1
|
Jeon WY, Jin SE, Sohn E, Jo K, Ha H, Shin HK, Lee MY. Anti-inflammatory and anti-allergic effects of Cheonwangbosim-dan water extract: An in vitro and in vivo study. Heliyon 2023; 9:e16172. [PMID: 37215857 PMCID: PMC10199208 DOI: 10.1016/j.heliyon.2023.e16172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 04/23/2023] [Accepted: 05/08/2023] [Indexed: 05/24/2023] Open
Abstract
Ethnopharmacological relevance Cheonwangbosim-dan is a traditional herbal prescription that is widely used to improve or treat physical and mental illnesses in East Asian countries.Aim of the study: The aim of the present study was to investigate the preventive and protective effects of a Cheonwangbosim-dan water extract (CBDW) against allergic inflammation using in vitro and in vivo models. Materials and methods BEAS-2B and MC/9 cells were treated with various concentrations of CBDW and stimulated with different inducers of inflammatory mediators. The production of various inflammatory mediators was subsequently evaluated. BALB/c mice were sensitized and challenged by repeated application of ovalbumin (OVA). CBDW was administered by oral gavage once daily for 10 consecutive days. We assessed the number of inflammatory cells and production of Th2 cytokines in bronchoalveolar lavage fluid (BALF), the plasma levels of total and OVA-specific immunoglobulin E (IgE), and histological changes in lung tissue. Results Our findings showed that CBDW significantly decreased the levels of various inflammatory mediators (eotaxin-1, eotaxin-3, RANTES, LTC4, TNF-α, MMP-9, 5-LO, ICAM-1, and VCAM-1) in vitro, significantly reduced the accumulation of total inflammatory cells, the production of Th2 cytokines (IL-5 and IL-13), the levels of IgE (total and OVA-specific) in vivo, and remarkably inhibited histological changes (infiltration of inflammatory cells and goblet cell hyperplasia) in vivo. Conclusions These results suggest that CBDW possesses anti-inflammatory and anti-allergic properties by lowering allergic inflammation.
Collapse
Affiliation(s)
- Woo-Young Jeon
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Seong Eun Jin
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Eunjin Sohn
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Kyuhyung Jo
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Hyekyung Ha
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Hyeun-Kyoo Shin
- KM Science Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| | - Mee-Young Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine, 1672 Yuseong-daero, Yuseong-gu, Daejeon, 34054, Republic of Korea
| |
Collapse
|
2
|
Farmanzadeh A, Qujeq D, Yousefi T. The Interaction Network of MicroRNAs with Cytokines and Signaling Pathways in Allergic Asthma. Microrna 2022; 11:104-117. [PMID: 35507792 DOI: 10.2174/2211536611666220428134324] [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] [Received: 10/11/2021] [Revised: 01/15/2022] [Accepted: 03/10/2022] [Indexed: 01/01/2023]
Abstract
Allergic asthma is a complicated disease that is affected by many factors. Numerous cytokines and signaling pathways are attributed to the cause of asthma symptoms. MicroRNAs (miRNAs) are a group of small non-coding single-stranded RNA molecules that are involved in gene silencing and posttranscriptional regulation of gene expression by targeting mRNAs. In pathological conditions, altered expression of microRNAs differentially regulates cytokines and signaling pathways and therefore, can be the underlying reason for the pathogenesis of allergic asthma. Indeed, microRNAs participate in airway inflammation via inducing airway structural cells and activating immune responses by targeting cytokines and signaling pathways. Thus, to make a complete understanding of allergic asthma, it is necessary to investigate the communication network of microRNAs with cytokines and signaling pathways which is contributed to the pathogenesis of allergic asthma. Here, we shed light on this aspect of asthma pathology by Summarizing our current knowledge of this topic.
Collapse
Affiliation(s)
- Ali Farmanzadeh
- Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran.,Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| | - Tooba Yousefi
- Department of Clinical Biochemistry, Faculty of Medicine, Babol University of Medical Sciences, Babol, Iran
| |
Collapse
|
3
|
MicroRNA-629-3p Promotes Interleukin-13-Induced Bronchial Epithelial Cell Injury and Inflammation by Targeting FOXA2. Cell Biochem Biophys 2022; 80:457-466. [DOI: 10.1007/s12013-022-01072-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 02/14/2022] [Indexed: 11/03/2022]
|
4
|
Kawakami K, Miyasaka T, Nakamura Y, Metoki H, Miyata S, Sato M, Sora I, Yamauchi K, Kawakami K, Blendy JA, Kawano T, Shimokawa H, Takayanagi M, Ohno I, Takahashi T. The A118G single-nucleotide polymorphism in OPRM1 is a risk factor for asthma severity. Allergol Int 2022; 71:55-65. [PMID: 34688555 DOI: 10.1016/j.alit.2021.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 07/16/2021] [Accepted: 07/20/2021] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Although population studies have implicated emotional burden in asthma severity, the underlying genetic risk factors are not completely understood. We aimed to evaluate the genetic influence of a functional single-nucleotide polymorphism (SNP) in the stress-related μ-opioid receptor gene (OPRM1; A118G SNP, rs1799971) on asthma severity. METHODS We initially assessed disease severity in asthmatic outpatients carrying A118G. Using an ovalbumin-induced experimental asthma rodent model harboring the functionally equivalent SNP, we investigated the mechanism by which this SNP influences the allergic immune response. RESULTS Among 292 outpatients, 168 underwent airway hyperresponsiveness (AHR) to methacholine testing. Compared with patients carrying the AA and AG genotypes, those carrying the GG genotype exhibited enhanced AHR. The stress levels were presumed to be moderate among patients and were comparable among genotypes. Compared with Oprm1 AA mice, GG mice demonstrated aggravated asthma-related features and increased pulmonary interleukin-4+CD4+ effector and effector memory T cells under everyday life stress conditions. Intraperitoneal naloxone methiodide injection reduced effector CD4+ T cell elevation associated with increased eosinophil numbers in bronchoalveolar lavage fluid of GG mice to the levels in AA mice, suggesting that elevated Th2 cell generation in the bronchial lymph node (BLN) of GG mice induces enhanced eosinophilic inflammation. CONCLUSIONS Without forced stress exposure, patients with asthma carrying the OPRM1 GG genotype exhibit enhanced AHR, attributable to enhanced Th2 cell differentiation in the regional lymph node. Further research is necessary to elucidate the role of the OPRM1 A118G genotype in the Th2 cell differentiation pathway in the BLN.
Collapse
Affiliation(s)
- Kaori Kawakami
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomomitsu Miyasaka
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan.
| | - Yutaka Nakamura
- Division of Pulmonary Medicine, Allergy, and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Hirohito Metoki
- Division of Public Health, Hygiene and Epidemiology, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Satoshi Miyata
- Teikyo University Graduate School of Public Health, Tokyo, Japan
| | - Miki Sato
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Ichiro Sora
- Department of Psychiatry, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Kohei Yamauchi
- Division of Pulmonary Medicine, Allergy, and Rheumatology, Department of Internal Medicine, Iwate Medical University School of Medicine, Iwate, Japan
| | - Kazuyoshi Kawakami
- Department of Medical Microbiology, Mycology and Immunology, Tohoku University Graduate School of Medicine, Sendai, Japan; Department of Intelligent Network for Infection Control, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Julie A Blendy
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania Medical School, Philadelphia, PA, USA
| | - Tasuku Kawano
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Hiroaki Shimokawa
- Department of Cardiovascular Medicine, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Motoaki Takayanagi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Isao Ohno
- Center for Medical Education, Faculty of Medicine, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Tomoko Takahashi
- Division of Pathophysiology, Department of Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| |
Collapse
|
5
|
Johansson K, Woodruff PG, Ansel KM. Regulation of airway immunity by epithelial miRNAs. Immunol Rev 2021; 304:141-153. [PMID: 34549450 PMCID: PMC9135676 DOI: 10.1111/imr.13028] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 02/07/2023]
Abstract
The airway epithelium is essential to protect the host from inhaled pathogens and particles. It maintains immune homeostasis and mediates tissue repair after injury. Inflammatory diseases of the airways are associated with failure of epithelial functions, including loss of barrier integrity that results in increased tissue permeability and immune activation; excessive mucus secretion and impaired mucociliary clearance that leads to airflow obstruction and microbial overgrowth; and dysregulation of cellular signals that promotes inflammation and alters tissue structure and airway reactivity. MicroRNAs play crucial roles in mounting appropriate cellular responses to environmental stimuli and preventing disease, using a common machinery and mechanism to regulate gene expression in epithelial cells, immune cells of hematopoietic origin, and other cellular components of the airways. Respiratory diseases are accompanied by dramatic changes in epithelial miRNA expression that drive persistent immune dysregulation. In this review, we discuss responses of the epithelium that promote airway immunopathology, with a focus on miRNAs that contribute to the breakdown of essential epithelial functions. We emphasize the emerging role of miRNAs in regulation of epithelial responses in respiratory health and their value as diagnostic and therapeutic targets.
Collapse
Affiliation(s)
- Kristina Johansson
- Department of Medical Biochemistry, University of Gothenburg, Gothenburg, Sweden
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| | - Prescott G. Woodruff
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Medicine, Division of Pulmonary, Critical Care, Sleep and Allergy, University of California, San Francisco, California, USA
- Cardiovascular Research Institute, University of California, San Francisco, California, USA
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center, University of California, San Francisco, California, USA
- Department of Microbiology and Immunology, University of California, San Francisco, California, USA
| |
Collapse
|
6
|
Weidner J, Bartel S, Kılıç A, Zissler UM, Renz H, Schwarze J, Schmidt‐Weber CB, Maes T, Rebane A, Krauss‐Etschmann S, Rådinger M. Spotlight on microRNAs in allergy and asthma. Allergy 2021; 76:1661-1678. [PMID: 33128813 PMCID: PMC8246745 DOI: 10.1111/all.14646] [Citation(s) in RCA: 112] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 10/16/2020] [Accepted: 10/25/2020] [Indexed: 12/14/2022]
Abstract
In past 10 years, microRNAs (miRNAs) have gained scientific attention due to their importance in the pathophysiology of allergic diseases and their potential as biomarkers in liquid biopsies. They act as master post‐transcriptional regulators that control most cellular processes. As one miRNA can target several mRNAs, often within the same pathway, dysregulated expression of miRNAs may alter particular cellular responses and contribute, or lead, to the development of various diseases. In this review, we give an overview of the current research on miRNAs in allergic diseases, including atopic dermatitis, allergic rhinitis, and asthma. Specifically, we discuss how individual miRNAs function in the regulation of immune responses in epithelial cells and specialized immune cells in response to different environmental factors and respiratory viruses. In addition, we review insights obtained from experiments with murine models of allergic airway and skin inflammation and offer an overview of studies focusing on miRNA discovery using profiling techniques and bioinformatic modeling of the network effect of multiple miRNAs. In conclusion, we highlight the importance of research into miRNA function in allergy and asthma to improve our knowledge of the molecular mechanisms involved in the pathogenesis of this heterogeneous group of diseases.
Collapse
Affiliation(s)
- Julie Weidner
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Sabine Bartel
- Department of Pathology and Medical Biology GRIAC Research Institute University Medical Center Groningen University of Groningen Groningen The Netherlands
| | - Ayse Kılıç
- Channing Division of Network Medicine Brigham and Women's Hospital Boston MA USA
| | - Ulrich M. Zissler
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Harald Renz
- Institut für Laboratoriumsmedizin und Pathobiochemie Philipps University of Marburg Marburg Germany
| | - Jürgen Schwarze
- Centre for Inflammation Research The University of Edinburgh Edinburgh UK
| | - Carsten B. Schmidt‐Weber
- Center for Allergy and Environment (ZAUM) Technical University of Munich and Helmholtz Center MunichGerman Research Center for Environmental Health Munich Germany
| | - Tania Maes
- Department of Respiratory Medicine Ghent University Ghent Belgium
| | - Ana Rebane
- Institute of Biomedicine and Translational Medicine University of Tartu Tartu Estonia
| | - Susanne Krauss‐Etschmann
- Research Center Borstel Borstel Germany
- Institute of Experimental Medicine Christian‐Albrechts University Kiel Kiel Germany
| | - Madeleine Rådinger
- Department of Internal Medicine and Clinical Nutrition Krefting Research Centre Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
7
|
Inhibitory Effects of Gyeji-Tang on MMP-9 Activity and the Expression of Adhesion Molecules in IL-4- and TNF-α-Stimulated BEAS-2B Cells. PLANTS 2021; 10:plants10050951. [PMID: 34068739 PMCID: PMC8151808 DOI: 10.3390/plants10050951] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 05/06/2021] [Accepted: 05/07/2021] [Indexed: 01/13/2023]
Abstract
Gyeji-tang (GJT), a traditional herbal formula composed of five herbal medicines, is commonly used to treat the common cold, exogenous febrile disease, fever and headaches in Korea, China and Japan. Although various pharmacological activities of GJT have been reported in several studies, the effect of GJT water extract (GJTWE) on airway inflammation has not yet been investigated. This study aimed to evaluate the effects of GJTWE on airway inflammation-related factors using human bronchial epithelial BEAS-2B cells, and to identify the phytochemicals in GJTWE by ultra-performance liquid chromatography-diode array detector-tandem mass spectrometry (UPLC-DAD-MS/MS) analysis. GJTWE significantly decreased the production of chemokines, including eotaxin-3, eotaxin-1, regulated on activation normal T-cell expressed and secreted (RANTES), and matrix metalloproteinase-9, and the expression of the adhesion molecules, intercellular adhesion molecule-1 and vascular cell adhesion molecule-1, in interleukin-4 + tumor necrosis factor-α (IT)-stimulated BEAS-2B cells. In the UPLC-DAD-MS/MS analysis, 21 phytochemicals, including six flavonoids, two chalcones, five terpenoids, six phenolics, one phenylpropanoid and one coumarin, were identified in GJTWE. The findings suggested that GJTWE might exhibit anti-inflammatory effects on airway inflammation by regulating the expression of inflammatory response-related factors in IT-stimulated BEAS-2B cells; further studies are required to determine the bioactive compounds involved in the inhibition of airway inflammation.
Collapse
|
8
|
Paul S, Ruiz-Manriquez LM, Ledesma-Pacheco SJ, Benavides-Aguilar JA, Torres-Copado A, Morales-Rodríguez JI, De Donato M, Srivastava A. Roles of microRNAs in chronic pediatric diseases and their use as potential biomarkers: A review. Arch Biochem Biophys 2021; 699:108763. [PMID: 33460581 DOI: 10.1016/j.abb.2021.108763] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 12/22/2020] [Accepted: 01/10/2021] [Indexed: 02/09/2023]
Abstract
MicroRNAs (miRNAs) are small non-coding highly conserved RNA molecules that can act as master regulators of gene expression in a sequence-specific manner either by translation repression or mRNA degradation, influencing a wide range of biologic processes that are essential for the maintenance of cellular homeostasis. Chronic pediatric diseases are the leading cause of death worldwide among children and the recent evidence indicates that aberrant miRNA expression significantly contributes to the development of chronic pediatric diseases. This review focuses on the role of miRNAs in five major chronic pediatric diseases including bronchial asthma, congenital heart diseases, cystic fibrosis, type 1 diabetes mellitus, and epilepsy, and their potential use as novel biomarkers for the diagnosis and prognosis of these disorders.
Collapse
Affiliation(s)
- Sujay Paul
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico.
| | - Luis M Ruiz-Manriquez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - S Janin Ledesma-Pacheco
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Javier A Benavides-Aguilar
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Andrea Torres-Copado
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Jonathan I Morales-Rodríguez
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Marcos De Donato
- Tecnologico de Monterrey, School of Engineering and Sciences, Campus Queretaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130, Queretaro, Mexico
| | - Aashish Srivastava
- Section of Bioinformatics, Clinical Laboratory, Haukeland University Hospital, Bergen, 5021, Norway; Department of Clinical Science, University of Bergen, Bergen, 5021, Norway
| |
Collapse
|
9
|
Shastri MD, Chong WC, Dua K, Peterson GM, Patel RP, Mahmood MQ, Tambuwala M, Chellappan DK, Hansbro NG, Shukla SD, Hansbro PM. Emerging concepts and directed therapeutics for the management of asthma: regulating the regulators. Inflammopharmacology 2020; 29:15-33. [PMID: 33152094 DOI: 10.1007/s10787-020-00770-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 10/17/2020] [Indexed: 12/19/2022]
Abstract
Asthma is a common, heterogeneous and serious disease, its prevalence has steadily risen in most parts of the world, and the condition is often inadequately controlled in many patients. Hence, there is a major need for new therapeutic approaches. Mild-to-moderate asthma is considered a T-helper cell type-2-mediated inflammatory disorder that develops due to abnormal immune responses to otherwise innocuous allergens. Prolonged exposure to allergens and persistent inflammation results in myofibroblast infiltration and airway remodelling with mucus hypersecretion, airway smooth muscle hypertrophy, and excess collagen deposition. The airways become hyper-responsive to provocation resulting in the characteristic wheezing and obstructed airflow experienced by patients. Extensive research has progressed the understanding of the underlying mechanisms and the development of new treatments for the management of asthma. Here, we review the basis of the disease, covering new areas such as the role of vascularisation and microRNAs, as well as associated potential therapeutic interventions utilising reports from animal and human studies. We also cover novel drug delivery strategies that are being developed to enhance therapeutic efficacy and patient compliance. Potential avenues to explore to improve the future of asthma management are highlighted.
Collapse
Affiliation(s)
- Madhur D Shastri
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Wai Chin Chong
- Department of Molecular and Translational Science, Monash University, Clayton, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Ultimo, NSW, Australia.,Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Gregory M Peterson
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Rahul P Patel
- School of Pharmacy and Pharmacology, University of Tasmania, Hobart, Australia
| | - Malik Q Mahmood
- Faculty of Health, School of Medicine, Deakin University, Melbourne, Australia
| | - Murtaza Tambuwala
- School of Pharmacy and Pharmaceutical Science, Ulster University, Belfast, Northern Ireland, UK
| | - Dinesh K Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Kuala Lumpur, Malaysia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia.,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia.,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia
| | - Shakti D Shukla
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, School of Medicine and Public Health, The University of Newcastle, Callaghan, Australia. .,Centre for Inflammation, Centenary Institute, Sydney, NSW, 2050, Australia. .,Faculty of Science, School of Life Sciences, University of Technology Sydney, Ultimo, NSW, 2007, Australia.
| |
Collapse
|
10
|
miRNAs in depression vulnerability and resilience: novel targets for preventive strategies. J Neural Transm (Vienna) 2019; 126:1241-1258. [PMID: 31350592 PMCID: PMC6746676 DOI: 10.1007/s00702-019-02048-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 07/11/2019] [Indexed: 02/06/2023]
Abstract
The exposure to stressful experiences during the prenatal period and through the first years of life is known to affect the brain developmental trajectories, leading to an enhanced vulnerability for the development of several psychiatric disorders later in life. However, not all the subjects exposed to the same stressful experience develop a pathologic condition, as some of them, activating coping strategies, become more resilient. The disclosure of mechanisms associated with stress vulnerability or resilience may allow the identification of novel biological processes and potential molecules that, if properly targeted, may prevent susceptibility or potentiate resilience. Over the last years, miRNAs have been proposed as one of the epigenetic mechanisms mediating the long-lasting effects of stress. Accordingly, they are associated with the development of stress vulnerability or resilience-related strategies. Moreover, miRNAs have been proposed as possible biomarkers able to identify subjects at high risk to develop depression and to predict the response to pharmacological treatments. In this review, we aimed to provide an overview of findings from studies in rodents and humans focused on the involvement of miRNAs in the mechanisms of stress response with the final goal to identify distinct sets of miRNAs involved in stress vulnerability or resilience. In addition, we reviewed studies on alterations of miRNAs in the context of depression, showing data on the involvement of miRNAs in the pathogenesis of the disease and in the efficacy of pharmacological treatments, discussing the potential utility of miRNAs as peripheral biomarkers able to predict the treatment response.
Collapse
|
11
|
Feketea G, Bocsan CI, Popescu C, Gaman M, Stanciu LA, Zdrenghea MT. A Review of Macrophage MicroRNAs' Role in Human Asthma. Cells 2019; 8:cells8050420. [PMID: 31071965 PMCID: PMC6562863 DOI: 10.3390/cells8050420] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 05/01/2019] [Accepted: 05/07/2019] [Indexed: 02/07/2023] Open
Abstract
There is an imbalance in asthma between classically activated macrophages (M1 cells) and alternatively activated macrophages (M2 cells) in favor of the latter. MicroRNAs (miRNAs) play a critical role in regulating macrophage proliferation and differentiation and control the balance of M1 and M2 macrophage polarization, thereby controlling immune responses. Here we review the current published data concerning miRNAs with known correlation to a specific human macrophage phenotype and polarization, and their association with adult asthma. MiRNA-targeted therapy is still in the initial stages, but clinical trials are under recruitment or currently running for some miRNAs in other diseases. Regulating miRNA expression via their upregulation or downregulation could show potential as a novel therapy for improving treatment efficacy in asthma.
Collapse
Affiliation(s)
- Gavriela Feketea
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.
| | - Corina I Bocsan
- Department of Pharmacology, Toxicology and Clinical Pharmacology, Iuliu Haţieganu University of Medicine and Pharmacy, 400337 Cluj-Napoca, Romania.
| | - Cristian Popescu
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.
| | - Mihaela Gaman
- Department of Hematology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania.
| | - Luminita A Stanciu
- National Heart and Lung Institute, Imperial College London, London W2 1PG, UK.
| | - Mihnea T Zdrenghea
- Department of Hematology, Iuliu Haţieganu University of Medicine and Pharmacy, 400124 Cluj-Napoca, Romania.
- Department of Hematology, Ion Chiricuta Oncology Institute, 400010 Cluj-Napoca, Romania.
| |
Collapse
|
12
|
Qian FH, Deng X, Zhuang QX, Wei B, Zheng DD. miR‑625‑5p suppresses inflammatory responses by targeting AKT2 in human bronchial epithelial cells. Mol Med Rep 2019; 19:1951-1957. [PMID: 30628701 DOI: 10.3892/mmr.2019.9817] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2017] [Accepted: 10/02/2018] [Indexed: 12/07/2022] Open
Abstract
Asthma is a common chronic inflammatory airway disease; however, whether microRNAs (miRs) could be used in the treatment of asthma remains unclear. The aim of the present study was to investigate the role of miR‑625‑5p in the inflammatory response of human bronchial epithelial cells (HBECs). Inflammation in the HBEC line, 16HBEC, was induced using different concentrations of lipopolysaccharide (LPS), which demonstrated that 1 µg/ml LPS was an appropriate concentration for further experiments. The association between protein kinase B2 (AKT2) and miR‑625‑5p was verified using a luciferase reporter assay. LPS was added to 16HBECs following the administration of miR‑625‑5p mimics or miR‑625‑5p inhibitors, and cells with silenced or overexpressed AKT2 levels. miR‑625‑5p was expressed at a high level in LPS‑activated 16HBECs. Overexpression of miR‑625‑5p inhibited interleukin (IL)‑6 and tumor necrosis factor (TNF)‑α secretion in 16HBECs. Inhibition of miR‑625‑5p enhanced LPS‑induced IL‑6 and TNF‑α secretion. miR‑625‑5p negatively regulated the expression of AKT2 in 16HBECs. A dual‑luciferase reporter assay system confirmed that miR‑625‑5p directly targeted the 3'untranslated region of AKT2. Transfection with a small interfering RNA against AKT2 inhibited inhibitor of κB phosphorylation. In brief, miR‑625‑5p may protect LPS‑induced HBECs by targeting AKT2 and inhibiting the nuclear factor‑κB signaling pathway. Therefore, miR‑625‑5p may function as an inhibitor of asthma airway inflammation in HBECs by targeting AKT2.
Collapse
Affiliation(s)
- Fen-Hong Qian
- Department of Respiratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Xia Deng
- Department of Respiratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Qiong-Xin Zhuang
- Department of Respiratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Bin Wei
- Department of Respiratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| | - Dan-Dan Zheng
- Department of Respiratory Medicine, Affiliated Hospital of Jiangsu University, Zhenjiang, Jiangsu 212000, P.R. China
| |
Collapse
|
13
|
Henry RJ, Doran SJ, Barrett JP, Meadows VE, Sabirzhanov B, Stoica BA, Loane DJ, Faden AI. Inhibition of miR-155 Limits Neuroinflammation and Improves Functional Recovery After Experimental Traumatic Brain Injury in Mice. Neurotherapeutics 2019; 16:216-230. [PMID: 30225790 PMCID: PMC6361054 DOI: 10.1007/s13311-018-0665-9] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Micro-RNAs (miRs) are short, noncoding RNAs that negatively regulate gene expression at the post-transcriptional level and have been implicated in the pathophysiology of secondary damage after traumatic brain injury (TBI). Among miRs linked to inflammation, miR-155 has been implicated as a pro-inflammatory factor in a variety of organ systems. We examined the expression profile of miR-155, following experimental TBI (controlled cortical impact) in adult male C57Bl/6 mice, as well as the effects of acute or delayed administration of a miR-155 antagomir on post-traumatic neuroinflammatory responses and neurological recovery. Trauma robustly increased miR-155 expression in the injured cortex over 7 days. Similar TBI-induced miR-155 expression changes were also found in microglia/macrophages isolated from the injured cortex at 7 days post-injury. A miR-155 hairpin inhibitor (antagomir; 0.5 nmol), administered intracerebroventricularly (ICV) immediately after injury, attenuated neuroinflammatory markers at both 1 day and 7 days post-injury and reduced impairments in spatial working memory. Delayed ICV infusion of the miR-155 antagomir (0.5 nmol/day), beginning 24 h post-injury and continuing for 6 days, attenuated neuroinflammatory markers at 7 days post-injury and improved motor, but not cognitive, function through 28 days. The latter treatment limited NADPH oxidase 2 expression changes in microglia/macrophages in the injured cortex and reduced cortical lesion volume. In summary, TBI causes a robust and persistent neuroinflammatory response that is associated with increased miR-155 expression in microglia/macrophages, and miR-155 inhibition reduces post-traumatic neuroinflammatory responses and improves neurological recovery. Thus, miR-155 may be a therapeutic target for TBI-related neuroinflammation.
Collapse
Affiliation(s)
- Rebecca J. Henry
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Sarah J. Doran
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - James P. Barrett
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Victoria E. Meadows
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Boris Sabirzhanov
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - Bogdan A. Stoica
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
| | - David J. Loane
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
- Department of Anesthesiology, University of Maryland School of Medicine, 655 West Baltimore Street, No. 6-011, Baltimore, MD 21201 USA
| | - Alan I. Faden
- Department of Anesthesiology and Shock, Trauma and Anesthesiology Research (STAR) Center, University of Maryland School of Medicine, Baltimore, MD USA
- Department of Anesthesiology, University of Maryland School of Medicine, 685 West Baltimore Street, MSTF No. 6-02, Baltimore, MD 21201 USA
| |
Collapse
|
14
|
Tahamtan A, Teymoori-Rad M, Nakstad B, Salimi V. Anti-Inflammatory MicroRNAs and Their Potential for Inflammatory Diseases Treatment. Front Immunol 2018; 9:1377. [PMID: 29988529 PMCID: PMC6026627 DOI: 10.3389/fimmu.2018.01377] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2017] [Accepted: 06/04/2018] [Indexed: 12/27/2022] Open
Abstract
Inflammation is a complicated biological and pathophysiological cascade of responses to infections and injuries, and inflammatory mechanisms are closely related to many diseases. The magnitude, the complicated network of pro- and anti-inflammatory factors, and the direction of the inflammatory response can impact on the development and progression of various disorders. The currently available treatment strategies often target the symptoms and not the causes of inflammatory disease and may often be ineffective. Since the onset and termination of inflammation are crucial to prevent tissue damage, a range of mechanisms has evolved in nature to regulate the process including negative and positive feedback loops. In this regard, microRNAs (miRNAs) have emerged as key gene regulators to control inflammation, and it is speculated that they are fine-tune signaling regulators to allow for proper resolution and prevent uncontrolled progress of inflammatory reactions. In this review, we discuss recent findings related to significant roles of miRNAs in immune regulation, especially the potential utility of these molecules as novel anti-inflammatory agents to treat inflammatory diseases. Furthermore, we discuss the possibilities of using miRNAs as drugs in the form of miRNA mimics or miRNA antagonists.
Collapse
Affiliation(s)
- Alireza Tahamtan
- Infectious Diseases Research Centre, Golestan University of Medical Sciences, Gorgan, Iran.,Department of Microbiology, School of Medicine, Golestan University of Medical Sciences, Gorgan, Iran
| | - Majid Teymoori-Rad
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Britt Nakstad
- Department of Pediatric and Adolescent Medicine, Akershus University Hospital, Lørenskog, Norway.,Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Vahid Salimi
- Department of Virology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
15
|
Xu K, Tao W, Su Z. Propofol prevents IL-13-induced epithelial-mesenchymal transition in human colorectal cancer cells. Cell Biol Int 2018; 42:985-993. [PMID: 29569786 DOI: 10.1002/cbin.10964] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Accepted: 03/17/2018] [Indexed: 12/17/2022]
Abstract
Accumulating evidence showed that cytokines are involved in the development of cancer. IL-13 was showed to induce epithelial-mesenchymal transition and promote metastasis in colorectal cancer, providing a promising therapeutic target for cancer patients. Interestingly, recent studies showed that propofol, one of most common intravenous anesthetic agent, may have antitumor function in different cancer type. However, the impact of propofol on colorectal cancer and IL-13 induced epithelial-mesenchymal transition remains unknown. Herein, we found that propofol can effectively suppress cell proliferation in colorectal cell lines RKO and SW480 cells by using MTT assay. Furthermore, wound healing assay and migration assay demonstrated that propofol has the ability to inhibit epithelial-mesenchymal transition that induced by IL-13 in RKO and SW480 cells. Mechanistically, we found propofol treatment causes up-regulation of miR-361 and miR-135b, that suppress expression of STAT6 and thereafter leads to the inhibition of IL-13/STAT6/ZEB1 signaling pathway. In conclusion, our data for the first time demonstrated that propofol may serve as a novel therapeutic drug for targeting IL-13. The aggressive function of IL-13/STAT6/ZEB1 axis in colorectal cancer was impaired by propofol through miR-361 and miR-135b.
Collapse
Affiliation(s)
- Kejia Xu
- Department of Anaesthesiology, Tongren Hospital, Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Weimin Tao
- Department of Anaesthesiology, Tongren Hospital, Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| | - Zhe Su
- Department of Anaesthesiology, Tongren Hospital, Shanghai JiaoTong University School of Medicine, 1111 Xian Xia Road, Shanghai 200336, China
| |
Collapse
|
16
|
Chen H, Xu X, Cheng S, Xu Y, Xuefei Q, Cao Y, Xie J, Wang CY, Xu Y, Xiong W. Small interfering RNA directed against microRNA-155 delivered by a lentiviral vector attenuates asthmatic features in a mouse model of allergic asthma. Exp Ther Med 2017; 14:4391-4396. [PMID: 29104649 DOI: 10.3892/etm.2017.5093] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 06/15/2017] [Indexed: 01/21/2023] Open
Abstract
Asthma is a chronic T helper type 2 (Th2) cell-mediated inflammatory disease characterized by airway hyperresponsiveness (AHR) and airway inflammation. Although the majority of patients with asthma can achieve a good level of control with existing treatments, asthma runs a chronic course and the effectiveness of current treatment is not satisfactory for certain patients. MicroRNAs (miRNAs) are short noncoding RNAs that suppress gene expression at the post-transcriptional level; their role in regulating allergic inflammation remains largely unknown. The present study aimed to explore the role of miRNA-155 in the pathogenesis of asthma and its potential as a target for treatment. The expression of miRNA-155 increased in ovalbumin-sensitized and challenged mice compared with control mice, and lentiviral vector-delivered small interfering (si)RNA targeting miRNA-155 resulted in reduced AHR, airway inflammation and Th2 cytokine production. The data from the present study indicate that miRNA-155 serves an important role in the pathogenesis of asthma, and that lentiviral vector-delivered siRNA targeting miRNA-155 may serve as a novel approach for the treatment of allergic asthma.
Collapse
Affiliation(s)
- Huilong Chen
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Xiangqin Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Sheng Cheng
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yuzhu Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Qi Xuefei
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yong Cao
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Jungang Xie
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Cong-Yi Wang
- Center for Biomedical Research, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Yongjian Xu
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| | - Weining Xiong
- Department of Respiratory and Critical Care Medicine, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430030, P.R. China
| |
Collapse
|