicine, University of Catanzaro, 88100 Catanzaro, Italy; (F.P.); (D.M.A.-G.)
| | | | - Maddalena Falciani
- Pulmonary and Critical Care Medicine, Ospedale Scarlato, 84018 Scafati, Italy;
| | | | - Eleonora Caiazzo
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| | - Luca Gallelli
- Clinical Pharmacology and Pharmacovigilance Unit, Department of Health Sciences, Mater Domini Hospital, University of “Magna Graecia”, 88100 Catanzaro, Italy;
| | - Erika Cione
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, 87036 Rende, Italy;
| | - Bruno D’Agostino
- Department of Environmental Biological and Pharmaceutical Sciences and Technologies, University of Campania “Luigi Vanvitelli”, 81100 Caserta, Italy; (D.M.); (R.E.); (E.C.); (B.D.)
| |
Collapse
|
2
|
Listyoko AS, Okazaki R, Harada T, Inui G, Yamasaki A. Impact of obesity on airway remodeling in asthma: pathophysiological insights and clinical implications. FRONTIERS IN ALLERGY 2024; 5:1365801. [PMID: 38562155 PMCID: PMC10982419 DOI: 10.3389/falgy.2024.1365801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2024] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
The prevalence of obesity among asthma patients has surged in recent years, posing a significant risk factor for uncontrolled asthma. Beyond its impact on asthma severity and patients' quality of life, obesity is associated with reduced lung function, increased asthma exacerbations, hospitalizations, heightened airway hyperresponsiveness, and elevated asthma-related mortality. Obesity may lead to metabolic dysfunction and immune dysregulation, fostering chronic inflammation characterized by increased pro-inflammatory mediators and adipocytokines, elevated reactive oxygen species, and reduced antioxidant activity. This chronic inflammation holds the potential to induce airway remodeling in individuals with asthma and obesity. Airway remodeling encompasses structural and pathological changes, involving alterations in the airway's epithelial and subepithelial layers, hyperplasia and hypertrophy of airway smooth muscle, and changes in airway vascularity. In individuals with asthma and obesity, airway remodeling may underlie heightened airway hyperresponsiveness and increased asthma severity, ultimately contributing to the development of persistent airflow limitation, declining lung function, and a potential increase in asthma-related mortality. Despite efforts to address the impact of obesity on asthma outcomes, the intricate mechanisms linking obesity to asthma pathophysiology, particularly concerning airway remodeling, remain incompletely understood. This comprehensive review discusses current research investigating the influence of obesity on airway remodeling, to enhance our understanding of obesity's role in the context of asthma airway remodeling.
Collapse
Affiliation(s)
- Aditya Sri Listyoko
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
- Pulmonology and Respiratory Medicine Department, Faculty of Medicine, Brawijaya University-Dr. Saiful Anwar General Hospital, Malang, Indonesia
| | - Ryota Okazaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Tomoya Harada
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Genki Inui
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology, Department of Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Yonago, Japan
| |
Collapse
|
3
|
Pluangnooch P, Soontrapa K, Pudgerd A, Sridurongrit S. Expression of constitutively active TβRI leads to attenuation of ovalbumin-induced allergic airway inflammation associated with augmented M2 polarization of alveolar macrophage. Respir Investig 2024; 62:90-97. [PMID: 38007853 DOI: 10.1016/j.resinv.2023.10.005] [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: 06/17/2023] [Revised: 09/14/2023] [Accepted: 10/14/2023] [Indexed: 11/28/2023]
Abstract
BACKGROUND Transforming growth factor-β (Tgf-β) plays an important role in the pathogenesis of asthma through the regulation of T cells and airway epithelium. Its functions in alveolar macrophage (AM) during allergic airway inflammation remain unknown. METHODS A murine asthma model was induced with ovalbumin (ova) in TβRICA/Fsp1-Cre transgenic mice expressing constitutively active Tgf-β receptor type I (TβRICA) under the control of Fsp1-Cre transgene. Cells in the bronchoalveolar lavage (BAL) were collected to study immune cell infiltration in the lungs. Cytokine levels in BAL fluid were measured by enzyme-linked immunoassay (ELISA). Lungs were sectioned and stained with hematoxylin and eosin, periodic acid-Schiff, and trichrome for histopathologic evaluation. AMs were assessed by flow cytometry and were sorted for quantitative polymerase chain reaction analysis. RESULTS Our data indicated that TβRICA transcripts were induced in AMs of TβRICA/Fsp1-Cre mice. Following the ova challenges, TβRICA/Fsp1-Cre mice exhibited reduced cellular infiltration of the airway, reduced pulmonary fibrosis, and reduced bronchial mucus secretion as compared to ova-challenged wild-type mice. An alternatively activated macrophage (M2) polarization was significantly elevated in the lungs of ova-challenged TβRICA/Fsp1-Cre mice as reflected by increased numbers of AMs expressing M2 subtype marker, CD163, in the lungs and enhanced expression of CCR2 and CD206 in AMs. Moreover, TβRICA/Fsp1-Cre AMs showed augmented expression of transcription factors, Foxo1, and IRF4, which are known to be positive regulators for M2 polarization. CONCLUSIONS Expression of TβRICA in AMs promoted M2 polarization and ameliorated allergic airway inflammation in an ova-induced asthma mouse model.
Collapse
Affiliation(s)
- Panwadee Pluangnooch
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Kitipong Soontrapa
- Department of Pharmacology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Arnon Pudgerd
- Division of Anatomy, School of Medical Science, University of Phayao, Phayao 56000, Thailand
| | - Somyoth Sridurongrit
- Department of Anatomy, Faculty of Science, Mahidol University, Bangkok 10400, Thailand; Center of Excellence on Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok 10400, Thailand.
| |
Collapse
|
4
|
Song MA, Kim JY, Gorr MW, Miller RA, Karpurapu M, Nguyen J, Patel D, Archer KJ, Pabla N, Shields PG, Wold LE, Christman JW, Chung S. Sex-specific lung inflammation and mitochondrial damage in a model of electronic cigarette exposure in asthma. Am J Physiol Lung Cell Mol Physiol 2023; 325:L568-L579. [PMID: 37697923 PMCID: PMC11068405 DOI: 10.1152/ajplung.00033.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 07/14/2023] [Accepted: 08/30/2023] [Indexed: 09/13/2023] Open
Abstract
The prevalence of electronic cigarette (EC) use among adult with asthma has continued to increase over time, in part due to the belief of being less harmful than smoking. However, the extent of their toxicity and the involved mechanisms contributing to the deleterious impact of EC exposure on patients with preexisting asthma have not been delineated. In the present project, we tested the hypothesis that EC use contributes to respiratory damage and worsening inflammation in the lungs of patients with asthma. To define the consequences of EC exposure in established asthma, we used a mouse model with/without preexisting asthma for short-term exposure to EC aerosols. C57/BL6J mice were sensitized and challenged with a DRA (dust mite, ragweed, Aspergillus fumigates, 200 µg/mL) mixture and exposed daily to EC with nicotine (2% nicotine in 30:70 propylene glycol: vegetable glycerin) or filtered air for 2 wk. The mice were evaluated at 24 h after the final EC exposure. After EC exposure in asthmatic mice, lung inflammatory cell infiltration and goblet cell hyperplasia were increased, whereas EC alone did not cause airway inflammation. Our data also show that mitochondrial DNA (mtDNA) content and a key mtDNA regulator, mitochondrial transcription factor A (TFAM), are reduced in asthmatic EC-exposed mice in a sex-dependent manner. Together, these results indicate that TFAM loss in lung epithelium following EC contributes to male-predominant sex pathological differences, including mitochondrial damage, inflammation, and remodeling in asthmatic airways.NEW & NOTEWORTHY Respiratory immunity is dysregulated in preexisting asthma, and further perturbations by EC use could exacerbate asthma severity. However, the extent of their toxicity and the involved mechanisms contributing to the deleterious impact of EC exposure on patients with preexisting asthma have not been delineated. We found that EC has unique biological impacts in lungs and potential sex differences with loss of TFAM, a key mtDNA regulator, in lung epithelial region from our animal EC study.
Collapse
Affiliation(s)
- Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Ji Young Kim
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| | - Matthew W Gorr
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Roy A Miller
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Jackie Nguyen
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Devki Patel
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Kellie J Archer
- Division of Biostatistics, College of Public Health, The Ohio State University, Columbus, Ohio, United States
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, Ohio, United States
| | - Peter G Shields
- Comprehensive Cancer Center, The Ohio State University and James Cancer Hospital, Columbus, Ohio, United States
| | - Loren E Wold
- Division of Cardiac Surgery, Department of Surgery, College of Medicine, The Ohio State University, Columbus, Ohio, United States
| | - John W Christman
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care and Sleep Medicine, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, Ohio, United States
| |
Collapse
|
5
|
Zhang X, Evans TD, Chen S, Sergin I, Stitham J, Jeong SJ, Rodriguez-Velez A, Yeh YS, Park A, Jung IH, Diwan A, Schilling JD, Rom O, Yurdagul A, Epelman S, Cho J, Lodhi IJ, Mittendorfer B, Razani B. Loss of Macrophage mTORC2 Drives Atherosclerosis via FoxO1 and IL-1β Signaling. Circ Res 2023; 133:200-219. [PMID: 37350264 PMCID: PMC10527041 DOI: 10.1161/circresaha.122.321542] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 06/12/2023] [Indexed: 06/24/2023]
Abstract
BACKGROUND The mTOR (mechanistic target of rapamycin) pathway is a complex signaling cascade that regulates cellular growth, proliferation, metabolism, and survival. Although activation of mTOR signaling has been linked to atherosclerosis, its direct role in lesion progression and in plaque macrophages remains poorly understood. We previously demonstrated that mTORC1 (mTOR complex 1) activation promotes atherogenesis through inhibition of autophagy and increased apoptosis in macrophages. METHODS Using macrophage-specific Rictor- and mTOR-deficient mice, we now dissect the distinct functions of mTORC2 pathways in atherogenesis. RESULTS In contrast to the atheroprotective effect seen with blockade of macrophage mTORC1, macrophage-specific mTORC2-deficient mice exhibit an atherogenic phenotype, with larger, more complex lesions and increased cell death. In cultured macrophages, we show that mTORC2 signaling inhibits the FoxO1 (forkhead box protein O1) transcription factor, leading to suppression of proinflammatory pathways, especially the inflammasome/IL (interleukin)-1β response, a key mediator of vascular inflammation and atherosclerosis. In addition, administration of FoxO1 inhibitors efficiently rescued the proinflammatory response caused by mTORC2 deficiency both in vitro and in vivo. Interestingly, collective deletion of macrophage mTOR, which ablates mTORC1- and mTORC2-dependent pathways, leads to minimal change in plaque size or complexity, reflecting the balanced yet opposing roles of these signaling arms. CONCLUSIONS Our data provide the first mechanistic details of macrophage mTOR signaling in atherosclerosis and suggest that therapeutic measures aimed at modulating mTOR need to account for its dichotomous functions.
Collapse
Affiliation(s)
- Xiangyu Zhang
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Trent D. Evans
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Sunny Chen
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Ismail Sergin
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Jeremiah Stitham
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Se-Jin Jeong
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | | | - Yu-Sheng Yeh
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Arick Park
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - In-Hyuk Jung
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Abhinav Diwan
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| | - Joel D. Schilling
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
| | - Oren Rom
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Arif Yurdagul
- Department of Pathology and Translational Pathobiology and Department of Molecular and Cellular Physiology, Louisiana State University, Shreveport, LA
| | - Slava Epelman
- Ted Rogers Centre for Heart Research, Peter Munk Cardiac Center, Toronto General Hospital Research Institute, University Health Network and University of Toronto, Toronto, Canada
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
| | - Irfan J. Lodhi
- Division of Endocrinology, Metabolism, and Lipid Research, Washington University School of Medicine, St Louis, MO, USA
| | - Bettina Mittendorfer
- Division of Geriatrics and Nutritional Science, and Washington University School of Medicine, St Louis, MO, USA
| | - Babak Razani
- Department of Medicine and Vascular Medicine Institute, University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA
- Pittsburgh VA Medical Center, Pittsburgh, PA
- Cardiovascular Division, Washington University School of Medicine, St Louis, MO, USA
- Department of Pathology & Immunology, Washington University School of Medicine, St Louis, MO, USA
- John Cochran VA Medical Center, St. Louis, MO, USA
| |
Collapse
|
6
|
Liao W, Foo HYC, Tran TNQ, Chai CLL, Wong WSF. Calcaratarin D, a labdane diterpenoid, attenuates mouse asthma via modulating alveolar macrophage function. Br J Pharmacol 2023; 180:1056-1071. [PMID: 36440573 DOI: 10.1111/bph.15993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 10/04/2022] [Accepted: 11/16/2022] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Alveolar macrophages (AMs) contribute to airway inflammation and remodelling in allergic asthma. Calcaratarin D (CalD), a labdane diterpenoid from rhizomes of the medicinal plant Alpinia calcarata, has recently been shown to possess anti-inflammatory properties. The present study evaluated protective effects of CalD in a house dust mite (HDM)-induced asthma mouse model. EXPERIMENTAL APPROACH The effects of CalD on AMs in contributing to anti-inflammatory effects in asthma were investigated through in vivo, ex vivo, and in vitro experiments. KEY RESULTS CalD reduced total bronchoalveolar lavage fluid and differential cell count, serum IgE levels, mucus hypersecretion, and airway hyperresponsiveness in HDM-challenged mice. Additionally, CalD affected a wide array of pro-inflammatory cytokines and chemokines and oxidative damage markers in isolated lung tissues. CalD suppressed the HDM-induced increase in Arg1 (M2 macrophage marker) in AMs from lung tissue and reduced lung polyamine levels. CalD weakened antigen presentation capability of AMs by reducing CD80 expression, reduced AM-derived CCL17 and CCL22 levels, and lessened Th2 cytokines from CD4+ T-cells from asthma lung digest. CalD blocked the HDM-induced FoxO1/IRF4 pathway and restored impaired the Nrf2/HO-1 antioxidant pathway in lung tissues. CalD inhibited IL-4/IL-13-stimulated JAK1/STAT6 pathway, FoxO1 protein expression, and chemokine production in primary AMs. Structure-activity relationship study revealed the α,β-unsaturated γ-butyrolactone in CalD is capable of forming covalent bonds with cellular protein targets essential for its action. CONCLUSION AND IMPLICATIONS Our results demonstrate for the first time that CalD is a novel anti-inflammatory natural compound for allergic asthma that modulates AM function.
Collapse
Affiliation(s)
- Wupeng Liao
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Hazel Yu Ci Foo
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore
| | - Thi Ngoc Quy Tran
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore.,Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore
| | - Christina Li Lin Chai
- Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore
| | - Wai Shiu Fred Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, Singapore.,Singapore-HUJ Alliance for Research and Enterprise, National University of Singapore, Singapore.,Drug Discovery and Optimization Platform, Yong Loo Lin School of Medicine, National University Health System, Singapore
| |
Collapse
|
7
|
The Essential Role of FoxO1 in the Regulation of Macrophage Function. BIOMED RESEARCH INTERNATIONAL 2022; 2022:1068962. [PMID: 35993049 PMCID: PMC9388302 DOI: 10.1155/2022/1068962] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 07/16/2022] [Indexed: 11/17/2022]
Abstract
Macrophages are widely distributed in various tissues and organs. They not only participate in the regulation of innate and adaptive immune response, but also play an important role in tissue homeostasis. Dysregulation of macrophage function is closely related to the initiation, development and prognosis of multiple diseases, including infection and tumorigenesis. Forkhead box transcription factor O1 (FoxO1) is an important member among the forkhead box transcription factor family. Through directly binding to the promoter regions of downstream target genes, FoxO1 is implicated in cell proliferation, apoptosis, metabolic activities and other biological processes. In this review, we summarized the regulatory role of FoxO1 in macrophage phagocytosis, migration, differentiation and inflammatory activation. We also emphasized that macrophage reciprocally modulated FoxO1 activity via a post-translational modification (PTM) dominant manner.
Collapse
|
8
|
Kim JY, Stevens P, Karpurapu M, Lee H, Englert JA, Yan P, Lee TJ, Pabla N, Pietrzak M, Park GY, Christman JW, Chung S. Targeting ETosis by miR-155 inhibition mitigates mixed granulocytic asthmatic lung inflammation. Front Immunol 2022; 13:943554. [PMID: 35958610 PMCID: PMC9360579 DOI: 10.3389/fimmu.2022.943554] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/01/2022] [Indexed: 11/13/2022] Open
Abstract
Asthma is phenotypically heterogeneous with several distinctive pathological mechanistic pathways. Previous studies indicate that neutrophilic asthma has a poor response to standard asthma treatments comprising inhaled corticosteroids. Therefore, it is important to identify critical factors that contribute to increased numbers of neutrophils in asthma patients whose symptoms are poorly controlled by conventional therapy. Leukocytes release chromatin fibers, referred to as extracellular traps (ETs) consisting of double-stranded (ds) DNA, histones, and granule contents. Excessive components of ETs contribute to the pathophysiology of asthma; however, it is unclear how ETs drive asthma phenotypes and whether they could be a potential therapeutic target. We employed a mouse model of severe asthma that recapitulates the intricate immune responses of neutrophilic and eosinophilic airway inflammation identified in patients with severe asthma. We used both a pharmacologic approach using miR-155 inhibitor-laden exosomes and genetic approaches using miR-155 knockout mice. Our data show that ETs are present in the bronchoalveolar lavage fluid of patients with mild asthma subjected to experimental subsegmental bronchoprovocation to an allergen and a severe asthma mouse model, which resembles the complex immune responses identified in severe human asthma. Furthermore, we show that miR-155 contributes to the extracellular release of dsDNA, which exacerbates allergic lung inflammation, and the inhibition of miR-155 results in therapeutic benefit in severe asthma mice. Our findings show that targeting dsDNA release represents an attractive therapeutic target for mitigating neutrophilic asthma phenotype, which is clinically refractory to standard care.
Collapse
Affiliation(s)
- Ji Young Kim
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Patrick Stevens
- Comprehensive Cancer Center, Biomedical Informatics Shared Resources, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Manjula Karpurapu
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Hyunwook Lee
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Joshua A. Englert
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Pearlly Yan
- Comprehensive Cancer Center, Division of Hematology, Department of Internal Medicine, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Tae Jin Lee
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, United States
| | - Navjot Pabla
- Division of Pharmaceutics and Pharmacology, College of Pharmacy and Comprehensive Cancer Center, The Ohio State University, Columbus, OH, United States
| | - Maciej Pietrzak
- Comprehensive Cancer Center, Biomedical Informatics Shared Resources, The Ohio State University College of Medicine, Columbus, OH, United States
| | - Gye Young Park
- Section of Pulmonary, Critical Care, and Sleep Medicine, Department of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| | - John W. Christman
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| | - Sangwoon Chung
- Division of Pulmonary, Critical Care, and Sleep Medicine, Department of Internal Medicine, Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, Columbus, OH, United States
| |
Collapse
|
9
|
Jo WS, Kang S, Jeong SK, Bae MJ, Lee CG, Son Y, Lee HJ, Jeong MH, Kim SH, Moon C, Shin IS, Kim JS. Low Dose Rate Radiation Regulates M2-like Macrophages in an Allergic Airway Inflammation Mouse Model. Dose Response 2022; 20:15593258221117349. [PMID: 36003321 PMCID: PMC9393681 DOI: 10.1177/15593258221117349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Accepted: 07/15/2022] [Indexed: 11/25/2022] Open
Abstract
We investigated the effects of low dose rate radiation (LDR) on M1 and M2
macrophages in an ovalbumin-induced mouse model of allergic airway inflammation
and asthma. After exposure to LDR (1 Gy, 1.818 mGy/h) for 24 days, mice were
euthanized and the changes in the number of M1 and M2 macrophages in the
bronchoalveolar lavage fluid and lung, and M2-associated cytokine levels, were
assessed. LDR treatment not only restored the M2-rich microenvironment but also
ameliorated asthma-related progression in a macrophage-dependent manner. In an
ovalbumin-induced mouse model, LDR treatment significantly inhibited M2, but not
M1, macrophage infiltration. M2-specific changes in macrophage polarization
during chronic lung disease reversed the positive effects of LDR. Moreover, the
levels of cytokines, including chemokine (C-C motif) ligand (CCL) 24, CCL17,
transforming growth factor beta 1, and matrix metalloproteinase-9, decreased in
ovalbumin-sensitized/challenged mice upon exposure to LDR. Collectively, our
results indicate that LDR exposure suppressed asthmatic progression, including
mucin accumulation, inflammation, and Type 2 T helper (Th2) cytokine
(interleukin (IL)-4 and IL-13) production. In conclusion, LDR exposure decreased
Th2 cytokine secretion in M2 macrophages, resulting in a reduction in
eosinophilic inflammation in ovalbumin-sensitized/challenged mice.
Collapse
Affiliation(s)
- Wol Soon Jo
- Dongnam Institute of Radiological
& Medical Sciences, Busan, Republic of Korea
| | - Sohi Kang
- College of Veterinary Medicine and
BK21 Plus Project Team, Chonnam National
University, Gwangju, Republic of Korea
| | - Soo Kyung Jeong
- Dongnam Institute of Radiological
& Medical Sciences, Busan, Republic of Korea
| | - Min Ji Bae
- Dongnam Institute of Radiological
& Medical Sciences, Busan, Republic of Korea
| | - Chang Geun Lee
- Dongnam Institute of Radiological
& Medical Sciences, Busan, Republic of Korea
| | - Yeonghoon Son
- Korea Institute of Radiological &
Medical Sciences, Seoul, Republic of Korea
| | - Hae-June Lee
- Korea Institute of Radiological &
Medical Sciences, Seoul, Republic of Korea
| | - Min Ho Jeong
- Department of Microbiology, Dong-A University College of
Medicine, Busan, Republic of Korea
| | - Sung Ho Kim
- College of Veterinary Medicine and
BK21 Plus Project Team, Chonnam National
University, Gwangju, Republic of Korea
| | - Chongjong Moon
- College of Veterinary Medicine and
BK21 Plus Project Team, Chonnam National
University, Gwangju, Republic of Korea
| | - In Sik Shin
- College of Veterinary Medicine and
BK21 Plus Project Team, Chonnam National
University, Gwangju, Republic of Korea
- In Sik Shin, College of Veterinary Medicine
and BK21 Plus Project Team, Chonnam National University, Gwangju 61186, Republic
of Korea.
| | - Joong Sun Kim
- Dongnam Institute of Radiological
& Medical Sciences, Busan, Republic of Korea
- College of Veterinary Medicine and
BK21 Plus Project Team, Chonnam National
University, Gwangju, Republic of Korea
- Joong Sun Kim, College of Veterinary
Medicine and BK21 Plus Project Team, Chonnam National University, 77
Yongbong-ro, Buk-gu, Gwangju 61186, Republic of Korea.
| |
Collapse
|
10
|
Bozack AK, Colicino E, Rodosthenous RS, Bloomquist TR, Baccarelli AA, Wright RO, Wright RJ, Lee AG. Breast milk-derived extracellular vesicle miRNAs are associated with maternal asthma and atopy. Epigenomics 2022; 14:727-739. [PMID: 35638388 PMCID: PMC9280402 DOI: 10.2217/epi-2022-0090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Breast milk-derived extracellular vesicle (EV) miRNAs may program child health outcomes associated with maternal asthma and atopy. The authors investigated associations between maternal asthma/atopy and EV miRNAs in the Programming of Intergenerational Stress Mechanisms cohort. Methods: Breast milk-derived EV miRNAs collected 6.1 ± 5.9 weeks postnatally (n = 80 mothers) were profiled using the TaqMan OpenArray Human MicroRNA Panel. The authors assessed associations using adjusted robust regression. Results: Nine EV miRNAs were associated with asthma during pregnancy (a priori criteria: nominal p < 0.05; |Bregression| >0.2). miR-1290 was associated with asthma and atopy during pregnancy (p < 0.05; |Bregression| >0.2). Enriched Kyoto Encyclopedia of Genes and Genomes pathways included TGF-β signaling and extracellular matrix-receptor interaction (false discovery rate <0.05). Conclusion: In this study, maternal asthma and atopy were associated with breast milk-derived EV miRNAs. Additional studies are needed to understand whether EV miRNAs have direct effects on infant and child health.
Collapse
Affiliation(s)
- Anne K Bozack
- Department of Internal Medicine, Division of Pulmonary Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rodosthenis S Rodosthenous
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tessa R Bloomquist
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison G Lee
- Department of Internal Medicine, Division of Pulmonary Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
11
|
Yang J, Hu QC, Wang JP, Ren QQ, Wang XP, Luoreng ZM, Wei DW, Ma Y. RNA-Seq Reveals the Role of miR-29c in Regulating Inflammation and Oxidative Stress of Bovine Mammary Epithelial Cells. Front Vet Sci 2022; 9:865415. [PMID: 35433915 PMCID: PMC9011060 DOI: 10.3389/fvets.2022.865415] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 02/22/2022] [Indexed: 12/20/2022] Open
Abstract
Healthy mammary gland is essential for milk performance in dairy cows. MicroRNAs (miRNAs) are the key molecules to regulate the steady state of mammary gland in dairy cows. This study investigated the potential role of miR-29c in bovine mammary epithelial cells (bMECs). RNA sequencing (RNA-seq) was used to measure the transcriptome profile of bovine mammary epithelial cells line (MAC-T) transfected with miR-29c inhibitor or negative control (NC) inhibitor, and then differentially expressed genes (DEGs) were screened. The results showed that a total of 42 up-regulated and 27 down-regulated genes were found in the miR-29c inhibitor group compared with the NC inhibitor group. The functional enrichment of the above DEGs indicates that miR-29c is a potential regulator of oxidative stress and inflammatory response in bMECs through multiple genes, such as forkhead box O1 (FOXO1), tumor necrosis factor-alpha (TNF-α), and major histocompatibility complex, class II, DQ alpha 5 (BoLA-DQA5) in the various biological process and signaling pathways of stress-activated mitogen-activated protein kinase (MAPK) cascade, Epstein-Barr virus infection, inflammatory bowel disease, etc. The results imply that miR-29c plays an important role in a steady state of bMECs or cow mammary gland and may be a potential therapeutic target for mastitis in dairy cows.
Collapse
Affiliation(s)
- Jian Yang
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qi-Chao Hu
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Jin-Peng Wang
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Qian-Qian Ren
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Xing-Ping Wang
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
- *Correspondence: Xing-Ping Wang
| | - Zhuo-Ma Luoreng
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
- Zhuo-Ma Luoreng
| | - Da-Wei Wei
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| | - Yun Ma
- School of Agriculture, Ningxia University, Yinchuan, China
- Key Laboratory of Ruminant Molecular Cell Breeding, Ningxia Hui Autonomous Region, Yinchuan, China
| |
Collapse
|
12
|
Li XY, Wang YJ, Chen S, Pan LH, Li QM, Luo JP, Zha XQ. Laminaria japonica Polysaccharide Suppresses Atherosclerosis via Regulating Autophagy-Mediated Macrophage Polarization. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3633-3643. [PMID: 35167294 DOI: 10.1021/acs.jafc.1c07483] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
The present work aimed to explore the effect and underlying mechanism of a homogeneous Laminaria japonica polysaccharide (LJP61A) on macrophage polarization in high-fat-diet-fed LDLr-/- mice and Ox-LDL-induced macrophages. Results showed that LJP61A remarkably reduced the lesion burden in atherosclerotic mice, alleviated lipid deposition in Ox-LDL-stimulated macrophages, decreased the expression of M1 macrophage markers, and increased the expression of M2 macrophage markers, thus reducing the M1/M2 macrophage phenotype ratio. Meanwhile, the autophagic flux of macrophages was enhanced by LJP61A treatment in vitro and in vivo. 3-Methyladenine is an autophagic inhibitor. As expected, this inhibitor blocked the effects of LJP61A on macrophage polarization. SIRT1 and FoxO1 are two key upstream genes that control the autophagy behavior. We also found that LJP61A significantly up-regulated the expression of SIRT1 and FoxO1. However, these effects of LJP61A were abolished by the SIRT1 siRNA and FoxO1 inhibitor AS1842856. These results suggested that LJP61A reduced atherosclerosis in HFD-induced LDLr-/- mice via regulating autophagy-mediated macrophage polarization.
Collapse
Affiliation(s)
- Xue-Ying Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Yu-Jing Wang
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Shun Chen
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Li-Hua Pan
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Qiang-Ming Li
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Jian-Ping Luo
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| | - Xue-Qiang Zha
- Engineering Research Centre of Bioprocess of Ministry of Education, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- School of Food and Biological Engineering, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
- Key Laboratory of Metabolism and Regulation for Major Disease of Anhui Higher Education Institutes, Hefei University of Technology, No 193 Tunxi Road, Hefei 230009, People's Republic of China
| |
Collapse
|
13
|
Wang SR, Hu RD, Ma M, You X, Cui H, He Y, Xu D, Zhao ZB, Selmi C, Eric Gershwin M, Li L, Lian ZX. FoxO1 suppresses IL-10 producing B cell differentiation via negatively regulating Blimp-1 expression and contributes to allergic asthma progression. Mucosal Immunol 2022; 15:459-470. [PMID: 35322189 DOI: 10.1038/s41385-022-00504-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 02/16/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023]
Abstract
IL-10-producing B cells (B10) are involved in the prevention of autoimmune and allergic responses but its mechanisms remain poorly understood. We took advantage of the ovalbumin-induced asthma mouse model to demonstrate that the activity of FoxO1 is upregulated in lung B cells and correlates inversely with B10 cells, while showing decreased activity in ex vivo and in vitro induced B10 cells. We further observed that FoxO1 deficiency leads to increased frequency of B10 cells. These observations have in vivo clinical evidence, as B cell specific FoxO1 deficiency leads to reduced lung eosinophils and asthma remission in mice, and there are reduced regulatory B cells and increased FoxO1 activity in B cells of asthma patients. Single cell RNA-sequencing data demonstrated a negative correlation between the expression of Foxo1 and Il10 in B cells from the mouse spleen and lung and the human lung. For a biological mechanism, FoxO1 inhibits the expression of Prdm1, which encodes Blimp-1, a transcription factor of B10 cells. Our experimental evidence in both murine and human asthma demonstrates that FoxO1 is a negative regulator of B10 cell differentiation via negatively regulating Prdm1 and its expression in B cells contributes to allergic asthma disease.
Collapse
Affiliation(s)
- Song-Rong Wang
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Ren-Dong Hu
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Min Ma
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China
| | - Xing You
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou, 511442, P. R. China
| | - Haiyan Cui
- Department of Respiratory and Critical Care Medicine, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, 510630, China
| | - Yi He
- Department of Rheumatology and Immunology, The Third Affiliated Hospital, Southern Medical University, Guangzhou, Guangdong, 510630, China
| | - Damo Xu
- State Key Laboratory of Respiratory Disease for Allergy at Shenzhen University, Shenzhen Key Laboratory of Allergy & Immunology, Shenzhen University School of Medicine, Shenzhen, China
| | - Zhi-Bin Zhao
- Medical Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Carlo Selmi
- Rheumatology and Clinical Immunology, Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy.,Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - M Eric Gershwin
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, Davis, CA, USA
| | - Liang Li
- Medical Research Center, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Zhe-Xiong Lian
- Chronic Disease Laboratory, School of Medicine, South China University of Technology, Guangzhou, China. .,Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sicences, Guangzhou, China.
| |
Collapse
|
14
|
LncTRPM2-AS inhibits TRIM21-mediated TRPM2 ubiquitination and prevents autophagy-induced apoptosis of macrophages in asthma. Cell Death Dis 2021; 12:1153. [PMID: 34903714 PMCID: PMC8668916 DOI: 10.1038/s41419-021-04437-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2021] [Revised: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 11/24/2022]
Abstract
Long non-coding RNAs (lncRNAs) play a crucial role in macrophage development but little is known about their role in asthma. Here, we investigated the role of lncRNA lncTRPM2-AS in asthma and found that lncTRPM2-AS participates in the promotion of macrophage inflammation. Downregulation of lncTRPM2-AS promoted apoptosis and inhibited proliferation and production of cytokines including IL-1β, IL-4, IL-6, IL-10, TNF-α, and TGF-β. RNA-immunoprecipitation and mass spectrometry indicated that the protein TRPM2 interacted with both lncTRPM2-AS and the E3 ubiquitin ligase TRIM21. LncTRPM2-AS silencing enhanced the interaction between TRIM21 and TRPM2, resulting in elevated levels of ubiquitin-related degradation of TRPM2. Mutation analysis indicated that TRPM2 K1218 is a key site for TRIM21-dependent ubiquitination. Downregulation of lncTRPM2-AS significantly decreased intracellular calcium levels by restraining TRPM2 protein expression, which in turn decreased ROS levels and increased autophagy to promote macrophage apoptosis and reduce cytokine production, together inhibiting macrophage inflammation. Taken together, our findings demonstrate that lncTRPM2-AS blocks the ubiquitination of TRPM2 via TRIM21 and inhibits autophagy-induced apoptosis which may contribute to macrophage inflammation in asthma.
Collapse
|
15
|
Abu Khweek A, Joldrichsen MR, Kim E, Attia Z, Krause K, Daily K, Estfanous S, Hamilton K, Badr A, Anne MNK, Eltobgy M, Corps KN, Carafice C, Zhang X, Gavrilin MA, Boyaka PN, Amer AO. Caspase-11 regulates lung inflammation in response to house dust mites. Cell Immunol 2021; 370:104425. [PMID: 34800762 PMCID: PMC8714054 DOI: 10.1016/j.cellimm.2021.104425] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 08/15/2021] [Accepted: 08/17/2021] [Indexed: 10/20/2022]
Abstract
Asthma is an inflammatory lung disorder characterized by mucus hypersecretion, cellular infiltration, and bronchial hyper-responsiveness. House dust mites (HDM) are the most prevalent cause of allergic sensitization. Canonical and noncanonical inflammasomes are multiprotein complexes that assemble in response to pathogen or danger-associated molecular patterns (PAMPs or DAMPs). Murine caspase-11 engages the noncanonical inflammasome. We addressed the role of caspase-11 in mediating host responses to HDM and subsequent allergic inflammation using caspase-11-/- mice, which lack caspase-11 while express caspase-1. We found that HDM induce caspase-11 expression in vitro. The presence of IL-4 and IL-13 promote caspase-11 expression. Additionally, caspase-11-/- macrophages show reduced release of IL-6, IL-12, and KC, and express lower levels of costimulatory molecules (e.g., CD40, CD86 and MHCII) in response to HDM stimulation. Notably, HDM sensitization of caspase-11-/- mice resulted in similar levels of IgE responses and hypothermia in response to nasal HDM challenge compared to WT. However, analysis of cell numbers and cytokines in bronchiolar alveolar lavage fluid (BALF) and histopathology of representative lung segments demonstrate altered inflammatory responses and reduced neutrophilia in the airways of the caspase-11-/- mice. These findings indicate that caspase-11 regulates airway inflammation in response to HDM exposure.
Collapse
Affiliation(s)
- Arwa Abu Khweek
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA; Department of Biology and Biochemistry, Birzeit University, West Bank, Palestine
| | - Marisa R Joldrichsen
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Eunsoo Kim
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Zayed Attia
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Kathrin Krause
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kylene Daily
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Shady Estfanous
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kaitlin Hamilton
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Asmaa Badr
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Midhun N K Anne
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Mostafa Eltobgy
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Kara N Corps
- Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA
| | - Cierra Carafice
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA
| | - Xiaoli Zhang
- Center for Biostatistics, The Ohio State University, Columbus OH 43210, USA
| | - Mikhail A Gavrilin
- Department of Internal Medicine, The Ohio State University, Columbus OH 43210, USA
| | - Prosper N Boyaka
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA; Department of Veterinary Biosciences, The Ohio State University, Columbus OH 43210, USA; Infectious Diseases Institute, The Ohio State University, Columbus OH 43210, USA.
| | - Amal O Amer
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus OH 43210, USA.
| |
Collapse
|
16
|
Adiponectin and Asthma: Knowns, Unknowns and Controversies. Int J Mol Sci 2021; 22:ijms22168971. [PMID: 34445677 PMCID: PMC8396527 DOI: 10.3390/ijms22168971] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/11/2021] [Accepted: 08/16/2021] [Indexed: 12/14/2022] Open
Abstract
Adiponectin is an adipokine associated with the healthy obese phenotype. Adiponectin increases insulin sensitivity and has cardio and vascular protection actions. Studies related to adiponectin, a modulator of the innate and acquired immunity response, have suggested a role of this molecule in asthma. Studies based on various asthma animal models and on the key cells involved in the allergic response have provided important insights about this relation. Some of them indicated protection and others reversed the balance towards negative effects. Many of them described the cellular pathways activated by adiponectin, which are potentially beneficial for asthma prevention or for reduction in the risk of exacerbations. However, conclusive proofs about their efficiency still need to be provided. In this article, we will, briefly, present the general actions of adiponectin and the epidemiological studies supporting the relation with asthma. The main focus of the current review is on the mechanisms of adiponectin and the impact on the pathobiology of asthma. From this perspective, we will provide arguments for and against the positive influence of this molecule in asthma, also indicating the controversies and sketching out the potential directions of research to complete the picture.
Collapse
|
17
|
Shaw OM, Hurst RD, Cooney J, Sawyer GM, Dinnan H, Martell S. Boysenberry and apple juice concentrate reduced acute lung inflammation and increased M2 macrophage-associated cytokines in an acute mouse model of allergic airways disease. Food Sci Nutr 2021; 9:1491-1503. [PMID: 33747463 PMCID: PMC7958577 DOI: 10.1002/fsn3.2119] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 12/22/2020] [Accepted: 12/24/2020] [Indexed: 12/11/2022] Open
Abstract
Bioactive compounds including anthocyanins and other polyphenols are associated with reduced lung inflammation and improved lung function in asthma and other lung diseases. This study investigated the effects of a Boysenberry and apple juice concentrate, high in cyanidin glycosides, ellagitannins, and chlorogenic acid, on a mouse model of allergic airways inflammation. Male C57BL/6J mice were orally gavaged with 2.5 mg/kg of total anthocyanins (TAC) from BerriQi® Boysenberry and apple juice concentrate (0.2 mg/kg human equivalent dose) or water control 1 hr before an acute intranasal ovalbumin (OVA) challenge and were gavaged again 2 days after the intranasal challenge. Consumption of BerriQi® Boysenberry and apple juice concentrate significantly decreased OVA-induced infiltrating eosinophils, neutrophils, and T cells in the lung, and mucous production. Quantification of gene expression for arginase (Arg1), chitinase 3-like 3 (Ym-1), found in inflammatory zone (Fizz1), which have been associated with an anti-inflammatory macrophage phenotype (M2), found significantly increased Arg1 expression in the lung in the Boysenberry and apple juice concentrate treatment group. There was also increased production of M2-associated cytokines C-X-C motif chemokine ligand (CXCL) 10 and C-C motif chemokine ligand (CCL) 4. These results suggest that consumption of BerriQi® Boysenberry and apple juice concentrate promoted a shift toward an anti-inflammatory environment within the lung leading to reduced immune cell infiltration and tissue damage.
Collapse
Affiliation(s)
- Odette M. Shaw
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Roger D. Hurst
- Food Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Janine Cooney
- Biological Chemistry & Bioactives GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedHamiltonNew Zealand
| | - Gregory M. Sawyer
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Hannah Dinnan
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| | - Sheridan Martell
- Nutrition & Health GroupFood Innovation PortfolioThe New Zealand Institute for Plant and Food Research LimitedPalmerston NorthNew Zealand
| |
Collapse
|
18
|
Dilated cardiomyopathy impairs mitochondrial biogenesis and promotes inflammation in an age- and sex-dependent manner. Aging (Albany NY) 2020; 12:24117-24133. [PMID: 33303703 PMCID: PMC7762497 DOI: 10.18632/aging.202283] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Dilated cardiomyopathy (DCM) belongs to the myocardial diseases associated with a severe impairment of cardiac function, but the question of how sex and age affect this pathology has not been fully explored. Impaired energy homeostasis, mitochondrial dysfunction, and systemic inflammation are well-described phenomena associated with aging. In this study, we investigated if DCM affects these phenomena in a sex- and age-related manner. We analyzed the expression of mitochondrial and antioxidant proteins and the inflammatory state in DCM heart tissue from younger and older women and men. A significant downregulation of Sirt1 expression was detected in older DCM patients. Sex-related differences were observed in the phosphorylation of AMPK that only appeared in older males with DCM, possibly due to an alternative Sirt1 regulation mechanism. Furthermore, reduced expression of several mitochondrial proteins (TOM40, TIM23, Sirt3, and SOD2) and genes (cox1, nd4) was only detected in old DCM patients, suggesting that age has a greater effect than DCM on these alterations. Finally, an increased expression of inflammatory markers in older, failing hearts, with a stronger pro-inflammatory response in men, was observed. Together, these findings indicate that age- and sex-related increased inflammation and disturbance of mitochondrial homeostasis occurs in male individuals with DCM.
Collapse
|
19
|
Helling BA, Sobreira DR, Hansen GT, Sakabe NJ, Luo K, Billstrand C, Laxman B, Nicolae RI, Nicolae DL, Bochkov YA, Gern JE, Nobrega MA, White SR, Ober C. Altered transcriptional and chromatin responses to rhinovirus in bronchial epithelial cells from adults with asthma. Commun Biol 2020; 3:678. [PMID: 33188283 PMCID: PMC7666152 DOI: 10.1038/s42003-020-01411-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Accepted: 10/21/2020] [Indexed: 12/20/2022] Open
Abstract
There is a life-long relationship between rhinovirus (RV) infection and the development and clinical manifestations of asthma. In this study we demonstrate that cultured primary bronchial epithelial cells from adults with asthma (n = 9) show different transcriptional and chromatin responses to RV infection compared to those without asthma (n = 9). Both the number and magnitude of transcriptional and chromatin responses to RV were muted in cells from asthma cases compared to controls. Pathway analysis of the transcriptionally responsive genes revealed enrichments of apoptotic pathways in controls but inflammatory pathways in asthma cases. Using promoter capture Hi-C we tethered regions of RV-responsive chromatin to RV-responsive genes and showed enrichment of these regions and genes at asthma GWAS loci. Taken together, our studies indicate a delayed or prolonged inflammatory state in cells from asthma cases and highlight genes that may contribute to genetic risk for asthma.
Collapse
Affiliation(s)
- Britney A Helling
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA.
| | - Débora R Sobreira
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Grace T Hansen
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Noboru J Sakabe
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Kaixuan Luo
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | | | - Bharathi Laxman
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Raluca I Nicolae
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Dan L Nicolae
- Department of Statistics, University of Chicago, Chicago, IL, 60637, USA
| | - Yury A Bochkov
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - James E Gern
- Department of Pediatrics, University of Wisconsin, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Marcelo A Nobrega
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| | - Steven R White
- Section of Pulmonary and Critical Care Medicine, Department of Medicine, University of Chicago, Chicago, IL, 60637, USA
| | - Carole Ober
- Department of Human Genetics, University of Chicago, Chicago, IL, 60637, USA
| |
Collapse
|
20
|
Tang B, Qiu J, Hu S, Li L, Wang J. Role of stearyl-coenzyme A desaturase 1 in mediating the effects of palmitic acid on endoplasmic reticulum stress, inflammation, and apoptosis in goose primary hepatocytes. Anim Biosci 2020; 34:1210-1220. [PMID: 33152216 PMCID: PMC8255868 DOI: 10.5713/ajas.20.0444] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/05/2020] [Indexed: 11/27/2022] Open
Abstract
Objective Unlike mammals, goose fatty liver shows a strong tolerance to fatty acids without obvious injury. Stearyl-coenzyme A desaturase 1 (SCD1) serves crucial role in desaturation of saturated fatty acids (SAFs), but its role in the SAFs tolerance of goose hepatocytes has not been reported. This study was conducted to explore the role of SCD1 in regulating palmitic acid (PA) tolerance of goose primary hepatocytes. Methods 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide was examined to reflect the effect of PA on hepatocytes viability, and quantitative polymerase chain reaction was used to detect the mRNA levels of several genes related to endoplasmic reticulum (ER) stress, inflammation, and apoptosis, and the role of SCD1 in PA tolerance of goose hepatocytes was explored using RNA interfere. Results Our results indicated that goose hepatocytes exhibited a higher tolerant capacity to PA than human hepatic cell line (LO2 cells). In goose primary hepatocytes, the mRNA levels of fatty acid desaturation-related genes (SCD1 and fatty acid desaturase 2) and fatty acid elongate enzyme-related gene (elongase of very long chain fatty acids 6) were significantly upregulated with 0.6 mM PA treatment. However, in LO2 cells, expression of ER stress-related genes (x box-binding protein, binding immunoglobulin protein, and activating transcription factor 6), inflammatory response-related genes (interleukin-6 [IL-6], interleukin-1β [IL-1β], and interferon-γ) and apoptosis-related genes (bcl-2-associated X protein, b-cell lymphoma 2, Caspase-3, and Caspase-9) was significantly enhanced with 0.6 mM PA treatment. Additionally, small interfering RNA (siRNA) mediated downregulation of SCD1 significantly reduced the PA tolerance of goose primary hepatocytes under the treatment of 0.6 mM PA; meanwhile, the mRNA levels of inflammatory-related genes (IL-6 and IL-1β) and several key genes involved in the phosphoinositide 3-kinase (PI3K)/protein kinase B (AKT), forkhead box O1 (FoxO1), mammalian target of rapamycin and AMPK pathways (AKT1, AKT2, FoxO1, and sirtuin 1), as well as the protein expression of cytochrome C and the apoptosis rate were upregulated. Conclusion In conclusion, our data suggested that SCD1 was involved in enhancing the PA tolerance of goose primary hepatocytes by regulating inflammation- and apoptosis-related genes expression.
Collapse
Affiliation(s)
- Bincheng Tang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 61130, China
| | - Jiamin Qiu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 61130, China
| | - Shenqiang Hu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 61130, China
| | - Liang Li
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 61130, China
| | - Jiwen Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory, Sichuan Agricultural University, Chengdu, Sichuan, 61130, China
| |
Collapse
|
21
|
COVID-19: Proposing a Ketone-Based Metabolic Therapy as a Treatment to Blunt the Cytokine Storm. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:6401341. [PMID: 33014275 PMCID: PMC7519203 DOI: 10.1155/2020/6401341] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Revised: 06/22/2020] [Accepted: 08/17/2020] [Indexed: 02/07/2023]
Abstract
Human SARS-CoV-2 infection is characterized by a high mortality rate due to some patients developing a large innate immune response associated with a cytokine storm and acute respiratory distress syndrome (ARDS). This is characterized at the molecular level by decreased energy metabolism, altered redox state, oxidative damage, and cell death. Therapies that increase levels of (R)-beta-hydroxybutyrate (R-BHB), such as the ketogenic diet or consuming exogenous ketones, should restore altered energy metabolism and redox state. R-BHB activates anti-inflammatory GPR109A signaling and inhibits the NLRP3 inflammasome and histone deacetylases, while a ketogenic diet has been shown to protect mice from influenza virus infection through a protective γδ T cell response and by increasing electron transport chain gene expression to restore energy metabolism. During a virus-induced cytokine storm, metabolic flexibility is compromised due to increased levels of reactive oxygen species (ROS) and reactive nitrogen species (RNS) that damage, downregulate, or inactivate many enzymes of central metabolism including the pyruvate dehydrogenase complex (PDC). This leads to an energy and redox crisis that decreases B and T cell proliferation and results in increased cytokine production and cell death. It is hypothesized that a moderately high-fat diet together with exogenous ketone supplementation at the first signs of respiratory distress will increase mitochondrial metabolism by bypassing the block at PDC. R-BHB-mediated restoration of nucleotide coenzyme ratios and redox state should decrease ROS and RNS to blunt the innate immune response and the associated cytokine storm, allowing the proliferation of cells responsible for adaptive immunity. Limitations of the proposed therapy include the following: it is unknown if human immune and lung cell functions are enhanced by ketosis, the risk of ketoacidosis must be assessed prior to initiating treatment, and permissive dietary fat and carbohydrate levels for exogenous ketones to boost immune function are not yet established. The third limitation could be addressed by studies with influenza-infected mice. A clinical study is warranted where COVID-19 patients consume a permissive diet combined with ketone ester to raise blood ketone levels to 1 to 2 mM with measured outcomes of symptom severity, length of infection, and case fatality rate.
Collapse
|
22
|
Szczepankiewicz D, Langwiński W, Kołodziejski P, Pruszyńska-Oszmałek E, Sassek M, Nowakowska J, Chmurzyńska A, Nowak KW, Szczepankiewicz A. Allergic Inflammation Alters microRNA Expression Profile in Adipose Tissue in the Rat. Genes (Basel) 2020; 11:genes11091034. [PMID: 32887419 PMCID: PMC7564923 DOI: 10.3390/genes11091034] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 08/29/2020] [Accepted: 08/31/2020] [Indexed: 01/28/2023] Open
Abstract
Adipose tissue is a major source of circulating exosomal microRNAs (miRNAs) that are modulators of the immune response in various types of tissues and organs, including airways. Still, no evidence exists if allergic airway inflammation may affect fat tissue inflammation via alterations in the miRNA expression profile. Therefore, we investigated the miRNA expression profile in the adipose tissue upon induced allergic inflammation in the airways in the rat. Brown Norway rats were chronically sensitized to house dust mite extract for seven weeks. Body composition was performed using MiniSpec Plus. The eosinophil count and the total IgE level were determined to confirm the induction of allergic inflammation. MiRNA expression profiling was done using the next-generation sequencing with validation by qPCR. We found that allergic airway inflammation significantly increased fat in adipose tissue, glucose concentration, and the gene expression of adipose tissue-derived proinflammatory peptides (leptin, TNFα). In miRNA-seq analysis, we showed significant differences in the expression of 36 mature miRNAs, three precursors, and two miRNA families in adipose tissue of allergic rats. Two miRNAs—miRNA-151-5p and miRNA-423-3p—showed significantly increased expression in qPCR in adipose tissue and lungs of sensitized animals. Allergic airway inflammation affects fat tissue and alters miRNA expression profile in adipose tissue in the rat.
Collapse
Affiliation(s)
- Dawid Szczepankiewicz
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Wojciech Langwiński
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (W.L.); (J.N.)
| | - Paweł Kołodziejski
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Ewa Pruszyńska-Oszmałek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Maciej Sassek
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Joanna Nowakowska
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (W.L.); (J.N.)
| | - Agata Chmurzyńska
- Institute of Human Nutrition and Dietetics, Poznań University of Life Sciences, 60-624 Poznań, Poland;
| | - Krzysztof W. Nowak
- Department of Animal Physiology, Biochemistry and Biostructure, Poznań University of Life Sciences, 60-637 Poznań, Poland; (D.S.); (P.K.); (E.P.-O.); (M.S.); (K.W.N.)
| | - Aleksandra Szczepankiewicz
- Molecular and Cell Biology Unit, Department of Pediatric Pulmonology, Allergy and Clinical Immunology, Poznan University of Medical Sciences, 60-572 Poznań, Poland; (W.L.); (J.N.)
- Correspondence: ; Tel.: +48-61-88547643; Fax: +48-618547663
| |
Collapse
|
23
|
Transcriptome sequencing of circular RNA reveals a novel circular RNA-has_circ_0114427 in the regulation of inflammation in acute kidney injury. Clin Sci (Lond) 2020; 134:139-154. [PMID: 31930399 DOI: 10.1042/cs20190990] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 01/04/2020] [Accepted: 01/13/2020] [Indexed: 02/07/2023]
Abstract
Acute kidney injury (AKI) is a common serious syndrome characterized by rapid decrease of glomerular filtration rate and the progressive increase of serum creatinine. Circular RNAs (circRNAs) are regulatory RNAs that recently became popular among various diseases. However, the expression profile and function of circRNAs in AKI remain largely unknown. The main function of circRNAs is acting as competing endogenous RNAs (ceRNAs) by binding with microRNAs (miRNAs), as indicated by recent research. In the present study, we established cisplatin-induced AKI model in mice and isolated renal tubular tissues to extract circRNAs for next-generation sequencing (NGS) and bioinformatics analysis. We analyzed the composition, distribution and Gene Ontology terms of circRNAs in cisplatin-induced AKI and revealed differentially expressed circRNAs related to AKI. By finding homologous genes between mouse and human, we identified circRNA- circ-0114427 in humans. We further investigated its function in AKI cell model. Circ-0114427 expression was significantly up-regulated in different AKI cell models. Knockdown of circ-0114427 indicated that circ-0114427 bound to miR-494 as a miRNA sponge to regulate ATF3 expression and further affected the expression of downstream cytokine IL-6. Circ-0114427 regulates inflammatory progression in AKI's early stage via circ-0114427/miR-494/ATF3 pathway. Our findings reveal the expression profile of circRNAs in cisplatin-induced AKI and provide a novel insight into the regulatory mechanism of circRNAs, which may become a new molecular target resource for early diagnosis and treatment strategies.
Collapse
|
24
|
Genome‑wide analysis of DNA methylation and gene expression changes in an ovalbumin‑induced asthma mouse model. Mol Med Rep 2020; 22:1709-1716. [PMID: 32705270 PMCID: PMC7411290 DOI: 10.3892/mmr.2020.11245] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 02/04/2020] [Indexed: 12/12/2022] Open
Abstract
The aim of the present study was to establish an integrated network of DNA methylation and RNA expression in an ovalbumin (OVA)-induced asthma model, and to investigate the epigenetically-regulated genes involved in asthma development. Genome-wide CpG-DNA methylation profiling was conducted through the use of a methylated DNA immunoprecipitation microarray and RNA sequencing was performed using three lung samples from mice with OVA-induced asthma. A total of 35,401 differentially methylated regions (DMRs) were identified between mice with OVA-induced asthma and control mice. Of these, 3,060 were located in promoter regions and 370 of the genes containing these DMRs demonstrated an inverse correlation between methylation and gene expression. Kyoto Encyclopedia of Genes and Genomes pathway analysis identified that 368 genes were upregulated or downregulated in OVA-induced asthma samples, including genes involved in ‘chemokine signalling pathway’, ‘focal adhesion’, ‘leukocyte transendothelial migration’ and ‘vascular smooth muscle contraction signaling’ pathways. Integrated network analysis identified four hub genes, consisting of three upregulated genes [forkhead box O1 (FOXO1), SP1 transcription factor (SP1) and amyloid β precursor protein (APP)], and one downregulated gene [RUNX family transcription factor 1 (RUNX1)], all of which demonstrated an association between DNA methylation and gene expression. These genes were highly interconnected nodes in the Ingenuity Pathway Analysis module and were functionally significant. A total of four interconnected hub genes, FOXO1, RUNX1, SP1 and APP, were identified from the integrated DNA methylation and gene expression networks involved in asthma development. These results suggested that modulating these four genes could effectively control the development of asthma.
Collapse
|
25
|
Chung S, Lee YG, Karpurapu M, Englert JA, Ballinger MN, Davis IC, Park GY, Christman JW. Depletion of microRNA-451 in response to allergen exposure accentuates asthmatic inflammation by regulating Sirtuin2. Am J Physiol Lung Cell Mol Physiol 2020; 318:L921-L930. [PMID: 32159972 PMCID: PMC7272736 DOI: 10.1152/ajplung.00457.2019] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2019] [Revised: 03/06/2020] [Accepted: 03/10/2020] [Indexed: 01/13/2023] Open
Abstract
The incidence of asthma has increased from 5.5% to near 8% of the population, which is a major health concern. The hallmarks of asthma include eosinophilic airway inflammation that is associated with chronic airway remodeling. Allergic airway inflammation is characterized by a complex interplay of resident and inflammatory cells. MicroRNAs (miRNAs) are small noncoding RNAs that function as posttranscriptional modulators of gene expression. However, the role of miRNAs, specifically miR-451, in the regulation of allergic airway inflammation is unexplored. Our previous findings showed that oxidant stress regulates miR-451 gene expression in macrophages during an inflammatory process. In this paper, we examined the role of miR-451 in regulating macrophage phenotype using an experimental poly-allergenic murine model of allergic airway inflammation. We found that miR-451 contributes to the allergic induction of CCL17 in the lung and plays a key role in proasthmatic macrophage activation. Remarkably, administration of a Sirtuin 2 (Sirt2) inhibitor diminished alternate macrophage activation and markedly abrogated triple-allergen [dust mite, ragweed, Aspergillus fumigatus (DRA)]-induced lung inflammation. These data demonstrate a role for miR-451 in modulating allergic inflammation by influencing allergen-mediated macrophages phenotype.
Collapse
Affiliation(s)
- Sangwoon Chung
- Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Institute, Columbus, Ohio
| | - Yong Gyu Lee
- Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Institute, Columbus, Ohio
| | - Manjula Karpurapu
- Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Institute, Columbus, Ohio
| | - Joshua A Englert
- Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Institute, Columbus, Ohio
| | - Megan N Ballinger
- Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Institute, Columbus, Ohio
| | - Ian C Davis
- College of Veterinary Medicine, the Ohio State University, Columbus, Ohio
| | - Gye Young Park
- Department of Medicine, Section of Pulmonary, Critical Care, and Sleep Medicine, University of Illinois at Chicago, Chicago, Illinois
| | - John W Christman
- Pulmonary, Critical Care, and Sleep Medicine, the Ohio State University, Davis Heart and Lung Research Institute, Columbus, Ohio
| |
Collapse
|
26
|
Gaspar N, Zambito G, Löwik CMWG, Mezzanotte L. Active Nano-targeting of Macrophages. Curr Pharm Des 2020; 25:1951-1961. [PMID: 31291874 DOI: 10.2174/1381612825666190710114108] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Accepted: 06/25/2019] [Indexed: 01/01/2023]
Abstract
Macrophages play a role in almost every disease such as cancer, infections, injuries, metabolic and inflammatory diseases and are becoming an attractive therapeutic target. However, understanding macrophage diversity, tissue distribution and plasticity will help in defining precise targeting strategies and effective therapies. Active targeting of macrophages using nanoparticles for therapeutic purposes is still at its infancy but holds promises since macrophages have shown high specific uptake of nanoparticles. Here, we highlight recent progress in active nanotechnology-based systems gaining pivotal roles to target diverse macrophage subsets in diseased tissues.
Collapse
Affiliation(s)
- Natasa Gaspar
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Percuros B.V., Department of Developmental BioEngineering, University of Twente, Enschede, Netherlands
| | - Giorgia Zambito
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Medres-Medical Research gmbh, Cologne, Germany
| | - Clemens M W G Löwik
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Oncology, Lausanne University Hospital (CHUV), UNIL, Switzerland
| | - Laura Mezzanotte
- Optical Molecular Imaging, Department of Radiology and Nuclear Medicine, Erasmus Medical Center, Rotterdam, Netherlands.,Department of Molecular Genetics, Erasmus Medical Center, Rotterdam, Netherlands
| |
Collapse
|
27
|
Roux A, Bismuth G, Mangeney M. [FOXO1 transcription factor: a key player in T cell/HIV-1 interaction]. Med Sci (Paris) 2020; 36:24-26. [PMID: 32014093 DOI: 10.1051/medsci/2019256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Arthur Roux
- Institut Cochin, CNRS UMR8104, Inserm U1016, université Paris Descartes, 22 rue Méchain, 75014 Paris, France
| | - Georges Bismuth
- Institut Cochin, CNRS UMR8104, Inserm U1016, université Paris Descartes, 22 rue Méchain, 75014 Paris, France
| | - Marianne Mangeney
- Institut Cochin, CNRS UMR8104, Inserm U1016, université Paris Descartes, 22 rue Méchain, 75014 Paris, France
| |
Collapse
|
28
|
Draijer C, Speth JM, Penke LRK, Zaslona Z, Bazzill JD, Lugogo N, Huang YJ, Moon JJ, Peters-Golden M. Resident alveolar macrophage-derived vesicular SOCS3 dampens allergic airway inflammation. FASEB J 2020; 34:4718-4731. [PMID: 32030817 DOI: 10.1096/fj.201903089r] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/22/2020] [Accepted: 01/22/2020] [Indexed: 01/28/2023]
Abstract
Resident alveolar macrophages (AMs) suppress allergic inflammation in murine asthma models. Previously we reported that resident AMs can blunt inflammatory signaling in alveolar epithelial cells (ECs) by transcellular delivery of suppressor of cytokine signaling 3 (SOCS3) within extracellular vesicles (EVs). Here we examined the role of vesicular SOCS3 secretion as a mechanism by which AMs restrain allergic inflammatory responses in airway ECs. Bronchoalveolar lavage fluid (BALF) levels of SOCS3 were reduced in asthmatics and in allergen-challenged mice. Ex vivo SOCS3 secretion was reduced in AMs from challenged mice and this defect was mimicked by exposing normal AMs to cytokines associated with allergic inflammation. Both AM-derived EVs and synthetic SOCS3 liposomes inhibited the activation of STAT3 and STAT6 as well as cytokine gene expression in ECs challenged with IL-4/IL-13 and house dust mite (HDM) extract. This suppressive effect of EVs was lost when they were obtained from AMs exposed to allergic inflammation-associated cytokines. Finally, inflammatory cell recruitment and cytokine generation in the lungs of OVA-challenged mice were attenuated by intrapulmonary pretreatment with SOCS3 liposomes. Overall, AM secretion of SOCS3 within EVs serves as a brake on airway EC responses during allergic inflammation, but is impaired in asthma. Synthetic liposomes encapsulating SOCS3 can rescue this defect and may serve as a framework for novel therapeutic approaches targeting airway inflammation.
Collapse
Affiliation(s)
- Christina Draijer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Jennifer M Speth
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Loka R K Penke
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Zbigniew Zaslona
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Joseph D Bazzill
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Njira Lugogo
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Yvonne J Huang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - James J Moon
- Department of Pharmaceutical Sciences, University of Michigan, Ann Arbor, MI, USA.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA.,Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA.,Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI, USA
| |
Collapse
|
29
|
Shang Y, Sun Y, Xu J, Ge X, Hu Z, Xiao J, Ning Y, Dong Y, Bai C. Exosomes from mmu_circ_0001359-Modified ADSCs Attenuate Airway Remodeling by Enhancing FoxO1 Signaling-Mediated M2-like Macrophage Activation. MOLECULAR THERAPY-NUCLEIC ACIDS 2020; 19:951-960. [PMID: 32018116 PMCID: PMC6997502 DOI: 10.1016/j.omtn.2019.10.049] [Citation(s) in RCA: 57] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2019] [Revised: 10/03/2019] [Accepted: 10/23/2019] [Indexed: 02/07/2023]
Abstract
Asthma is the most common chronic disease and is characterized by airway remodeling and chronic inflammation. Increasingly, studies have found that the activation and M1 phenotypic transformation of macrophages play important roles in asthma progress, including airway remodeling. However, the reversal of M1 macrophages to the M2 phenotype has been shown to attenuate airway remodeling. Exosomes are nano-sized extracellular vesicles derived from endosomes; they play direct roles in governing physiological and pathological conditions by the intracellular transfer of bioactive cargo, such as proteins, enzymes, nucleic acids (microRNA [miRNA], mRNA, DNA), and metabolites. However, transfer mechanisms are unclear. To uncover potential therapeutic mechanisms, we constructed an ovalbumin-induced asthma mouse model and lipopolysaccharide-induced RAW264.7 macrophages cells. High-throughput sequencing showed that mmu_circ_0001359 was downregulated in asthmatic mice when compared with normal mice. Adipose-derived stem cell (ADSC)-exosome treatment suppressed inflammatory cytokine expression by the conversion of M1 macrophages to the M2 phenotype, under lipopolysaccharide-induced conditions. Exosomes from mmu_circ_0001359 overexpression in ADSCs increased therapeutic effects, in terms of cytokine expression, when compared with wild-type exosomes. Luciferase reporter assays confirmed that exosomes from mmu_circ_0001359-modified ADSCs attenuated airway remodeling by enhancing FoxO1 signaling-mediated M2-like macrophage activation, via sponging miR-183-5p. In conclusion, mmu_circ_0001359-enriched exosomes attenuated airway remodeling by promoting M2-like macrophages.
Collapse
Affiliation(s)
- Yan Shang
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| | - Yahong Sun
- Department of Respiratory Medicine, Haining People's Hospital of Zhejiang Province, Zhejiang 314400, China
| | - Jing Xu
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, Shanghai 200137, China
| | - Xiahui Ge
- Department of Respiratory Medicine, Seventh People's Hospital of Shanghai University of TCM, Shanghai 200137, China
| | - Zhenli Hu
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Jiang Xiao
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Yunye Ning
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Yuchao Dong
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China
| | - Chong Bai
- Department of Respiratory and Critical Care Medicine, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai 200433, China.
| |
Collapse
|
30
|
Feng Y, Yang C, Yan W. Expression of lncRNA MEG3 in asthma with different phenotypes and its relationship with course of disease. Exp Ther Med 2020; 19:2211-2217. [PMID: 32104286 PMCID: PMC7027329 DOI: 10.3892/etm.2020.8414] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2019] [Accepted: 11/01/2019] [Indexed: 12/16/2022] Open
Abstract
The purpose of this study was to explore the application value of lncRNA MEG3 in lung cancer. From March 2017 to March 2019, 119 asthma patients and 125 healthy people undergoing physical examination in the same period were selected as the research objects. The levels of lncRNA MEG3 in the peripheral blood of the two groups were compared, and the predictive value of MEG3 for asthma as well as the differences in different inflammatory phenotypes were analyzed. The expression of MEG3 was low in asthma patients (P<0.050), the diagnostic sensitivity and specificity for asthma were 79.83 and 66.40%, respectively (P<0.001), it was the lowest in mixed granulocytic asthma (P<0.050) and was negatively correlated with the course of disease (r=−0.666, P<0.001). Logistic regression analysis showed that course of disease, inflammatory phenotype and MEG3 were independent factors affecting recurrence of asthma (P<0.050). MEG3 was low expressed in asthma and had good predictive value for it; in mixed granulocytic asthma, its expression was the lowest and the course of disease was closely related. It might be the key to the diagnosis and treatment of asthma in the future.
Collapse
Affiliation(s)
- Yan Feng
- Department of Pathology, Wuhan No. 1 Hospital, Wuhan, Hubei 430022, P.R. China
| | - Chang Yang
- Department of Respiratory Medicine, Hubei No.3 People's Hospital of Jianghan University, Wuhan, Hubei 430030, P.R. China
| | - Wen Yan
- Department of Pathology, Wuhan No. 1 Hospital, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
31
|
Up-regulation of FOXO1 and reduced inflammation by β-hydroxybutyric acid are essential diet restriction benefits against liver injury. Proc Natl Acad Sci U S A 2019; 116:13533-13542. [PMID: 31196960 PMCID: PMC6613133 DOI: 10.1073/pnas.1820282116] [Citation(s) in RCA: 89] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Liver ischemia and reperfusion injury (IRI) is a major challenge in liver surgery. Diet restriction reduces liver damage by increasing stress resistance; however, the underlying molecular mechanisms remain unclear. We investigated the preventive effect of 12-h fasting on mouse liver IRI. Partial warm hepatic IRI model in wild-type male C57BL/6 mice was used. The control ischemia and reperfusion (IR) group of mice was given food and water ad libitum, while the fasting IR group was given water but not food for 12 h before ischemic insult. In 12-h fasting mice, serum liver-derived enzyme level and tissue damages due to IR were strongly suppressed. Serum β-hydroxybutyric acid (BHB) was significantly raised before ischemia and during reperfusion. Up-regulated BHB induced an increment in the expression of FOXO1 transcription factor by raising the level of acetylated histone. Antioxidative enzyme heme oxigenase 1 (HO-1), a target gene of FOXO1, then increased. Autophagy activity was also enhanced. Serum high-mobility group box 1 was remarkably lowered by the 12-h fasting, and activation of NF-κB and NLRP3 inflammasome was suppressed. Consequently, inflammatory cytokine production and liver injury were reduced. Exogenous BHB administration or histone deacetylase inhibitor administration into the control fed mice ameliorated liver IRI, while FOXO1 inhibitor administration to the 12-h fasting group exacerbated liver IRI. The 12-h fasting exerted beneficial effects on the prevention of liver IRI by increasing BHB, thus up-regulating FOXO1 and HO-1, and by reducing the inflammatory responses and apoptotic cell death via the down-regulation of NF-κB and NLRP3 inflammasome.
Collapse
|
32
|
Roux A, Leroy H, De Muylder B, Bracq L, Oussous S, Dusanter-Fourt I, Chougui G, Tacine R, Randriamampita C, Desjardins D, Le Grand R, Bouillaud F, Benichou S, Margottin-Goguet F, Cheynier R, Bismuth G, Mangeney M. FOXO1 transcription factor plays a key role in T cell-HIV-1 interaction. PLoS Pathog 2019; 15:e1007669. [PMID: 31042779 PMCID: PMC6513100 DOI: 10.1371/journal.ppat.1007669] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 05/13/2019] [Accepted: 02/28/2019] [Indexed: 12/21/2022] Open
Abstract
HIV-1 is dependent on the host cell for providing the metabolic resources for completion of its viral replication cycle. Thus, HIV-1 replicates efficiently only in activated CD4+ T cells. Barriers preventing HIV-1 replication in resting CD4+ T cells include a block that limits reverse transcription and also the lack of activity of several inducible transcription factors, such as NF-κB and NFAT. Because FOXO1 is a master regulator of T cell functions, we studied the effect of its inhibition on T cell/HIV-1 interactions. By using AS1842856, a FOXO1 pharmacologic inhibitor, we observe that FOXO1 inhibition induces a metabolic activation of T cells with a G0/G1 transition in the absence of any stimulatory signal. One parallel outcome of this change is the inhibition of the activity of the HIV restriction factor SAMHD1 and the activation of the NFAT pathway. FOXO1 inhibition by AS1842856 makes resting T cells permissive to HIV-1 infection. In addition, we found that FOXO1 inhibition by either AS1842856 treatment or upon FOXO1 knockdown induces the reactivation of HIV-1 latent proviruses in T cells. We conclude that FOXO1 has a central role in the HIV-1/T cell interaction and that inhibiting FOXO1 with drugs such as AS1842856 may be a new therapeutic shock-and-kill strategy to eliminate the HIV-1 reservoir in human T cells. HIV-1 is controlled by host restriction factors that interfere with its life cycle. However, the virus has equipped itself to counter these strategies. We report a new interplay between HIV-1 and human T lymphocytes through the FOXO1 transcription factor. By using AS1842856, a drug targeting FOXO1, we found that FOXO1 inhibition triggers metabolic activation and G0/G1 transition of resting T cells and also by the inactivation of the SAMHD1 viral restriction factor. FOXO1 inhibition makes resting CD4+ T cells permissive to HIV-1 infection. We finally found that pharmacologic (AS1842856 treatment) or genetic (shRNA) silencing of FOXO1 reactivate HIV-1 latent proviruses. Thus FOXO1 appears as an important player of the HIV-1/T-cell relationship and a new potential therapeutic target for intervention during HIV-1 infection.
Collapse
Affiliation(s)
- Arthur Roux
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Héloise Leroy
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Bénédicte De Muylder
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Lucie Bracq
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
- Institut Pasteur Shangai-Chinese Academy of Sciences, Shangai, China
- International Associated Laboratory (LIA VirHost), CNRS, Université Paris Descartes, Institut Pasteur Paris, and Institut Pasteur Shangai-Chinese Academy of Sciences, Shangai, China
| | - Samia Oussous
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Isabelle Dusanter-Fourt
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Ghina Chougui
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Rachida Tacine
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Clotilde Randriamampita
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Delphine Desjardins
- CEA, Université Paris Sud, INSERM -Immunology of Viral Infections and Autoimmune Diseases department (IMVA), U1184, IDMIT Department, Fontenay-aux-Roses, France
| | - Roger Le Grand
- CEA, Université Paris Sud, INSERM -Immunology of Viral Infections and Autoimmune Diseases department (IMVA), U1184, IDMIT Department, Fontenay-aux-Roses, France
| | - Frederic Bouillaud
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Serge Benichou
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
- Institut Pasteur Shangai-Chinese Academy of Sciences, Shangai, China
- International Associated Laboratory (LIA VirHost), CNRS, Université Paris Descartes, Institut Pasteur Paris, and Institut Pasteur Shangai-Chinese Academy of Sciences, Shangai, China
| | - Florence Margottin-Goguet
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Remi Cheynier
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Georges Bismuth
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
| | - Marianne Mangeney
- NSERM, U1016, Institut Cochin, Paris, France
- CNRS, UMR8104, Paris, France
- Université Paris Descartes, Paris, France
- * E-mail:
| |
Collapse
|
33
|
Kim SH, Das A, Choi HI, Kim KH, Chai JC, Choi MR, Binas B, Park KS, Lee YS, Jung KH, Chai YG. Forkhead box O1 (FOXO1) controls the migratory response of Toll-like receptor (TLR3)-stimulated human mesenchymal stromal cells. J Biol Chem 2019; 294:8424-8437. [PMID: 30944148 DOI: 10.1074/jbc.ra119.008673] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Indexed: 12/13/2022] Open
Abstract
Mesenchymal stromal cells (MSCs) can potently regulate the functions of immune cells and are being investigated for the management of inflammatory diseases. Toll-like receptor 3 (TLR3)-stimulated human MSCs (hMSCs) exhibit increased migration and chemotaxis within and toward damaged tissues. However, the regulatory mechanisms underlying these migratory activities are unclear. Therefore, we analyzed the migration capability and gene expression profiles of TLR3-stimulated hMSCs using RNA-Seq, wound healing, and transwell cell migration assay. Along with increased cell migration, the TLR3 stimulation also increased the expression of cytokines, chemokines, and cell migration-related genes. The promoter regions of the latter showed an enrichment of putative motifs for binding the transcription factors forkhead box O1 (FOXO1), FOXO3, NF-κB (NF-κB1), and RELA proto-oncogene and NF-κB subunit. Of note, FOXO1 inhibition by the FOXO1-selective inhibitor AS1842856 significantly reduced both migration and the expression of migration-related genes. In summary, our results indicate that TLR3 stimulation induces hMSC migration through the expression of FOXO1-activated genes.
Collapse
Affiliation(s)
- Sun Hwa Kim
- Department of Molecular & Life Science, Hanyang University, Seoul 04673, Republic of Korea
| | - Amitabh Das
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588
| | - Hae In Choi
- Department of Bionanotechnology, Hanyang University, Seoul 04673, Republic of Korea
| | - Ki Hoon Kim
- Department of Molecular & Life Science, Hanyang University, Seoul 04673, Republic of Korea
| | - Jin Choul Chai
- Department of Molecular & Life Science, Hanyang University, Seoul 04673, Republic of Korea
| | - Mi Ran Choi
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588
| | - Bert Binas
- Department of Molecular & Life Science, Hanyang University, Seoul 04673, Republic of Korea
| | - Kyoung Sun Park
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588
| | - Young Seek Lee
- Department of Molecular & Life Science, Hanyang University, Seoul 04673, Republic of Korea
| | - Kyoung Hwa Jung
- Institute of Natural Science & Technology, Hanyang University, Ansan 15588.
| | - Young Gyu Chai
- Department of Molecular & Life Science, Hanyang University, Seoul 04673, Republic of Korea; Department of Bionanotechnology, Hanyang University, Seoul 04673, Republic of Korea.
| |
Collapse
|
34
|
Chung S, Kim JY, Song MA, Park GY, Lee YG, Karpurapu M, Englert JA, Ballinger MN, Pabla N, Chung HY, Christman JW. FoxO1 is a critical regulator of M2-like macrophage activation in allergic asthma. Allergy 2019; 74:535-548. [PMID: 30288751 PMCID: PMC6393185 DOI: 10.1111/all.13626] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2018] [Revised: 08/13/2018] [Accepted: 08/25/2018] [Indexed: 12/14/2022]
Abstract
BACKGROUND The pathogenesis of asthma and airway obstruction is the result of an abnormal response to different environmental exposures. The scientific premise of our study was based on the finding that FoxO1 expression is increased in lung macrophages of mice after allergen exposure and human asthmatic patients. Macrophages are capable of switching from one functional phenotype to another, and it is important to understand the mechanisms involved in the transformation of macrophages and how their cellular function affects the peribronchial stromal microenvironment. METHODS We employed a murine asthma model, in which mice were treated by intranasal insufflation with allergens for 2-8 weeks. We used both a pharmacologic approach using a highly specific FoxO1 inhibitor and genetic approaches using FoxO1 knockout mice (FoxO1fl/fl LysMcre). Cytokine level in biological fluids was measured by ELISA and the expression of encoding molecules by NanoString assay and qRT-PCR. RESULTS We show that the levels of FoxO1 gene are significantly elevated in the airway macrophages of patients with mild asthma in response to subsegmental bronchial allergen challenge. Transcription factor FoxO1 regulates a pro-asthmatic phenotype of lung macrophages that is involved in the development and progression of chronic allergic airway disease. We have shown that inhibition of FoxO1 induced phenotypic conversion of lung macrophages and downregulates pro-asthmatic and pro-fibrotic gene expression by macrophages, which contribute to airway inflammation and airway remodeling in allergic asthma. CONCLUSION Targeting FoxO1 with its downstream regulator IRF4 is a novel therapeutic target for controlling allergic inflammation and potentially reversing fibrotic airway remodeling.
Collapse
Affiliation(s)
- Sangwoon Chung
- Pulmonary, Sleep and Critical Care Medicine, The Ohio State University, Wexner Medical Center, Davis Heart
and Lung Research Institute, Columbus, Ohio
| | - Ji Young Kim
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University
| | - Min-Ae Song
- Division of Environmental Health Sciences, College of Public Health, The Ohio State University
| | - Gye Young Park
- Pulmonary, Critical Care, Sleep and Allergy, University of Illinois at Chicago, Chicago, Illinois
| | - Yong Gyu Lee
- Pulmonary, Sleep and Critical Care Medicine, The Ohio State University, Wexner Medical Center, Davis Heart
and Lung Research Institute, Columbus, Ohio
| | - Manjula Karpurapu
- Pulmonary, Sleep and Critical Care Medicine, The Ohio State University, Wexner Medical Center, Davis Heart
and Lung Research Institute, Columbus, Ohio
| | - Joshua A. Englert
- Pulmonary, Sleep and Critical Care Medicine, The Ohio State University, Wexner Medical Center, Davis Heart
and Lung Research Institute, Columbus, Ohio
| | - Megan N. Ballinger
- Pulmonary, Sleep and Critical Care Medicine, The Ohio State University, Wexner Medical Center, Davis Heart
and Lung Research Institute, Columbus, Ohio
| | - Navjot Pabla
- Division of Pharmaceutics, College of Pharmacy, The Ohio State University
| | - Hae Young Chung
- Department of Pharmacy, College of Pharmacy, Pusan National University, Busan, Korea
| | - John W. Christman
- Pulmonary, Sleep and Critical Care Medicine, The Ohio State University, Wexner Medical Center, Davis Heart
and Lung Research Institute, Columbus, Ohio
| |
Collapse
|
35
|
Xi J, Chen Y, Jing J, Zhang Y, Liang C, Hao Z, Zhang L. Sirtuin 3 suppresses the formation of renal calcium oxalate crystals through promoting M2 polarization of macrophages. J Cell Physiol 2018; 234:11463-11473. [PMID: 30588609 DOI: 10.1002/jcp.27803] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Accepted: 11/01/2018] [Indexed: 01/10/2023]
Affiliation(s)
- Junhua Xi
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei Anhui China
- Institute of Urology, Anhui Medical University Hefei Anhui China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University Hefei Anhui China
- Department of Urology Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei) Hefei Anhui China
| | - Yang Chen
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei Anhui China
- Institute of Urology, Anhui Medical University Hefei Anhui China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University Hefei Anhui China
| | - Junfeng Jing
- Department of Urology Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei) Hefei Anhui China
| | - Yanbin Zhang
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei Anhui China
- Institute of Urology, Anhui Medical University Hefei Anhui China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University Hefei Anhui China
- Department of Urology Hefei Hospital Affiliated to Anhui Medical University (The Second People's Hospital of Hefei) Hefei Anhui China
| | - Chaozhao Liang
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei Anhui China
- Institute of Urology, Anhui Medical University Hefei Anhui China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University Hefei Anhui China
| | - Zongyao Hao
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei Anhui China
- Institute of Urology, Anhui Medical University Hefei Anhui China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University Hefei Anhui China
| | - Li Zhang
- Department of Urology The First Affiliated Hospital of Anhui Medical University Hefei Anhui China
- Institute of Urology, Anhui Medical University Hefei Anhui China
- Anhui Province Key Laboratory of Genitourinary Diseases, Anhui Medical University Hefei Anhui China
| |
Collapse
|
36
|
Resolution of chronic inflammatory disease: universal and tissue-specific concepts. Nat Commun 2018; 9:3261. [PMID: 30111884 PMCID: PMC6093916 DOI: 10.1038/s41467-018-05800-6] [Citation(s) in RCA: 269] [Impact Index Per Article: 38.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Accepted: 06/22/2018] [Indexed: 12/21/2022] Open
Abstract
Inflammation and its resolution is under-studied in medicine despite being essential for understanding the development of chronic inflammatory disease. In this review article, we discuss the resolution of inflammation in both a biological and translational context. We introduce the concept of impaired resolution leading to diseases like rheumatoid arthritis, Crohn's disease, and asthma, as well as the cellular and molecular components that contribute to resolution of joint, gut, and lung inflammation, respectively. Finally, we discuss potential intervention strategies for fostering the resolution process, and their implications for the therapy of inflammatory diseases.
Collapse
|
37
|
Legrand JMD, Roy E, Baz B, Mukhopadhyay P, Wong HY, Ram R, Morahan G, Walker G, Khosrotehrani K. Genetic variation in the mitogen-activated protein kinase/extracellular signal-regulated kinase pathway affects contact hypersensitivity responses. J Allergy Clin Immunol 2018; 142:981-984.e7. [PMID: 29753814 DOI: 10.1016/j.jaci.2018.04.028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2017] [Revised: 01/17/2018] [Accepted: 04/20/2018] [Indexed: 12/31/2022]
Affiliation(s)
- Julien M D Legrand
- UQ Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Australia
| | - Edwige Roy
- UQ Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Australia
| | - Batoul Baz
- UQ Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Australia
| | | | - Ho Yi Wong
- UQ Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Australia
| | - Ramesh Ram
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Perth, Australia
| | - Grant Morahan
- Centre for Diabetes Research, Harry Perkins Institute of Medical Research, Perth, Australia
| | - Graeme Walker
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Kiarash Khosrotehrani
- UQ Diamantina Institute, Translational Research Institute, University of Queensland, Brisbane, Australia.
| |
Collapse
|
38
|
Pyrzynska B, Dwojak M, Zerrouqi A, Morlino G, Zapala P, Miazek N, Zagozdzon A, Bojarczuk K, Bobrowicz M, Siernicka M, Machnicki MM, Gobessi S, Barankiewicz J, Lech-Maranda E, Efremov DG, Juszczynski P, Calado D, Golab J, Winiarska M. FOXO1 promotes resistance of non-Hodgkin lymphomas to anti-CD20-based therapy. Oncoimmunology 2018; 7:e1423183. [PMID: 29721381 PMCID: PMC5927521 DOI: 10.1080/2162402x.2017.1423183] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Revised: 12/23/2017] [Accepted: 12/28/2017] [Indexed: 12/16/2022] Open
Abstract
Diminished overall survival rate of non-Hodgkin lymphoma (NHL) patients treated with a combination regimen of rituximab, cyclophosphamide, doxorubicin, vincristine and prednisone (R-CHOP) has been recently linked to recurrent somatic mutations activating FOXO1. Despite of the clinical relevance of this finding, the molecular mechanism driving resistance to R-CHOP therapy remains largely unknown. Herein, we investigated the potential role of FOXO1 in the therapeutic efficacy of rituximab, the only targeted therapy included in the R-CHOP regimen. We found CD20 transcription is negatively regulated by FOXO1 in NHL cell lines and in human lymphoma specimens carrying activating mutations of FOXO1. Furthermore, both the expression of exogenous mutants of FOXO1 and the inhibition of AKT led to FOXO1 activation in lymphoma cells, increased binding to MS4A1 promoter and diminished CD20 expression levels. In contrast, a disruption of FOXO1 with CRISPR/Cas9 genome-editing (sgFOXO1) resulted in CD20 upregulation, improved the cytotoxicity induced by rituximab and the survival of mice with sgFOXO1 tumors. Accordingly, pharmacological inhibition of FOXO1 activity in primary samples upregulated surface CD20 levels. Importantly, FOXO1 was required for the downregulation of CD20 levels by the clinically tested inhibitors of BTK, SYK, PI3K and AKT. Taken together, these results indicate for the first time that the AKT-unresponsive mutants of FOXO1 are important determinant of cell response to rituximab-induced cytotoxicity, and suggest that the genetic status of FOXO1 together with its transcriptional activity need further attention while designing anti-CD20 antibodies based regimens for the therapy of pre-selected lymphomas.
Collapse
Affiliation(s)
- Beata Pyrzynska
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Michal Dwojak
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Warsaw, Poland
| | | | | | - Piotr Zapala
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | - Nina Miazek
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | | | - Kamil Bojarczuk
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
| | | | - Marta Siernicka
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Warsaw, Poland
| | - Marcin M. Machnicki
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Warsaw, Poland
| | - Stefania Gobessi
- Department of Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Joanna Barankiewicz
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- Department of Hematology and Transfusion Medicine, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Ewa Lech-Maranda
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
- Department of Hematology and Transfusion Medicine, Center of Postgraduate Medical Education, Warsaw, Poland
| | - Dimitar G. Efremov
- Department of Molecular Hematology, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy
| | - Przemyslaw Juszczynski
- Department of Hematology, Institute of Hematology and Transfusion Medicine, Warsaw, Poland
| | - Dinis Calado
- The Francis Crick Institute, London, United Kingdom
- Peter Gorer Department of Immunobiology, Kings College London, United Kingdom
| | - Jakub Golab
- Department of Immunology, Medical University of Warsaw, Warsaw, Poland
- Centre for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | | |
Collapse
|
39
|
Buttrick TS, Wang W, Yung C, Trieu KG, Patel K, Khoury SJ, Ai X, Elyaman W. Foxo1 Promotes Th9 Cell Differentiation and Airway Allergy. Sci Rep 2018; 8:818. [PMID: 29339772 PMCID: PMC5770389 DOI: 10.1038/s41598-018-19315-z] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2017] [Accepted: 12/21/2017] [Indexed: 12/31/2022] Open
Abstract
T helper 9 (Th9) cells are effector CD4+ T cells that are characterized by the production of interleukin-9 (IL-9) and have been associated with allergic responses. Here, we found that the expression of the transcription factor forkhead box O1 (Foxo1) was induced in Th9 and Foxo1 plays a crucial role in the differentiation of Th9 cells. Pharmacological inhibition of Foxo1 or genetic disruption of Foxo1 in CD4+ T cells caused a reduction in IL-9 expression while upregulating IL-17A and IFNγ production. Furthermore, chromatin immunoprecipitation (ChIP) followed by luciferase assays revealed direct binding of Foxo1 to both the Il9 and Irf4 promoters and induces their transactivation. Lastly, adoptive transfer of Th9 cells into lungs induced asthma-like symptoms that were ameliorated by Foxo1 inhibitor, AS1842856. Together, our findings demonstrate a novel regulator of Th9 cells with a direct implication in allergic inflammation.
Collapse
Affiliation(s)
- Thomas S Buttrick
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Wei Wang
- Pulmonary and Critical Care, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Christina Yung
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, 10032, USA
| | - Kenneth G Trieu
- Pulmonary and Critical Care, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Kruti Patel
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Samia J Khoury
- Ann Romney Center for Neurologic Diseases, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA.,Abu Haidar Neuroscience Institute, American University of Beirut Medical Center, Beirut, Lebanon
| | - Xingbin Ai
- Pulmonary and Critical Care, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, 02115, USA
| | - Wassim Elyaman
- Center for Translational and Computational Neuroimmunology, Columbia University Medical Center, New York, NY, 10032, USA.
| |
Collapse
|
40
|
FoxO1 is a regulator of MHC-II expression and anti-tumor effect of tumor-associated macrophages. Oncogene 2017; 37:1192-1204. [PMID: 29238041 DOI: 10.1038/s41388-017-0048-4] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Revised: 09/27/2017] [Accepted: 10/09/2017] [Indexed: 01/19/2023]
Abstract
Macrophages are a critical component in host immune responses against tumor. In this work we investigated the role of forkhead box O1 (FoxO1) in the transcriptional regulation in macrophages, which affects the anti-tumor functions of tumor-associated macrophages (TAMs). First, we showed that TAMs expressed reduced levels of FoxO1, which was associated with their protumoral M2 polarization state. The suppression of FoxO1 expression in TAM was induced by the hypoxic condition in the tumor microenviroment. Next, we confirmed that FoxO1 positively regulates MHC-II genes by binding to the promoter region of Ciita gene, the master activator of multiple MHC-II genes. Loss of FoxO1 in TAMs resulted in reduced MHC-II expression. Furthermore, we used FoxO1 conditional knockout mice to show that FoxO1 deficiency in myeloid cells exacerbates tumor growth. These results demonstrate that the protumoral property of TAMs is induced by the hypoxia-triggered FoxO1 deficiency, which could be a potential target of novel anti-tumor therapies.
Collapse
|
41
|
Malik S, Sadhu S, Elesela S, Pandey RP, Chawla AS, Sharma D, Panda L, Rathore D, Ghosh B, Ahuja V, Awasthi A. Transcription factor Foxo1 is essential for IL-9 induction in T helper cells. Nat Commun 2017; 8:815. [PMID: 28993609 PMCID: PMC5634439 DOI: 10.1038/s41467-017-00674-6] [Citation(s) in RCA: 84] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2017] [Accepted: 07/16/2017] [Indexed: 11/09/2022] Open
Abstract
Interleukin 9 (IL-9)-producing helper T (Th9) cells have a crucial function in allergic inflammation, autoimmunity, immunity to extracellular pathogens and anti-tumor immune responses. In addition to Th9, Th2, Th17 and Foxp3+ regulatory T (Treg) cells produce IL-9. A transcription factor that is critical for IL-9 induction in Th2, Th9 and Th17 cells has not been identified. Here we show that the forkhead family transcription factor Foxo1 is required for IL-9 induction in Th9 and Th17 cells. We further show that inhibition of AKT enhances IL-9 induction in Th9 cells while it reciprocally regulates IL-9 and IL-17 in Th17 cells via Foxo1. Mechanistically, Foxo1 binds and transactivates IL-9 and IRF4 promoters in Th9, Th17 and iTreg cells. Furthermore, loss of Foxo1 attenuates IL-9 in mouse and human Th9 and Th17 cells, and ameliorates allergic inflammation in asthma. Our findings thus identify that Foxo1 is essential for IL-9 induction in Th9 and Th17 cells.The transcription factor Foxo1 can control regulatory T cell and Th1 function. Here the authors show that Foxo1 is also critical for IL-9 production by Th9 cells and other IL-9-producing cells.
Collapse
Affiliation(s)
- Sakshi Malik
- Center for Human Microbial Ecology, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana, 121 001, India
| | - Srikanth Sadhu
- Center for Human Microbial Ecology, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana, 121 001, India
| | - Srikanth Elesela
- Center for Human Microbial Ecology, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana, 121 001, India
| | - Ramendra Pati Pandey
- Center for Human Microbial Ecology, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana, 121 001, India
| | | | - Deepak Sharma
- Department of Biotechnology, Indian Institute of Technology Roorkee, Roorkee, 247667, India
| | - Lipsa Panda
- Institute of Genomics and Integrative Biology (IGIB), Mall Road, New Delhi, 110007, India
| | - Deepak Rathore
- Center for Human Microbial Ecology, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana, 121 001, India
| | - Balram Ghosh
- Institute of Genomics and Integrative Biology (IGIB), Mall Road, New Delhi, 110007, India
| | - Vineet Ahuja
- Department of Gastroenterology, All India Institute of Medical Sciences (AIIMS), Ansari Nagar, New Delhi, 110029, India
| | - Amit Awasthi
- Center for Human Microbial Ecology, Translational Health Science & Technology Institute, NCR Biotech Science Cluster, 3rd Milestone Gurgaon-Faridabad Expressway, Faridabad, Haryana, 121 001, India.
| |
Collapse
|
42
|
Davis JS, Sun M, Kho AT, Moore KG, Sylvia JM, Weiss ST, Lu Q, Tantisira KG. Circulating microRNAs and association with methacholine PC20 in the Childhood Asthma Management Program (CAMP) cohort. PLoS One 2017; 12:e0180329. [PMID: 28749975 PMCID: PMC5531511 DOI: 10.1371/journal.pone.0180329] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2017] [Accepted: 06/08/2017] [Indexed: 11/18/2022] Open
Abstract
Introduction Circulating microRNAs (miRNA) are promising biomarkers for human diseases. Our study hypothesizes that circulating miRNA would reveal candidate biomarkers related to airway hyperresponsiveness (AHR) and provide biologic insights into asthma epigenetic influences. Methods Serum samples obtained at randomization for 160 children in the Childhood Asthma Management Program were profiled using a TaqMan miRNA array set. The association of the isolated miRNA with methacholine PC20 was assessed. Network and pathway analyses were performed. Functional validation of two significant miRNAs was performed in human airway smooth muscle cells (HASMs). Results Of 155 well-detected circulating miRNAs, eight were significantly associated with PC20 with the strongest association with miR-296-5p. Pathway analysis revealed miR-16-5p as a network hub, and involvement of multiple miRNAs interacting with genes in the FoxO and Hippo signaling pathways by KEGG analysis. Functional validation of two miRNA in HASM showed effects on cell growth and diameter. Conclusion Reduced circulatory miRNA expression at baseline is associated with an increase in PC20. These miRNA provide biologic insights into, and may serve as biomarkers of, asthma severity. miR-16-5p and -30d-5p regulate airway smooth muscle phenotypes critically involved in asthma pathogenesis, supporting a mechanistic link to these findings. Functional ASM phenotypes may be directly relevant to AHR.
Collapse
Affiliation(s)
- Joshua S. Davis
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Maoyun Sun
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Alvin T. Kho
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Computational Health Informatics Program, Boston Children’s Hospital, Boston, Massachusetts, United States of America
| | - Kip G. Moore
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Jody M. Sylvia
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Scott T. Weiss
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Quan Lu
- Molecular and Integrative Physiological Sciences Program, Department of Environmental Health, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Kelan G. Tantisira
- Channing Division of Network Medicine, Brigham and Women’s Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- * E-mail:
| |
Collapse
|
43
|
Draijer C, Peters-Golden M. Alveolar Macrophages in Allergic Asthma: the Forgotten Cell Awakes. Curr Allergy Asthma Rep 2017; 17:12. [PMID: 28233154 DOI: 10.1007/s11882-017-0681-6] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW The role of alveolar macrophages in innate immune responses has long been appreciated. Here, we review recent studies evaluating the participation of these cells in allergic inflammation. RECENT FINDINGS Immediately after allergen exposure, monocytes are rapidly recruited from the bloodstream and serve to promote acute inflammation. By contrast, resident alveolar macrophages play a predominantly suppressive role in an effort to restore homeostasis. As inflammation becomes established after repeated exposures, alveolar macrophages can polarize across a continuum of activation phenotypes, losing their suppressive functions and gaining pathogenic functions. Future research should focus on the diverse roles of monocytes/macrophages during various types and phases of allergic inflammation. These properties could lead us to new therapeutic opportunities.
Collapse
Affiliation(s)
- Christina Draijer
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA
| | - Marc Peters-Golden
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, University of Michigan Medical School, 6301 MSRB III, 1150 W. Medical Center Drive, Ann Arbor, MI, 48109-5642, USA.
| |
Collapse
|
44
|
Sundaresan S, Puthanveetil P. Is FoxO1 the culprit, partner in crime, or a protector in systemic inflammation? Am J Physiol Cell Physiol 2017. [PMID: 28637677 DOI: 10.1152/ajpcell.00194.2016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Sinju Sundaresan
- Division of Gastroenterology, Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan; and
| | - Prasanth Puthanveetil
- Department of Biopharmaceutical Sciences, College of Pharmacy, Roosevelt University, Schaumburg, Illinois
| |
Collapse
|
45
|
Thomsen LN, Thomsen PD, Downing A, Talbot R, Berg LC. FOXO1, PXK, PYCARD and SAMD9L are differentially expressed by fibroblast-like cells in equine synovial membrane compared to joint capsule. BMC Vet Res 2017; 13:106. [PMID: 28410619 PMCID: PMC5391632 DOI: 10.1186/s12917-017-1003-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Accepted: 03/28/2017] [Indexed: 11/11/2022] Open
Abstract
Background The synovial membrane lines the luminal side of the joint capsule in synovial joints. It maintains joint homeostasis and plays a crucial role in equine joint pathology. When trauma or inflammation is induced in a joint, the synovial membrane influences progression of joint damage. Equine synovial membrane research is hampered by a lack of markers of fibroblast-like synoviocytes (FLS) to distinguish FLS from other fibroblast-like cells in musculoskeletal connective tissues. The aim of this study is to identify potential FLS markers of the equine synovial membrane using microarray to compare between gene expression in equine synovial membrane and the joint capsule in metacarpophalangeal joints. Results Microarray analysis of tissues from 6 horses resulted in 1167 up-regulated genes in synovial membrane compared with joint capsule. Pathway analysis resulted in 241 candidate genes. Of these, 15 genes were selected for further confirmation as genes potentially expressed by fibroblast-like synoviocytes. Four genes: FOXO1, PXK, PYCARD and SAMD9L were confirmed in 9 horses by qPCR as differentially expressed in synovial membrane compared to joint capsule. Conclusions In conclusion, FOXO1, PXK, PYCARD and SAMD9L were confirmed as differentially expressed in synovial membrane compared to joint capsule. These four genes are potential markers of fibroblast-like synoviocytes of the synovial membrane. As these genes are overexpressed in synovial membrane compared to joint capsule, these genes could shed light on synovial membrane physiology and its role in joint disease.
Collapse
Affiliation(s)
- Line Nymann Thomsen
- Department of Veterinary Clinical Sciences, University of Copenhagen, Hoejbakkegaards alle 5, 2630, Taastrup, Denmark
| | - Preben Dybdahl Thomsen
- Department of Veterinary and Animal Sciences, University of Copenhagen, Groennegaardsvej 7, 1870, Frederiksberg C, Denmark
| | - Alison Downing
- Edinburgh Genomics, Ashworth Laboratories, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Richard Talbot
- Edinburgh Genomics, Ashworth Laboratories, University of Edinburgh, Edinburgh, EH9 3FL, UK
| | - Lise Charlotte Berg
- Department of Veterinary Clinical Sciences, University of Copenhagen, Hoejbakkegaards alle 5, 2630, Taastrup, Denmark.
| |
Collapse
|
46
|
Wang Y, Zhu J, Zhang L, Zhang Z, He L, Mou Y, Deng Y, Cao Y, Yang P, Su Y, Zhao J, Zhang S, Yu Q, Hu J, Chen Z, Ning Q, Xiang X, Xu Y, Wang CY, Xiong W. Role of C/EBP homologous protein and endoplasmic reticulum stress in asthma exacerbation by regulating the IL-4/signal transducer and activator of transcription 6/transcription factor EC/IL-4 receptor α positive feedback loop in M2 macrophages. J Allergy Clin Immunol 2017; 140:1550-1561.e8. [PMID: 28238747 DOI: 10.1016/j.jaci.2017.01.024] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 12/30/2016] [Accepted: 01/23/2017] [Indexed: 12/16/2022]
Abstract
BACKGROUND C/EBP homologous protein (Chop), a marker of endoplasmic reticulum (ER) stress, exhibits aberrant expression patterns during asthma development. However, its exact role in asthma pathogenesis is not fully understood. OBJECTIVES We aimed to determine the function and mechanism of Chop in the pathogenesis of allergic asthma in patients and animals. METHODS Studies were conducted in asthmatic patients and Chop-/- mice to dissect the role of Chop and ER stress in asthma pathogenesis. An ovalbumin (OVA)-induced allergic airway inflammation model was used to address the effect of Chop deficiency on asthma development. Next, the effect of Chop deficiency on macrophage polarization and related signaling pathways was investigated to demonstrate the underlying mechanisms. RESULTS Asthmatic patients and mice after OVA induction exhibited aberrant Chop expression along with ER stress. Specifically, Chop was noted to be specifically overexpressed in macrophages, and mice deficient in Chop were protected from OVA-induced allergic airway inflammation, as manifested by attenuated airway inflammation, remodeling, and hyperresponsiveness. Chop was found to exacerbate allergic airway inflammation by enhancing M2 programming in macrophages. Mechanistic studies characterized an IL-4/signal transducer and activator of transcription 6/transcription factor EC (Tfec)/IL-4 receptor α positive feedback regulatory loop, in which IL-4 induces Chop expression, which then promotes signal transducer and activator of transcription 6 signaling to transcribe Tfec expression. Finally, Tfec transcribes IL-4 receptor α expression to promote M2 programming in macrophages. CONCLUSIONS Chop and ER stress are implicated in asthma pathogenesis, which involves regulation of M2 programming in macrophages.
Collapse
Affiliation(s)
- Yi Wang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Jianghui Zhu
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Lei Zhang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Zhijun Zhang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Long He
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Yong Mou
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yanhan Deng
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Yong Cao
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Ping Yang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Yunchao Su
- Department of Pharmacology and Toxicology, Medical College of Georgia, Georgia Regents University, Augusta, Ga
| | - Jianping Zhao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Shu Zhang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Qilin Yu
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China
| | - Jifa Hu
- Department of Sponsored Program Administration, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Zhishui Chen
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Sponsored Program Administration, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Qin Ning
- Department of Infectious Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Xudong Xiang
- Department of Emergency Medicine, Institute of Emergency Medicine and Rare Diseases, Second Xiangya Hospital, Central South University, Changsha, China
| | - Yongjian Xu
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China
| | - Cong-Yi Wang
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Sponsored Program Administration, Tongji Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China.
| | - Weining Xiong
- Center for Biomedical Research, Key Laboratory of Organ Transplantation, Ministry of Education, Key Laboratory of Organ Transplantation, Ministry of Health, Wuhan, China; Department of Respiratory and Critical Care Medicine, Key Laboratory of Pulmonary Diseases of Health Ministry, Key Cite of National Clinical Research Center for Respiratory Disease, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences & Technology, Wuhan, China.
| |
Collapse
|
47
|
Mamun AA, Liu F. Role of IRF4-Mediated Inflammation: Implication in Neurodegenerative Diseases. NEUROLOGY & NEUROTHERAPY OPEN ACCESS JOURNAL 2017; 2:107. [PMID: 39473489 PMCID: PMC11521387 DOI: 10.23880/nnoaj-16000107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Neuro-inflammation is a common feature of various central nervous system (CNS) disorders, including stroke, Alzheimer's disease, Multiple sclerosis, etc., and has a significant impact on the outcomes. Regulation of the immune response has therapeutic value. Interferon regulatory factor 4 (IRF4) is a hemopoietic transcription factor critical for activation of microglia/macrophages and modulation of inflammatory responses. The effects of IRF4 signaling on inflammation are pleiotropic, and vary depending on immune cell types and the pathological microenvironment that is regulated by both pro- and anti-inflammatory cytokines. Mechanistically, IRF4 is a quintessential 'context-dependent' transcription factor that regulates distinct groups of inflammatory mediators in a differential manner depending on their activation in different cell types including phagocytes, T-cell subtypes, and neuronal cells. In this review, we summarized the recent findings of IRF4 in the context of immune responses in different cell types with diverse pathological conditions. The primary goal of this review is to understand the signaling pathways and beneficial functions of IRF4, in hope of developing effective therapeutic strategies targeting the immune responses to neurodegenerative diseases.
Collapse
Affiliation(s)
- AA Mamun
- Department of Neurology, McGovern Medical School, The University of Texas Medical School, USA
| | - F Liu
- Department of Neurology, McGovern Medical School, The University of Texas Medical School, USA
| |
Collapse
|
48
|
Man K, Kutyavin VI, Chawla A. Tissue Immunometabolism: Development, Physiology, and Pathobiology. Cell Metab 2017; 25:11-26. [PMID: 27693378 PMCID: PMC5226870 DOI: 10.1016/j.cmet.2016.08.016] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2016] [Revised: 08/15/2016] [Accepted: 08/24/2016] [Indexed: 02/07/2023]
Abstract
Evolution of metazoans resulted in the specialization of cellular and tissue function. This was accomplished by division of labor, which allowed tissue parenchymal cells to prioritize their core functions while ancillary functions were delegated to tissue accessory cells, such as immune, stromal, and endothelial cells. In metabolic organs, the accessory cells communicate with their clients, the tissue parenchymal cells, to optimize cellular processes, allowing organisms to adapt to changes in their environment. Here, we discuss tissue immunometabolism from this vantage point and use examples from adipose tissues (white, beige, and brown) and liver to outline the general principles by which accessory cells support metabolic homeostasis in parenchymal cells. A corollary of this model is that disruption of communication between client and accessory cells might predispose metabolic organs to the development of disease.
Collapse
Affiliation(s)
- Kevin Man
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Vassily I Kutyavin
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA
| | - Ajay Chawla
- Cardiovascular Research Institute, University of California, San Francisco, CA 94143-0795, USA; Departments of Physiology and Medicine, University of California, San Francisco, CA 94143-0795, USA.
| |
Collapse
|
49
|
Lanza GM, Jenkins J, Schmieder AH, Moldobaeva A, Cui G, Zhang H, Yang X, Zhong Q, Keupp J, Sergin I, Paranandi KS, Eldridge L, Allen JS, Williams T, Scott MJ, Razani B, Wagner EM. Anti-angiogenic Nanotherapy Inhibits Airway Remodeling and Hyper-responsiveness of Dust Mite Triggered Asthma in the Brown Norway Rat. Am J Cancer Res 2017; 7:377-389. [PMID: 28042341 PMCID: PMC5197071 DOI: 10.7150/thno.16627] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2016] [Accepted: 10/04/2016] [Indexed: 12/16/2022] Open
Abstract
Although angiogenesis is a hallmark feature of asthmatic inflammatory responses, therapeutic anti-angiogenesis interventions have received little attention. Objective: Assess the effectiveness of anti-angiogenic Sn2 lipase-labile prodrugs delivered via αvβ3-micellar nanotherapy to suppress microvascular expansion, bronchial remodeling, and airway hyper-responsiveness in Brown Norway rats exposed to serial house dust mite (HDM) inhalation challenges. Results: Anti-neovascular effectiveness of αvβ3-mixed micelles incorporating docetaxel-prodrug (Dxtl-PD) or fumagillin-prodrug (Fum-PD) were shown to robustly suppress neovascular expansion (p<0.01) in the upper airways/bronchi of HDM rats using simultaneous 19F/1H MR neovascular imaging, which was corroborated by adjunctive fluorescent microscopy. Micelles without a drug payload (αvβ3-No-Drug) served as a carrier-only control. Morphometric measurements of HDM rat airway size (perimeter) and vessel number at 21d revealed classic vascular expansion in control rats but less vascularity (p<0.001) after the anti-angiogenic nanotherapies. CD31 RNA expression independently corroborated the decrease in airway microvasculature. Methacholine (MCh) induced respiratory system resistance (Rrs) was high in the HDM rats receiving αvβ3-No-Drug micelles while αvβ3-Dxtl-PD or αvβ3-Fum-PD micelles markedly and equivalently attenuated airway hyper-responsiveness and improved airway compliance. Total inflammatory BAL cells among HDM challenged rats did not differ with treatment, but αvβ3+ macrophages/monocytes were significantly reduced by both nanotherapies (p<0.001), most notably by the αvβ3-Dxtl-PD micelles. Additionally, αvβ3-Dxtl-PD decreased BAL eosinophil and αvβ3+ CD45+ leukocytes relative to αvβ3-No-Drug micelles, whereas αvβ3-Fum-PD micelles did not. Conclusion: These results demonstrate the potential of targeted anti-angiogenesis nanotherapy to ameliorate the inflammatory hallmarks of asthma in a clinically relevant rodent model.
Collapse
|