1
|
Zhuang C, Kang M, Oh J, Lee M. Pulmonary delivery of cell membrane-derived nanovesicles carrying anti-miRNA155 oligonucleotides ameliorates LPS-induced acute lung injury. Regen Biomater 2024; 11:rbae092. [PMID: 39220743 PMCID: PMC11364520 DOI: 10.1093/rb/rbae092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/11/2024] [Accepted: 08/02/2024] [Indexed: 09/04/2024] Open
Abstract
Acute lung injury (ALI) is a devastating inflammatory disease. MicroRNA155 (miR155) in alveolar macrophages and lung epithelial cells enhances inflammatory reactions by inhibiting the suppressor of cytokine signaling 1 (SOCS1) in ALI. Anti-miR155 oligonucleotide (AMO155) have been suggested as a potential therapeutic reagent for ALI. However, a safe and efficient carrier is required for delivery of AMO155 into the lungs for ALI therapy. In this study, cell membrane-derived nanovesicles (CMNVs) were produced from cell membranes of LA4 mouse lung epithelial cells and evaluated as a carrier of AMO155 into the lungs. For preparation of CMNVs, cell membranes were isolated from LA4 cells and CMNVs were produced by extrusion. Cholesterol-conjugated AMO155 (AMO155c) was loaded into CMNVs and extracellular vesicles (EVs) by sonication. The physical characterization indicated that CMNVs with AMO155c (AMO155c/CMNV) were membrane-structured vesicles with a size of ∼120 nm. The delivery efficiency and therapeutic efficacy of CMNVs were compared with those of EVs or polyethylenimine (25 kDa, PEI25k). The delivery efficiency of AMO155c by CMNVs was similar to that by EVs. As a result, the miR155 levels were reduced by AMO155c/CMNV and AMO155c/EV. AMO155c/CMNV were administered intratracheally into the ALI models. The SOCS1 levels were increased more efficiently by AMO155c/CMNV than by the others, suggesting that miR155 effectively was inhibited by AMO155c/CMNV. In addition, the inflammatory cytokines were reduced more effectively by AMO155c/CMNV than they were by AMO155c/EV and AMO155c/PEI25k, reducing inflammation reactions. The results suggest that CMNVs are a useful carrier of AMO155c in the treatment of ALI.
Collapse
Affiliation(s)
- Chuanyu Zhuang
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04173, Republic of Korea
| | - Minji Kang
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04173, Republic of Korea
| | - Jihun Oh
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04173, Republic of Korea
| | - Minhyung Lee
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul 04173, Republic of Korea
| |
Collapse
|
2
|
Schneble D, El-Gazzar A, Kargarpour Z, Kramer M, Metekol S, Stoshikj S, Idzko M. Cell-type-specific role of P2Y2 receptor in HDM-driven model of allergic airway inflammation. Front Immunol 2023; 14:1209097. [PMID: 37790940 PMCID: PMC10543084 DOI: 10.3389/fimmu.2023.1209097] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Accepted: 08/28/2023] [Indexed: 10/05/2023] Open
Abstract
Allergic airway inflammation (AAI) is a chronic respiratory disease that is considered a severe restriction in daily life and is accompanied by a constant risk of acute aggravation. It is characterized by IgE-dependent activation of mast cells, infiltration of eosinophils, and activated T-helper cell type 2 (Th2) lymphocytes into airway mucosa. Purinergic receptor signaling is known to play a crucial role in inducing and maintaining allergic airway inflammation. Previous studies in an ovalbumin (OVA)-alum mouse model demonstrated a contribution of the P2Y2 purinergic receptor subtype (P2RY2) in allergic airway inflammation. However, conflicting data concerning the mechanism by which P2RY2 triggers AAI has been reported. Thus, we aimed at elucidating the cell-type-specific role of P2RY2 signaling in house dust mite (HDM)-driven model of allergic airway inflammation. Thereupon, HDM-driven AAI was induced in conditional knockout mice, deficient or intact for P2ry2 in either alveolar epithelial cells, hematopoietic cells, myeloid cells, helper T cells, or dendritic cells. To analyze the functional role of P2RY2 in these mice models, flow cytometry of bronchoalveolar lavage fluid (BALF), cytokine measurement of BALF, invasive lung function measurement, HDM re-stimulation of mediastinal lymph node (MLN) cells, and lung histology were performed. Mice that were subjected to an HDM-based model of allergic airway inflammation resulted in reduced signs of acute airway inflammation including eosinophilia in BALF, peribronchial inflammation, Th2 cytokine production, and bronchial hyperresponsiveness in mice deficient for P2ry2 in alveolar epithelial cells, hematopoietic cells, myeloid cells, or dendritic cells. Furthermore, the migration of bone-marrow-derived dendritic cells and bone-marrow-derived monocytes, both deficient in P2ry2, towards ATP was impaired. Additionally, we found reduced levels of MCP-1/CCL2 and IL-8 homologues in the BALF of mice deficient in P2ry2 in myeloid cells and lower concentrations of IL-33 in the lung tissue of mice deficient in P2ry2 in alveolar epithelial cells. In summary, our results show that P2RY2 contributes to HDM-induced airway inflammation by mediating proinflammatory cytokine production in airway epithelial cells, monocytes, and dendritic cells and drives the recruitment of lung dendritic cells and monocytes.
Collapse
Affiliation(s)
- Dominik Schneble
- Department of Pneumology, Medical Center – University of Freiburg, Freiburg, Germany
| | - Ahmed El-Gazzar
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Zahra Kargarpour
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Markus Kramer
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Seda Metekol
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Slagjana Stoshikj
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| | - Marco Idzko
- Department of Pneumology, Medical Center – University of Freiburg, Freiburg, Germany
- Department of Pulmonology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
3
|
Shrestha J, Paudel KR, Nazari H, Dharwal V, Bazaz SR, Johansen MD, Dua K, Hansbro PM, Warkiani ME. Advanced models for respiratory disease and drug studies. Med Res Rev 2023; 43:1470-1503. [PMID: 37119028 PMCID: PMC10946967 DOI: 10.1002/med.21956] [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: 02/15/2022] [Revised: 02/02/2023] [Accepted: 03/17/2023] [Indexed: 04/30/2023]
Abstract
The global burden of respiratory diseases is enormous, with many millions of people suffering and dying prematurely every year. The global COVID-19 pandemic witnessed recently, along with increased air pollution and wildfire events, increases the urgency of identifying the most effective therapeutic measures to combat these diseases even further. Despite increasing expenditure and extensive collaborative efforts to identify and develop the most effective and safe treatments, the failure rates of drugs evaluated in human clinical trials are high. To reverse these trends and minimize the cost of drug development, ineffective drug candidates must be eliminated as early as possible by employing new, efficient, and accurate preclinical screening approaches. Animal models have been the mainstay of pulmonary research as they recapitulate the complex physiological processes, Multiorgan interplay, disease phenotypes of disease, and the pharmacokinetic behavior of drugs. Recently, the use of advanced culture technologies such as organoids and lung-on-a-chip models has gained increasing attention because of their potential to reproduce human diseased states and physiology, with clinically relevant responses to drugs and toxins. This review provides an overview of different animal models for studying respiratory diseases and evaluating drugs. We also highlight recent progress in cell culture technologies to advance integrated models and discuss current challenges and present future perspectives.
Collapse
Affiliation(s)
- Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Hojjatollah Nazari
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Vivek Dharwal
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sajad Razavi Bazaz
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Matt D. Johansen
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of HealthUniversity of TechnologySydneyNew South WalesAustralia
- Faculty of Health, Australian Research Centre in Complementary & Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M. Hansbro
- Centre for InflammationCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Majid Ebrahimi Warkiani
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
- Institute for Biomedical Materials and Devices, Faculty of ScienceUniversity of Technology SydneyUltimoNew South WalesAustralia
| |
Collapse
|
4
|
Chen C, Zeng J, Lu J. Critical role of epigenetic modification in the pathogenesis of atopic dermatitis. Indian J Dermatol Venereol Leprol 2023; 89:700-709. [PMID: 37067130 DOI: 10.25259/ijdvl_298_2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2021] [Accepted: 10/24/2022] [Indexed: 03/31/2023]
Abstract
Atopic dermatitis is a chronic inflammatory skin disease characterised by recurrent eczema-like lesions and severe pruritus, along with drying and decrustation of skin. Current research relates the pathogenesis of atopic dermatitis mainly to genetic susceptibility, abnormal skin barrier function, immune disorders, Staphylococcus aureus colonisation, microbiological dysfunction and vitamin D insufficiency. Epigenetic modifications are distinct genetic phenotypes resulting from environment-driven changes in chromosome functions in the absence of nuclear DNA sequence variation. Classic epigenetic events include DNA methylation, histone protein modifications and non-coding RNA regulation. Increasing evidence has indicated that epigenetic events are involved in the pathogenesis of atopic dermatitis by their effects on multiple signalling pathways which in turn influence the above factors. This review primarily analyses the function of epigenetic regulation in the pathogenesis of atopic dermatitis. In addition, it tries to make recommendations for personalised epigenetic treatment strategies for atopic dermatitis in the future.
Collapse
Affiliation(s)
- Chunli Chen
- Department of Dermatology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jinrong Zeng
- Department of Dermatology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jianyun Lu
- Department of Dermatology, the Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| |
Collapse
|
5
|
Vanders RL, Gomez HM, Hsu AC, Daly K, Wark PAB, Horvat JC, Hansbro PM. Inflammatory and antiviral responses to influenza A virus infection are dysregulated in pregnant mice with allergic airway disease. Am J Physiol Lung Cell Mol Physiol 2023; 325:L385-L398. [PMID: 37463835 DOI: 10.1152/ajplung.00232.2022] [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: 07/24/2022] [Revised: 06/23/2023] [Accepted: 06/27/2023] [Indexed: 07/20/2023] Open
Abstract
Influenza A virus (IAV) infections are increased during pregnancy especially with asthma as a comorbidity, leading to asthma exacerbations, secondary bacterial infections, intensive care unit admissions, and mortality. We aimed to define the processes involved in increased susceptibility and severity of IAV infections during pregnancy, especially with asthma. We sensitized mice to house dust mite (HDM), induced pregnancy, and challenged with HDM to induce allergic airway disease (AAD). At midpregnancy, we induced IAV infection. We assessed viral titers, airway inflammation, lung antiviral responses, mucus hypersecretion, and airway hyperresponsiveness (AHR). During early IAV infection, pregnant mice with AAD had increased mRNA expression of the inflammatory markers Il13 and IL17 and reduced mRNA expression of the neutrophil chemoattractant marker Kc. These mice had increased mucous hyperplasia and increased AHR. miR155, miR574, miR223, and miR1187 were also reduced during early infection, as was mRNA expression of the antiviral β-defensins, Bd1, Bd2, and Spd and IFNs, Ifnα, Ifnβ, and Ifnλ. During late infection, Il17 was still increased as was eosinophil infiltration in the lungs. mRNA expression of Kc was reduced, as was neutrophil infiltration and mRNA expression of the antiviral markers Ifnβ, Ifnλ, and Ifnγ and Ip10, Tlr3, Tlr9, Pkr, and Mx1. Mucous hyperplasia was still significantly increased as was AHR. Early phase IAV infection in pregnancy with asthma heightens underlying inflammatory asthmatic phenotype and reduces antiviral responses.NEW & NOTEWORTHY Influenza A virus (IAV) infection during pregnancy with asthma is a major health concern leading to increased morbidity for both mother and baby. Using murine models, we show that IAV infection in pregnancy with allergic airway disease is associated with impaired global antiviral and antimicrobial responses, increased lung inflammation, mucus hypersecretion, and airway hyperresponsiveness (AHR). Targeting specific β-defensins or microRNAs (miRNAs) may prove useful in future treatments for IAV infection during pregnancy.
Collapse
Affiliation(s)
- Rebecca L Vanders
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Henry M Gomez
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Alan C Hsu
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Katie Daly
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Peter A B Wark
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, New South Wales, Australia
- Vaccines, Infection, Viruses and Asthma Research Program, Hunter Medical Research Institute, Newcastle, New South Wales, Australia
- Faculty of Science, School of Life Sciences, Centre for Inflammation, Centenary Institute and University of Technology Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
6
|
Wang W, Li Y, Fan J, Qu X, Shang D, Qin Q, Xu T, Hamid Q, Dang X, Chang Y, Xu D. MiR-365-3p is a negative regulator in IL-17-mediated asthmatic inflammation. Front Immunol 2022; 13:953714. [PMID: 35958620 PMCID: PMC9361323 DOI: 10.3389/fimmu.2022.953714] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 06/30/2022] [Indexed: 12/07/2022] Open
Abstract
Background Interleukin-17, the major proinflammatory cytokine secreted by Th17 cells, makes essential contribution to pathogenesis of severe asthma, while the detailed mechanisms, especially the involvement of microRNAs which are also important participants in asthma progression, remains largely unclear. Methods In this study, we established a house dust mite (HDM) extract-induced murine asthmatic models and the miRNA expression in the lung tissues of mice were profiled by miRNA microarray assay. The effect of miR-365-3p on IL-17-mediated inflammation was examined by qRT-PCR and immunoblotting analysis. The involvement of ARRB2 as target gene of miR-365-3p was verified by overexpression or RNA interference. Results HDM extract-induced asthmatic inflammation was proved to be IL17-mediated and miR-365-3p was screened out to be the only miRNA exclusively responsive to IL-17. miR-365-3p, whose expression was significantly downregulated upon IL-17 stimulation, was demonstrated to exert remarkable anti-inflammatory effect to decrease IL-17-provoked inflammatory cytokines (KC/IL-8 and IL-6) in both airway epithelial cells and macrophages of murine and human origins, verifying its universal antagonizing activity against IL-17-initiated inflammation across the two species. ARRB2 was characterized as the key target of miR-365-3p to negate IL-17-induced inflammatory cytokines. Conclusion Taken together, our data supported the notion that miR-365-3p, which was diminished by IL-17 in murine and human asthmatic pathogenesis, functioned as an essential negative mediator in IL-17-stimuated inflammatory response by targeting ARRB2, which would shed new light to the understanding and therapeutics thereof of asthmatic inflammation.
Collapse
Affiliation(s)
- Weijia Wang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Ying Li
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Jiaqi Fan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Xiaoyan Qu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
| | - Dong Shang
- Department of Respiration, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Qiaohong Qin
- Institute of Basic and Translational Medicine, Xi’an Medical University, Xi’an, China
| | - Tun Xu
- School of Automation Science and Engineering, Faculty of Electronic and Information Engineering, Xi’an Jiaotong University, Xi’an, China
| | - Qutayba Hamid
- Meakins-Christie Laboratories and Respiratory Division, The Research Institute of the McGill University Health Centre and Department of Medicine, McGill University, Montreal, QC, Canada
- College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | - Xiaomin Dang
- Department of Respiration, The First Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Ying Chang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Dan Xu, ; Ying Chang,
| | - Dan Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi’an Jiaotong University, Xi’an, China
- *Correspondence: Dan Xu, ; Ying Chang,
| |
Collapse
|
7
|
Beeraka NM, Zhou R, Wang X, Vikram P R H, Kumar TP, Liu J, Greeshma MV, Mandal SP, Gurupadayya BM, Fan R. Immune Repertoire and Advancements in Nanotherapeutics for the Impediment of Severe Steroid Resistant Asthma (SSR). Int J Nanomedicine 2022; 17:2121-2138. [PMID: 35592101 PMCID: PMC9112344 DOI: 10.2147/ijn.s364693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Accepted: 04/17/2022] [Indexed: 11/28/2022] Open
Abstract
Severe steroid-resistant asthma (SSR) patients do not respond to the corticosteroid therapies due to the heterogeneity, and genome-wide variations. However, there are very limited reports pertinent to the molecular signaling underlying SSR and making pharmacologists, and formulation scientists to identify the effective therapeutic targets in order to produce novel therapies using novel drug delivery systems (NDDS). We have substantially searched literature for the peer-reviewed and published reports delineating the role of glucocorticoid-altered gene expression, and the mechanisms responsible for SSR asthma, and NDDS for treating SSR asthma using public databases PubMed, National Library of Medicine (NLM), google scholar, and medline. Subsequently, we described reports underlying the SSR pathophysiology through several immunological and inflammatory phenotypes. Furthermore, various therapeutic strategies and the role of signaling pathways such as mORC1-STAT3-FGFBP1, NLRP3 inflammasomes, miR-21/PI3K/HDAC2 axis, PI3K were delineated and these can be considered as the therapeutic targets for mitigating the pathophysiology of SSR asthma. Finally, the possibility of nanomedicine-based formulation and their applications in order to enhance the long term retention of several antioxidant and anti-asthmatic drug molecules as a significant therapeutic modality against SSR asthma was described vividly.
Collapse
Affiliation(s)
- Narasimha M Beeraka
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Department of Human Anatomy, Sechenov First Moscow State Medical University (Sechenov University), Moscow, 119991, Russia
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical college, Mysuru, Karnataka, India
| | - Runze Zhou
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Xiaoyan Wang
- Endocrinology Department, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Hemanth Vikram P R
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, 570015, Karnataka, India
| | - Tegginamath Pramod Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysore, Karnataka, 570015, India
| | - Junqi Liu
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - M V Greeshma
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Academy of Higher Education and Research (JSS AHER), JSS Medical college, Mysuru, Karnataka, India
| | - Subhankar P Mandal
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSSAHER), Mysuru, 570015, Karnataka, India
| | - B M Gurupadayya
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
| | - Ruitai Fan
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, People’s Republic of China
- Correspondence: Ruitai Fan, Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital of Zhengzhou University, 1 Jianshedong Str., Zhengzhou, 450052, People’s Republic of China, Email
| |
Collapse
|
8
|
Abstract
MicroRNAs (miRNAs) are key players in gene regulation that target specific mRNAs for degradation or translational repression. Each miRNA is synthesized as a miRNA duplex comprising two strands (5p and 3p). However, only one of the two strands becomes active and is selectively incorporated into the RNA-induced silencing complex in a process known as miRNA strand selection. Recently, significant progress has been made in understanding the factors and processes involved in strand selection. Here, we explore the selection and functionality of the miRNA star strand (either 5p or 3p), which is generally present in the cell at low levels compared to its partner strand and, historically, has been thought to possess no biological activity. We also highlight the concepts of miRNA arm switching and miRNA isomerism. Finally, we offer insights into the impact of aberrant strand selection on immunity and cancer. Leading us through this journey is miR-155, a well-established regulator of immunity and cancer, and the increasing evidence that its 3p strand plays a role in these arenas. Interestingly, the miR-155-5p/-3p ratio appears to vary dependent on the timing of the immune response, and the 3p strand seems to play a regulatory role upon its partner 5p strand.
Collapse
Affiliation(s)
- Owen Dawson
- School of Pharmacy, University of Nottingham, Nottingham NG7 2RD, UK
| | | |
Collapse
|
9
|
Rodrigo-Muñoz JM, Gil-Martínez M, Lorente-Sorolla C, García-Latorre R, Valverde-Monge M, Quirce S, Sastre J, del Pozo V. miR-144-3p Is a Biomarker Related to Severe Corticosteroid-Dependent Asthma. Front Immunol 2022; 13:858722. [PMID: 35432357 PMCID: PMC9010740 DOI: 10.3389/fimmu.2022.858722] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/08/2022] [Indexed: 01/07/2023] Open
Abstract
MicroRNAs are non-coding molecules that act both as regulators of the epigenetic landscape and as biomarkers for diseases, including asthma. In the era of personalized medicine, there is a need for novel disease-associated biomarkers that can help in classifying diseases into phenotypes for treatment selection. Currently, severe eosinophilic asthma is one of the most widely studied phenotypes in clinical practice, as many patients require higher and higher doses of corticosteroids, which in some cases fail to achieve the desired outcome. Such patients may only benefit from alternative drugs such as biologics, for which novel biomarkers are necessary. The objective of the study was to study the expression of miR-144-3p in order to discover its possible use as a diagnostic biomarker for severe asthma. For this purpose, miR-144-3p was evaluated in airway biopsies and serum from asthmatics and healthy individuals. mRNA was studied in asthmatic biopsies and smooth muscle cells transfected with miR-144-3p mimic. An in silico regulation of miR-144-3p was performed using miRSystem, miRDB, STRING, and ShinyGO for pathway analysis. From our experimental procedures, we found that miR-144-3p is a biomarker associated with asthma severity and corticosteroid treatment. MiR-144-3p is increased in asthmatic lungs, and its presence correlates directly with blood eosinophilia and with the expression of genes involved in asthma pathophysiology in the airways. When studied in serum, this miRNA was increased in severe asthmatics and associated with higher doses of corticosteroids, thereby making it a potential biomarker for severe asthma previously treated with higher doses of corticosteroids. Thus, we can conclude that miR-144-3p is associated with severe diseases in both the airways and serum of asthmatics, and this association is related to corticosteroid treatment.
Collapse
Affiliation(s)
- José M. Rodrigo-Muñoz
- Department of Immunology, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Marta Gil-Martínez
- Department of Immunology, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
| | - Clara Lorente-Sorolla
- Department of Immunology, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
| | - Raquel García-Latorre
- Department of Immunology, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
| | - Marcela Valverde-Monge
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Allergy, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
| | - Santiago Quirce
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Allergy, Hospital La Paz-Institute for Health Research (IdiPAZ), Madrid, Spain
| | - Joaquín Sastre
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), Madrid, Spain
- Department of Allergy, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
| | - Victoria del Pozo
- Department of Immunology, Instituto de Investigación Sanitaria (IIS)-Fundación Jiménez Díaz, Madrid, Spain
- Centro de Investigación Biomédica en Red (CIBER) de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
10
|
Hey J, Paulsen M, Toth R, Weichenhan D, Butz S, Schatterny J, Liebers R, Lutsik P, Plass C, Mall MA. Epigenetic reprogramming of airway macrophages promotes polarization and inflammation in muco-obstructive lung disease. Nat Commun 2021; 12:6520. [PMID: 34764283 PMCID: PMC8586227 DOI: 10.1038/s41467-021-26777-9] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Accepted: 10/19/2021] [Indexed: 12/12/2022] Open
Abstract
Lung diseases, such as cystic fibrosis and COPD, are characterized by mucus obstruction and chronic airway inflammation, but their mechanistic link remains poorly understood. Here, we focus on the function of the mucostatic airway microenvironment on epigenetic reprogramming of airway macrophages (AM) and resulting transcriptomic and phenotypical changes. Using a mouse model of muco-obstructive lung disease (Scnn1b-transgenic), we identify epigenetically controlled, differentially regulated pathways and transcription factors involved in inflammatory responses and macrophage polarization. Functionally, AMs from Scnn1b-transgenic mice have reduced efferocytosis and phagocytosis, and excessive inflammatory responses upon lipopolysaccharide challenge, mediated through enhanced Irf1 function and expression. Ex vivo stimulation of wild-type AMs with native mucus impairs efferocytosis and phagocytosis capacities. In addition, mucus induces gene expression changes, comparable with those observed in AMs from Scnn1b-transgenic mice. Our data show that mucostasis induces epigenetic reprogramming of AMs, leading to changes favoring tissue damage and disease progression. Targeting these altered AMs may support therapeutic approaches in patients with muco-obstructive lung diseases.
Collapse
Affiliation(s)
- Joschka Hey
- grid.7497.d0000 0004 0492 0584Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Ruprecht Karl University of Heidelberg, Heidelberg, Germany ,grid.452624.3Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany
| | - Michelle Paulsen
- Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany. .,Department of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany. .,Novo Nordisk Foundation Center for Stem Cell Biology, University of Copenhagen, Copenhagen, Denmark.
| | - Reka Toth
- grid.7497.d0000 0004 0492 0584Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.7497.d0000 0004 0492 0584Division of Molecular Thoracic Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Dieter Weichenhan
- grid.7497.d0000 0004 0492 0584Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Simone Butz
- grid.452624.3Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Department of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Jolanthe Schatterny
- grid.452624.3Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Department of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany
| | - Reinhard Liebers
- grid.7497.d0000 0004 0492 0584Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany ,grid.461742.2Present Address: National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Pavlo Lutsik
- grid.7497.d0000 0004 0492 0584Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Christoph Plass
- Division of Cancer Epigenomics, German Cancer Research Center (DKFZ), Heidelberg, Germany. .,Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany.
| | - Marcus A. Mall
- grid.452624.3Translational Lung Research Center Heidelberg (TLRC), German Center for Lung Research (DZL), Heidelberg, Germany ,grid.7700.00000 0001 2190 4373Department of Translational Pulmonology, University of Heidelberg, Heidelberg, Germany ,grid.7468.d0000 0001 2248 7639Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, Corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany ,grid.484013.aBerlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, Germany ,grid.452624.3German Center for Lung Research (DZL), Associated Partner, Berlin, Germany
| |
Collapse
|
11
|
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
|
12
|
MicroRNA Targets for Asthma Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1303:89-105. [PMID: 33788189 DOI: 10.1007/978-3-030-63046-1_6] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Asthma is a chronic inflammatory obstructive lung disease that is stratified into endotypes. Th2 high asthma is due to an imbalance of Th1/Th2 signaling leading to abnormally high levels of Th2 cytokines, IL-4, IL-5, and IL-13 and in some cases a reduction in type I interferons. Some asthmatics express Th2 low, Th1/Th17 high phenotypes with or without eosinophilia. Most asthmatics with Th2 high phenotype respond to beta-adrenergic agonists, muscarinic antagonists, and inhaled corticosteroids. However, 5-10% of asthmatics are not well controlled by these therapies despite significant advances in lung immunology and the pathogenesis of severe asthma. This problem is being addressed by developing novel classes of anti-inflammatory agents. Numerous studies have established efficacy of targeting pro-inflammatory microRNAs in mouse models of mild/moderate and severe asthma. Current approaches employ microRNA mimics and antagonists designed for use in vivo. Chemically modified oligonucleotides have enhanced stability in blood, increased cell permeability, and optimized target specificity. Delivery to lung tissue limits clinical applications, but it is a tractable problem. Future studies need to define the most effective microRNA targets and effective delivery systems. Successful oligonucleotide drug candidates must have adequate lung cell uptake, high target specificity, and efficacy with tolerable off-target effects.
Collapse
|
13
|
Siddiqui S, Johansson K, Joo A, Bonser LR, Koh KD, Le Tonqueze O, Bolourchi S, Bautista RA, Zlock L, Roth TL, Marson A, Bhakta NR, Ansel KM, Finkbeiner WE, Erle DJ, Woodruff PG. Epithelial miR-141 regulates IL-13-induced airway mucus production. JCI Insight 2021; 6:139019. [PMID: 33682796 PMCID: PMC8021117 DOI: 10.1172/jci.insight.139019] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 01/20/2021] [Indexed: 12/17/2022] Open
Abstract
IL-13-induced goblet cell metaplasia contributes to airway remodeling and pathological mucus hypersecretion in asthma. miRNAs are potent modulators of cellular responses, but their role in mucus regulation is largely unexplored. We hypothesized that airway epithelial miRNAs play roles in IL-13-induced mucus regulation. miR-141 is highly expressed in human and mouse airway epithelium, is altered in bronchial brushings from asthmatic subjects at baseline, and is induced shortly after airway allergen exposure. We established a CRISPR/Cas9-based protocol to target miR-141 in primary human bronchial epithelial cells that were differentiated at air-liquid-interface, and goblet cell hyperplasia was induced by IL-13 stimulation. miR-141 disruption resulted in decreased goblet cell frequency, intracellular MUC5AC, and total secreted mucus. These effects correlated with a reduction in a goblet cell gene expression signature and enrichment of a basal cell gene expression signature defined by single cell RNA sequencing. Furthermore, intranasal administration of a sequence-specific mmu-miR-141-3p inhibitor in mice decreased Aspergillus-induced secreted mucus and mucus-producing cells in the lung and reduced airway hyperresponsiveness without affecting cellular inflammation. In conclusion, we have identified a miRNA that regulates pathological airway mucus production and is amenable to therapeutic manipulation through an inhaled route.
Collapse
Affiliation(s)
- Sana Siddiqui
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | - Kristina Johansson
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
- Department of Microbiology and Immunology
| | - Alex Joo
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | | | - Kyung Duk Koh
- Lung Biology Center
- Cardiovascular Research Institute
| | | | - Samaneh Bolourchi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | - Rodriel A. Bautista
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
| | | | - Theodore L. Roth
- Department of Microbiology and Immunology
- Biomedical Sciences Graduate Program, and
- Diabetes Center, UCSF, San Francisco, California, USA
- Innovative Genomics Institute, University of California, Berkeley, California, USA
| | - Alexander Marson
- Department of Microbiology and Immunology
- Innovative Genomics Institute, University of California, Berkeley, California, USA
- J. David Gladstone Institutes, San Francisco, California, USA
- Department of Medicine, Division of Infectious Diseases, UCSF, San Francisco, California, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
- Chan Zuckerberg Biohub, San Francisco, California, USA
| | - Nirav R. Bhakta
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
| | - K. Mark Ansel
- Sandler Asthma Basic Research Center
- Department of Microbiology and Immunology
| | | | - David J. Erle
- Lung Biology Center
- Cardiovascular Research Institute
| | - Prescott G. Woodruff
- Department of Medicine, Division of Pulmonary and Critical Care Medicine
- Sandler Asthma Basic Research Center
- Cardiovascular Research Institute
| |
Collapse
|
14
|
Khan S, Haque S. Trauma, mental health, and everyday functioning among Rohingya refugee people living in short- and long-term resettlements. Soc Psychiatry Psychiatr Epidemiol 2021; 56:497-512. [PMID: 33015727 DOI: 10.1007/s00127-020-01962-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 09/23/2020] [Indexed: 12/14/2022]
Abstract
PURPOSE This study investigated if Rohingya refugee people resettled in camps in rural Bangladesh and urban locations in Malaysia had different levels of trauma, mental health and everyday functioning. The study also examined if direct and indirect exposure to traumatic events could predict PTSD, depression, generalized anxiety, and everyday functioning in the two groups separately. An attempt was also made to see if the relations between trauma and mental health were different across the two settings. METHODS This was a cross-sectional study, for which we conveniently recruited 100 adult Rohingyas, 50 from each country; the majority was males. Rohingyas in Bangladesh fled Myanmar's Rakhine State following a major military crackdown in 2017, whereas Rohingyas in Malaysia fled Rakhine gradually over the last three decades because of recurrent violence and military operations. We assessed trauma (cumulative trauma, direct trauma, and indirect trauma), PTSD, depression, generalized anxiety, and everyday functioning of the participants using traumatic event questionnaire, PTSD-8, PHQ-9, GAD-7, and WHODAS-2.0. RESULTS The Bangladeshi cohort experienced more types of traumatic events (i.e., cumulative trauma) than did the Malaysian cohort (d = 0.58). Although the two cohorts did not differ in terms of indirect exposure to traumatic incidents (i.e., indirect trauma), the Malaysian cohort had direct exposure to traumatic events (i.e., direct trauma) more frequently than did the Bangladeshi cohort (d = 1.22). The Bangladeshi cohort showed higher PTSD (d = 1.67), depression (d = 0.81), generalized anxiety (d = 1.49), and functional impairment (d = 2.51) than those in Malaysia. Hierarchical linear regression analyses showed that after controlling for demographic variables, both direct and indirect trauma significantly predicted PTSD, depression, and functional impairment among Rohingyas in Bangladesh, with direct trauma being the stronger predictor. However, similar analyses showed that only indirect trauma predicted PTSD among Rohingyas in Malaysia, while all other effects were nonsignificant. The results also showed that the predictive relationship between direct trauma and PTSD was different across the two countries. With the same level of direct trauma, a participant from Malaysia would score 0.256 points lower in PTSD than a participant from Bangladesh. CONCLUSION The recently experienced direct and indirect trauma have impaired mental health and everyday functioning among the Bangladeshi cohort. However, only indirect trauma was active to cause PTSD in the Malaysian cohort as direct trauma was weakening due to the time elapsed since migration. We discuss the results in the context of the current theories of trauma and mental health and suggest therapeutic interventions for the refugee population.
Collapse
Affiliation(s)
- Sanjida Khan
- Department of Psychology, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor DE, Malaysia
| | - Shamsul Haque
- Department of Psychology, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Jalan Lagoon Selatan, 47500, Bandar Sunway, Selangor DE, Malaysia.
| |
Collapse
|
15
|
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
|
16
|
Zhang QM, Ni WW, Li Y, Zhang X, Hou JC, Meng XC, Li AL, Jiang ZM. Analysis of altered miRNA profiling in the colon of a mouse model with β-lactoglobulin allergy. Allergol Immunopathol (Madr) 2020; 48:666-674. [PMID: 33131977 DOI: 10.1016/j.aller.2020.05.007] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2020] [Revised: 05/05/2020] [Accepted: 05/08/2020] [Indexed: 12/07/2022]
Abstract
OBJECTIVES The differences in the expression profiles of colonic miRNAs between β-lactoglobulin (β-Lg) allergic mice and normal mice were analyzed to investigate the important role of the miRNA regulation mechanism in the pathogenesis of cow's milk allergy. METHODS The present study performed Illumina sequencing to characterize the miRNA profile changes in mouse colon responding to β-Lg challenge. Target genes were predicted by TargetScan 50 and miRanda 3.3a algorithms and assessed by GO and KEGG analysis. The expression levels of selected miRNAs and cytokine production were verified by cell transfection and quantitative RT-PCR. RESULTS A total of 15 miRNAs were diversely expressed between the colon of the normal and β-Lg-sensitized mice (P < 0.05, fold change of >1.50 or <0.67), including six up-regulated miRNAs and nine down-regulated miRNAs, among which seven miRNAs were validated using qRT-PCR. GO enrichment and KEGG pathway analyses further revealed that biological process, protein binding, cytoplasm and the pathways of cancer were significantly enriched, which were closely connected to the allergic inflammation development. Additionally, six key functional interaction pairs in β-Lg allergy were identified in miRNA prediction algorithms and verified using qRT-PCR. CONCLUSIONS We can conclude that our results suggested that the miRNAs regulation network participated in the pathogenesis of cow's milk allergy.
Collapse
Affiliation(s)
- Q-M Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China
| | - W-W Ni
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China
| | - Y Li
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China
| | - X Zhang
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China
| | - J-C Hou
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China
| | - X C Meng
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China
| | - A-L Li
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China.
| | - Z-M Jiang
- Key Laboratory of Dairy Science, Ministry of Education, Food College, Northeast Agriculture University, Harbin, China.
| |
Collapse
|
17
|
Mehta M, Satija S, Paudel KR, Malyla V, Kannaujiya VK, Chellappan DK, Bebawy M, Hansbro PM, Wich PR, Dua K. Targeting respiratory diseases using miRNA inhibitor based nanotherapeutics: Current status and future perspectives. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2020; 31:102303. [PMID: 32980549 DOI: 10.1016/j.nano.2020.102303] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 09/10/2020] [Accepted: 09/11/2020] [Indexed: 12/24/2022]
Abstract
MicroRNAs (miRNAs) play a fundamental role in the developmental and physiological processes that occur in both animals and plants. AntagomiRs are synthetic antagonists of miRNA, which prevent the target mRNA from suppression. Therapeutic approaches that modulate miRNAs have immense potential in the treatment of chronic respiratory disorders. However, the successful delivery of miRNAs/antagomiRs to the lungs remains a major challenge in clinical applications. A range of materials, namely, polymer nanoparticles, lipid nanocapsules and inorganic nanoparticles, has shown promising results for intracellular delivery of miRNA in chronic respiratory disorders. This review discusses the current understanding of miRNA biology, the biological roles of antagomiRs in chronic respiratory disease and the recent advances in the therapeutic utilization of antagomiRs as disease biomarkers. Furthermore our review provides a common platform to debate on the nature of antagomiRs and also addresses the viewpoint on the new generation of delivery systems that target antagomiRs in respiratory diseases.
Collapse
Affiliation(s)
- Meenu Mehta
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia; Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
| | - Saurabh Satija
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia; School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab, India
| | - Keshav R Paudel
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia; School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia
| | - Vamshikrishna Malyla
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia; Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia
| | | | - Dinesh Kumar Chellappan
- Department of Life Sciences, School of Pharmacy, International Medical University, Bukit Jalil, Kuala Lumpur, Malaysia
| | - Mary Bebawy
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia; School of Life Sciences, University of Technology Sydney, Ultimo, NSW, Australia
| | - Peter R Wich
- School of Chemical Engineering, University of New South Wales, Sydney, NSW, Australia; Centre for Nanomedicine, University of New South Wales, Sydney, NSW, Australia
| | - Kamal Dua
- Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, NSW, Australia; Centre for Inflammation, Centenary Institute, Sydney, NSW, Australia; Priority Research Centre for Healthy Lungs, University of Newcastle & Hunter Medical Research Institute, New Lambton Heights, Newcastle, NSW, Australia; School of Pharmaceutical Sciences, Shoolini University, Solan, Himachal Pradesh, India.
| |
Collapse
|
18
|
Blomme EE, Provoost S, Bazzan E, Van Eeckhoutte HP, Roffel MP, Pollaris L, Bontinck A, Bonato M, Vandenbroucke L, Verhamme F, Joos GF, Cosio MG, Vanoirbeek JAJ, Brusselle GG, Saetta M, Maes T. Innate lymphoid cells in isocyanate-induced asthma: role of microRNA-155. Eur Respir J 2020; 56:13993003.01289-2019. [PMID: 32499335 DOI: 10.1183/13993003.01289-2019] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 05/10/2020] [Indexed: 11/05/2022]
Abstract
BACKGROUND Occupational asthma, induced by workplace exposures to low molecular weight agents such as toluene 2,4-diisocyanate (TDI), causes a significant burden to patients and society. Little is known about innate lymphoid cells (ILCs) in TDI-induced asthma. A critical regulator of ILC function is microRNA-155, a microRNA associated with asthma. OBJECTIVE To determine whether TDI exposure modifies the number of ILCs in the lung and whether microRNA-155 contributes to TDI-induced airway inflammation and hyperresponsiveness. METHODS C57BL/6 wild-type and microRNA-155 knockout mice were sensitised and challenged with TDI or vehicle. Intracellular cytokine expression in ILCs and T-cells was evaluated in bronchoalveolar lavage (BAL) fluid using flow cytometry. Peribronchial eosinophilia and goblet cells were evaluated on lung tissue, and airway hyperresponsiveness was measured using the forced oscillation technique. Putative type 2 ILCs (ILC2) were identified in bronchial biopsies of subjects with TDI-induced occupational asthma using immunohistochemistry. Human bronchial epithelial cells were exposed to TDI or vehicle. RESULTS TDI-exposed mice had higher numbers of airway goblet cells, BAL eosinophils, CD4+ T-cells and ILCs, with a predominant type 2 response, and tended to have airway hyperresponsiveness. In TDI-exposed microRNA-155 knockout mice, inflammation and airway hyperresponsiveness were attenuated. TDI exposure induced IL-33 expression in human bronchial epithelial cells and in murine lungs, which was microRNA-155 dependent in mice. GATA3+CD3- cells, presumably ILC2, were present in bronchial biopsies. CONCLUSION TDI exposure is associated with increased numbers of ILCs. The proinflammatory microRNA-155 is crucial in a murine model of TDI asthma, suggesting its involvement in the pathogenesis of occupational asthma due to low molecular weight agents.
Collapse
Affiliation(s)
- Evy E Blomme
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Sharen Provoost
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Erica Bazzan
- Dept of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Hannelore P Van Eeckhoutte
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Mirjam P Roffel
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium.,University of Groningen, University Medical Center Groningen, GRIAC (Groningen Research Institute for Asthma and COPD), Groningen, The Netherlands
| | - Lore Pollaris
- Centre for Environment and Health, KU Leuven, Leuven, Belgium
| | - Annelies Bontinck
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Matteo Bonato
- Dept of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Louise Vandenbroucke
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Fien Verhamme
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Guy F Joos
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Manuel G Cosio
- Dept of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy.,Meakins Christie Laboratories, Respiratory Division, McGill University, Montreal, QC, Canada
| | | | - Guy G Brusselle
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| | - Marina Saetta
- Dept of Cardiac, Thoracic, Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Tania Maes
- Dept of Respiratory Medicine, Laboratory for Translational Research in Obstructive Pulmonary Diseases, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
19
|
Zhu Y, Liu Y, Zhu X, Wang Z, Wang M. Upregulation of miR-155 regulates group 2 innate lymphoid cells by targeting c-maf in allergic rhinitis. Eur J Pharmacol 2020; 887:173564. [PMID: 32946865 DOI: 10.1016/j.ejphar.2020.173564] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/13/2022]
Abstract
Group 2 innate lymphoid cells (ILC2s) and Th2 type immune response are critically involved in the pathogenesis of allergic rhinitis (AR), and this pathological process is influenced by microRNAs-mediated post-transcriptional regulation. The present study investigated the adaptation and function of miR-155 in AR patients and mouse model. We found that significantly increased miR-155 expression (1.63 ± 0.12 vs. 0.92 ± 0.11 in human, and 1.68 ± 0.15 vs. 1.06 ± 0.06 in mice) and ILC2s activity in nasal mucosa and serum in AR patients and mice. Administration of miR-155 antagomir significantly reduced the activity of ILC2s in nasal mucosa, suppressed the production of Th2 cytokines in serum and nasal mucosa, and alleviated the airway inflammation and allergic symptoms in AR mice, while miR-155 agomir increased ILC2s activity and production of Th2 cytokines and induced airway inflammation and allergic symptoms in control mice. Meanwhile, the expression of transcriptional factor c-Maf (0.57 ± 0.05 vs. 0.37 ± 0.04) in nasal mucosa in AR mice, which was significantly recovered by miR-155 antagomir (0.56 ± 0.04). Treatment with miR-155 agomir decreased c-Maf expression in nasal mucosa in control mice. This synchronized with the similar pattern in the current observations that miR-155 regulated Th2 cytokine (IL-4, IL-5, IL-9 and IL-13) production, airway inflammation and allergic symptoms in AR mice. Together, upregulation miR-155 suppressed the expression of transcriptional factor c-Maf and was critically involved in the ILC2s activation, which contributed to the airway inflammation and allergic symptoms in AR.
Collapse
Affiliation(s)
- Yaqiong Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Yuehui Liu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, Jiangxi, China.
| | - Xinhua Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Zhi Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, Jiangxi, China
| | - Meiqun Wang
- Department of Otorhinolaryngology Head and Neck Surgery, Second Affiliated Hospital of Nanchang University, Jiangxi, China
| |
Collapse
|
20
|
Tan BWQ, Sim WL, Cheong JK, Kuan WS, Tran T, Lim HF. MicroRNAs in chronic airway diseases: Clinical correlation and translational applications. Pharmacol Res 2020; 160:105045. [PMID: 32590100 DOI: 10.1016/j.phrs.2020.105045] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Revised: 06/18/2020] [Accepted: 06/19/2020] [Indexed: 12/19/2022]
Abstract
MicroRNAs (miRNAs) are short single-stranded RNAs that have pivotal roles in disease pathophysiology through transcriptional and translational modulation of important genes. It has been implicated in the development of many diseases, such as stroke, cardiovascular conditions, cancers and inflammatory airway diseases. There is recent evidence that miRNAs play important roles in the pathogenesis of asthma and chronic obstructive pulmonary disease (COPD), and could help to distinguish between T2-low (non-eosinophilic, steroid-insensitive) versus T2-high (eosinophilic, steroid-sensitive) disease endotypes. As these are the two most prevalent chronic respiratory diseases globally, with rising disease burden, miRNA research might lead to the development of new diagnostic and therapeutic targets. Research involving miRNAs in airway disease is challenging because: (i) asthma and COPD are heterogeneous inflammatory airway diseases; there are overlapping but distinct inter- and intra-disease differences in the immunological pathophysiology, (ii) there exists more than 2000 known miRNAs and a single miRNA can regulate multiple targets, (iii) differential effects of miRNAs could be present in different cellular subtypes and tissues, and (iv) dysregulated miRNA expression might be a direct consequence of an indirect effect of airway disease onset or progression. As miRNAs are actively secreted in fluids and remain relatively stable, they have the potential for biomarker development and therapeutic targets. In this review, we summarize the preclinical data on potential miRNA biomarkers that mediate different pathophysiological mechanisms in airway disease. We discuss the framework for biomarker development using miRNA and highlight the need for careful patient characterization and endotyping in the screening and validation cohorts, profiling both airway and blood samples to determine the biological fluids of choice in different disease states or severity, and adopting an untargeted approach. Collaboration between the various stakeholders - pharmaceutical companies, laboratory professionals and clinician-scientists is crucial to reduce the difficulties and cost required to bring miRNA research into the translational stage for airway diseases.
Collapse
Affiliation(s)
- Bryce W Q Tan
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Wei Liang Sim
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Jit Kong Cheong
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Win Sen Kuan
- Department of Emergency Medicine, National University Hospital, National University Health System, Singapore
| | - Thai Tran
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | - Hui Fang Lim
- Division of Respiratory & Critical Care Medicine, Department of Medicine, National University Hospital, National University Health System, Singapore; Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore.
| |
Collapse
|
21
|
Jaiswal A, Maurya M, Maurya P, Barthwal MK. Lin28B Regulates Angiotensin II-Mediated Let-7c/miR-99a MicroRNA Formation Consequently Affecting Macrophage Polarization and Allergic Inflammation. Inflammation 2020; 43:1846-1861. [DOI: 10.1007/s10753-020-01258-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
22
|
Xu W, Wang Y, Ma Y, Yang J. MiR-223 plays a protecting role in neutrophilic asthmatic mice through the inhibition of NLRP3 inflammasome. Respir Res 2020; 21:116. [PMID: 32423405 PMCID: PMC7236263 DOI: 10.1186/s12931-020-01374-4] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Accepted: 04/24/2020] [Indexed: 12/11/2022] Open
Abstract
Background Neutrophilic subtype asthma occurs in approximately 15–25% of the asthma cases and is associated with severe airflow obstruction, corticosteroid resistance. MicroRNA plays a vital role in regulating many immune processes, but how miRNA circuits coordinate airway inflammation during neutrophilic asthma is unclear. Methods To investigate the molecular mechanism of miR-223 in regulation of neutrophilic airway inflammation, miR-223 knockout mice were used to the OVA/CFA-induced neutrophilic asthma or treated with NLRP3 inhibitor and IL-1β receptor antagonist. Based on the results obtained, wide-type mice were subsequently treated with miR-223 agomirs or negative control agomirs, and the effects on airway inflammation were assessed using morphometric techniques, quantitative RT-PCR, western blot, ELISA and other molecular approaches. Results The expression of miR-223 was upregulated in lung tissues of experimental mice model. Furthermore, miR-223−/− mice led to aggravated neutrophilic airway inflammation with heightened histopathological, inflammatory cells and cytokines readouts. Moreover, miR-223−/− mice also presented with enhanced NLRP3 inflammasome level with elevated IL-1β. Blocking NLRP3 or IL-1β diminished this phenotype. Finally, overexpression of miR-223 via treatment with miR-223 agomirs attenuated airway inflammation, NLRP3 levels and IL-1β release. Conclusions The findings of this study revealed a crucial role for miR-223 in regulating the immunoinflammatory responses by depressing the NLRP3/ IL-1β axis in neutrophilic asthma.
Collapse
Affiliation(s)
- Wenjuan Xu
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Yimin Wang
- National Clinical Research Center for Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Ying Ma
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China
| | - Jiong Yang
- Department of Respiratory and Critical Care Medicine, Zhongnan Hospital of Wuhan University, 169 Donghu Road, Wuhan, Hubei, 430071, People's Republic of China.
| |
Collapse
|
23
|
Carregal-Romero S, Fadón L, Berra E, Ruíz-Cabello J. MicroRNA Nanotherapeutics for Lung Targeting. Insights into Pulmonary Hypertension. Int J Mol Sci 2020; 21:ijms21093253. [PMID: 32375361 PMCID: PMC7246754 DOI: 10.3390/ijms21093253] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2020] [Revised: 04/26/2020] [Accepted: 04/30/2020] [Indexed: 02/07/2023] Open
Abstract
In this review, the potential future role of microRNA-based therapies and their specific application in lung diseases is reported with special attention to pulmonary hypertension. Current limitations of these therapies will be pointed out in order to address the challenges that they need to face to reach clinical applications. In this context, the encapsulation of microRNA-based therapies in nanovectors has shown improvements as compared to chemically modified microRNAs toward enhanced stability, efficacy, reduced side effects, and local administration. All these concepts will contextualize in this review the recent achievements and expectations reported for the treatment of pulmonary hypertension.
Collapse
Affiliation(s)
- Susana Carregal-Romero
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014 San Sebastián, Spain; (S.C.-R.); (L.F.)
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
| | - Lucía Fadón
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014 San Sebastián, Spain; (S.C.-R.); (L.F.)
| | - Edurne Berra
- Center for Cooperative Research in Bioscience (CIC bioGUNE), Buiding 800, Science and Technology Park of Bizkaia, 48160 Derio, Spain;
| | - Jesús Ruíz-Cabello
- Center for Cooperative Research in Biomaterials (CIC biomaGUNE), Basque Research and Technology Alliance (BRTA), Paseo de Miramón 182, 20014 San Sebastián, Spain; (S.C.-R.); (L.F.)
- CIBER de Enfermedades Respiratorias (CIBERES), 28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, 48013 Bilbao, Spain
- Departamento de Química en Ciencias Farmacéuticas, Universidad Complutense de Madrid, 28040 Madrid, Spain
- Correspondence:
| |
Collapse
|
24
|
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
|
25
|
Gene Expression and Epigenetic Changes in Mice Following Inhalation of Copper(II) Oxide Nanoparticles. NANOMATERIALS 2020; 10:nano10030550. [PMID: 32197515 PMCID: PMC7153614 DOI: 10.3390/nano10030550] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 03/12/2020] [Accepted: 03/17/2020] [Indexed: 12/13/2022]
Abstract
We investigated the transcriptomic response and epigenetic changes in the lungs of mice exposed to inhalation of copper(II) oxide nanoparticles (CuO NPs) (8 × 105 NPs/m3) for periods of 3 days, 2 weeks, 6 weeks, and 3 months. A whole genome transcriptome and miRNA analysis was performed using next generation sequencing. Global DNA methylation was assessed by ELISA. The inhalation resulted in the deregulation of mRNA transcripts: we detected 170, 590, 534, and 1551 differentially expressed transcripts after 3 days, 2 weeks, 6 weeks, and 3 months of inhalation, respectively. Biological processes and pathways affected by inhalation, differed between 3 days exposure (collagen formation) and longer treatments (immune response). Periods of two weeks exposure further induced apoptotic processes, 6 weeks of inhalation affected the cell cycle, and 3 months of treatment impacted the processes related to cell adhesion. The expression of miRNA was not affected by 3 days of inhalation. Prolonged exposure periods modified miRNA levels, although the numbers were relatively low (17, 18, and 38 miRNAs, for periods of 2 weeks, 6 weeks, and 3 months, respectively). Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways analysis based on miRNA–mRNA interactions, revealed the deregulation of processes implicated in the immune response and carcinogenesis. Global DNA methylation was not significantly affected in any of the exposure periods. In summary, the inhalation of CuO NPs impacted on both mRNA and miRNA expression. A significant transcriptomic response was already observed after 3 days of exposure. The affected biological processes and pathways indicated the negative impacts on the immune system and potential role in carcinogenesis.
Collapse
|
26
|
E-Lacerda RR, Anhê GF, Page CP, Riffo-Vasquez Y. Sex differences in the influence of obesity on a murine model of allergic lung inflammation. Clin Exp Allergy 2019; 50:256-266. [PMID: 31765033 DOI: 10.1111/cea.13541] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 11/05/2019] [Accepted: 11/10/2019] [Indexed: 12/19/2022]
Abstract
BACKGROUND Despite the overwhelming evidence showing the influence of sex or obesity in the development of respiratory diseases in humans and animals, the mechanisms by which these combined two factors influence allergic asthma are not well understood. OBJECTIVE We have investigated the interaction between sex and weight gain in an experimental model of lung allergic inflammation induced by chicken egg ovalbumin (OVA) in mice. METHODS Animals were fed a high-fat diet for 8 weeks and then sensitized and challenged with OVA. RESULTS Our results demonstrate that in comparison with males, high-fat diet (HFD) allergic female mice exhibit a reduction in the number of leucocytes in the lung lumen when challenged with OVA and, in contrast, an accumulation of these cells in the lung tissue. In addition, we also observed that allergic HFD female mice presented a robust lung remodelling in comparison with HFD males, evidenced by higher deposition of collagen in the airways and TGF-β in lung fluid. Measuring epithelial adhesion molecule expression, we observed that female mice presented a significantly lower expression of CD103 than males in BAL cells, regardless of the diet. Similarly, HFD female mice express lower levels of EpCAM in lung tissue in comparison with males and lean females. Levels of A20/TNFAIP3 expression in lung tissue demonstrated that HFD female mice express lower levels of these regulatory factors than all the other groups. However, this reduction was not accompanied by an increase in activated NF-κB. CONCLUSIONS Our results present evidence that the interaction between sex and weight gain alters the progression of allergic asthma in mice with females developing airway remodelling at a much earlier stage than males. CLINICAL RELEVANCE These data may contribute to a better understanding of the clinical differences in the development and severity of allergic asthma observed between men and women of reproductive age.
Collapse
Affiliation(s)
- Rodrigo Rodrigues E-Lacerda
- Sackler Institute of Pulmonary Pharmacology, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK.,Departamento de Farmacologia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Gabriel Forato Anhê
- Departamento de Farmacologia, Universidade Estadual de Campinas, Campinas, Brazil
| | - Clive Peter Page
- Sackler Institute of Pulmonary Pharmacology, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - Yanira Riffo-Vasquez
- Sackler Institute of Pulmonary Pharmacology, School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| |
Collapse
|
27
|
Dutta RK, Chinnapaiyan S, Unwalla H. Aberrant MicroRNAomics in Pulmonary Complications: Implications in Lung Health and Diseases. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:413-431. [PMID: 31655261 PMCID: PMC6831837 DOI: 10.1016/j.omtn.2019.09.007] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 09/09/2019] [Accepted: 09/11/2019] [Indexed: 02/07/2023]
Abstract
Over the last few decades, evolutionarily conserved molecular networks have emerged as important regulators in the expression and function of eukaryotic genomes. Recently, miRNAs (miRNAs), a large family of small, non-coding regulatory RNAs were identified in these networks as regulators of endogenous genes by exerting post-transcriptional gene regulation activity in a broad range of eukaryotic species. Dysregulation of miRNA expression correlates with aberrant gene expression and can play an essential role in human health and disease. In the context of the lung, miRNAs have been implicated in organogenesis programming, such as proliferation, differentiation, and morphogenesis. Gain- or loss-of-function studies revealed their pivotal roles as regulators of disease development, potential therapeutic candidates/targets, and clinical biomarkers. An altered microRNAome has been attributed to several pulmonary diseases, such as asthma, chronic pulmonary obstructive disease, cystic fibrosis, lung cancer, and idiopathic pulmonary fibrosis. Considering the relevant roles and functions of miRNAs under physiological and pathological conditions, they may lead to the invention of new diagnostic and therapeutic tools. This review will focus on recent advances in understanding the role of miRNAs in lung development, lung health, and diseases, while also exploring the progress and prospects of their application as therapeutic leads or as biomarkers.
Collapse
Affiliation(s)
- Rajib Kumar Dutta
- Department of Immunology and Nano-medicine, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Srinivasan Chinnapaiyan
- Department of Immunology and Nano-medicine, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Hoshang Unwalla
- Department of Immunology and Nano-medicine, Institute of Neuroimmune Pharmacology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| |
Collapse
|
28
|
MicroRNA-155-5p is a key regulator of allergic inflammation, modulating the epithelial barrier by targeting PKIα. Cell Death Dis 2019; 10:884. [PMID: 31767859 PMCID: PMC6877533 DOI: 10.1038/s41419-019-2124-x] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 10/24/2019] [Accepted: 10/28/2019] [Indexed: 12/13/2022]
Abstract
Recent studies have demonstrated that microRNA-155-5p (miR-155-5p) plays an essential role in the regulation of allergen-induced inflammation and is overexpressed in the skin of patients with atopic dermatitis (AD), although the mechanism is unknown. In this study, silencing miR-155-5p attenuated the thickening of the epidermis in AD model and reduced the infiltration of inflammatory cells and the secretion of Th2 cytokines. Protein kinase inhibitor α (PKIα) was identified as a direct target of miR-155-5p and correlated negatively with miR-155-5p in our AD model. Fluorescence in situ hybridization showed that miR-155-5p-expressing cells were predominantly present in the epidermis. When epithelial cells were transfected with an miR-155-5p inhibitor, the expression of PKIα, occludin, and CLDN16 increased and that of TSLP decreased significantly, whereas the overexpression of miR-155-5p resulted in the opposite changes. The increased expression of PKIα and tight junction (TJ) proteins, with reduced TSLP and IL-33, was also detected in miR-155-5p-blocked mice, in both the initial and elicitation stages of AD. The expression of TJ proteins also decreased when cells were transfected with PKIα siRNA. TJ proteins increased and TSLP and IL-33 decreased significantly after the overexpression of PKIα. Our data provide the first evidence that miR-155-5p is critical for the allergic inflammation in a mouse model of AD by directly regulating PKIα and thus epithelial TJ expression. These findings suggest new therapeutic strategies that target miR-155-5p in patients with allergic disorders.
Collapse
|
29
|
Wadhwa R, Dua K, Adcock IM, Horvat JC, Kim RY, Hansbro PM. Cellular mechanisms underlying steroid-resistant asthma. Eur Respir Rev 2019; 28:28/153/190096. [PMID: 31636089 DOI: 10.1183/16000617.0096-2019] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Accepted: 09/19/2019] [Indexed: 01/04/2023] Open
Abstract
Severe steroid-resistant asthma is clinically important, as patients with this form of the disease do not respond to mainstay corticosteroid therapies. The heterogeneity of this form of asthma and poor understanding of the pathological mechanisms involved hinder the identification of therapeutic targets and the development of more effective therapies. A major limiting factor in the understanding of severe steroid-resistant asthma is the existence of multiple endotypes represented by different immunological and inflammatory phenotypes, particularly in adults. Several clinical and experimental studies have revealed associations between specific respiratory infections and steroid-resistant asthma in adults. Here, we discuss recent findings from other authors as well as our own studies that have developed novel experimental models for interrogating the association between respiratory infections and severe steroid-resistant asthma. These models have enabled the identification of new therapies using macrolides, as well as several novel disease mechanisms, including the microRNA-21/phosphoinositide 3-kinase/histone deacetylase 2 axis and NLRP3 inflammasomes, and highlight the potential of these mechanisms as therapeutic targets.
Collapse
Affiliation(s)
- Ridhima Wadhwa
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia.,Both authors contributed equally
| | - Kamal Dua
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Discipline of Pharmacy, Graduate School of Health, University of Technology Sydney, Sydney, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Both authors contributed equally
| | - Ian M Adcock
- The Airways Disease Section, National Heart and Lung Institute, Imperial College London, London, UK
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia
| | - Richard Y Kim
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Faculty of Science, University of Technology Sydney, Sydney, Australia
| | - Philip M Hansbro
- Centre for Inflammation, Centenary Institute, Sydney, Australia.,Priority Research Centre for Healthy Lungs, University of Newcastle and Hunter Medical Research Institute, Newcastle, Australia.,Faculty of Science, University of Technology Sydney, Sydney, Australia
| |
Collapse
|
30
|
Gulei D, Raduly L, Broseghini E, Ferracin M, Berindan-Neagoe I. The extensive role of miR-155 in malignant and non-malignant diseases. Mol Aspects Med 2019; 70:33-56. [PMID: 31558293 DOI: 10.1016/j.mam.2019.09.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 02/06/2023]
Abstract
MicroRNAs (miRNAs) have rapidly emerged as key molecules in cancer initiation and development, showing the capability to regulate pivotal oncogenic pathways. MiR-155 has gathered an increased attention especially in oncology, but also in non-malignanat pathologies. Nowadays, this noncoding RNA is one of the most important miRNAs in cancer, due to the extensive signaling network associated with it, implication in immune system regulation and also deregulation in disease states. Therefore, numerous research protocols are focused on preclinical modulation of miR-155 for therapeutic purposes, or investigation of its dynamic expression for diagnostic/prognostic assessments, with the final intention of bringing this miRNA into the clinical setting. This review comprehensively presents the extended role of miR-155 in cancer and other pathologies, where its expression is dysregulated. Finally, we assess the future steps toward miR-155 based therapeutics.
Collapse
Affiliation(s)
- Diana Gulei
- MEDFUTURE - Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Marinescu 23 Street, Cluj-Napoca, Romania.
| | - Lajos Raduly
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, No. 23, 400337, Cluj-Napoca, Romania
| | - Elisabetta Broseghini
- Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Bologna, Italy
| | - Manuela Ferracin
- Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, No. 23, 400337, Cluj-Napoca, Romania; Department of Experimental, Diagnostic and Specialty Medicine, DIMES, University of Bologna, Bologna, Italy
| | - Ioana Berindan-Neagoe
- MEDFUTURE - Research Center for Advanced Medicine, University of Medicine and Pharmacy Iuliu-Hatieganu, Marinescu 23 Street, Cluj-Napoca, Romania; Research Center for Functional Genomics, Biomedicine and Translational Medicine, "Iuliu Hatieganu" University of Medicine and Pharmacy, Marinescu 23 Street, No. 23, 400337, Cluj-Napoca, Romania; Department of Functional Genomics and Experimental Pathology, The Oncology Institute "Prof. Dr. Ion Chiricuţă", Cluj-Napoca, 400015, Romania.
| |
Collapse
|
31
|
Murdaca G, Tonacci A, Negrini S, Greco M, Borro M, Puppo F, Gangemi S. Effects of AntagomiRs on Different Lung Diseases in Human, Cellular, and Animal Models. Int J Mol Sci 2019; 20:E3938. [PMID: 31412612 PMCID: PMC6719072 DOI: 10.3390/ijms20163938] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 07/14/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022] Open
Abstract
INTRODUCTION MiRNAs have been shown to play a crucial role among lung cancer, pulmonary fibrosis, tuberculosis (TBC) infection, and bronchial hypersensitivity, thus including chronic obstructive pulmonary disease (COPD) and asthma. The oncogenic effect of several miRNAs has been recently ruled out. In order to act on miRNAs turnover, antagomiRs have been developed. MATERIALS AND METHODS The systematic review was conducted under the PRISMA guidelines (registration number is: CRD42019134173). The PubMed database was searched between 1 January 2000 and 30 April 2019 under the following search strategy: (((antagomiR) OR (mirna antagonists) OR (mirna antagonist)) AND ((lung[MeSH Terms]) OR ("lung diseases"[MeSH Terms]))). We included original articles, published in English, whereas exclusion criteria included reviews, meta-analyses, single case reports, and studies published in a language other than English. RESULTS AND CONCLUSIONS A total of 68 articles matching the inclusion criteria were retrieved. Overall, the use of antagomiR was seen to be efficient in downregulating the specific miRNA they are conceived for. The usefulness of antagomiRs was demonstrated in humans, animal models, and cell lines. To our best knowledge, this is the first article to encompass evidence regarding miRNAs and their respective antagomiRs in the lung, in order to provide readers a comprehensive review upon major lung disorders.
Collapse
Affiliation(s)
- Giuseppe Murdaca
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, 16132 Genoa, Italy.
| | - Alessandro Tonacci
- Clinical Physiology Institute, National Research Council of Italy (IFC-CNR), 56124 Pisa, Italy
| | - Simone Negrini
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Monica Greco
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Matteo Borro
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Francesco Puppo
- Clinical Immunology Unit, Department of Internal Medicine, University of Genoa and Ospedale Policlinico San Martino, 16132 Genoa, Italy
| | - Sebastiano Gangemi
- School and Operative Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University of Messina, 98125 Messina, Italy
| |
Collapse
|
32
|
Specjalski K, Jassem E. MicroRNAs: Potential Biomarkers and Targets of Therapy in Allergic Diseases? Arch Immunol Ther Exp (Warsz) 2019; 67:213-223. [PMID: 31139837 PMCID: PMC6597590 DOI: 10.1007/s00005-019-00547-4] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2019] [Accepted: 05/13/2019] [Indexed: 12/17/2022]
Abstract
MicroRNAs (miRNAs) are small non-coding RNA molecules that are 18-22 nucleotides long and highly conserved throughout evolution. Currently, they are considered one of the fundamental regulatory mechanisms of genes expression. It has been demonstrated that miRNAs are involved in many biologic processes, such as signal transduction, cell proliferation and differentiation, apoptosis and stress responses. More recently, the role of miRNA has also been revealed in numerous immunological and inflammatory disorders, including allergic inflammation. Specific miRNA profiles were demonstrated in asthma, allergic rhinitis and atopic dermatitis. A core set of miRNAs involved in atopic diseases include upregulated miR-21, miR-223, miR-146a, miR-142-5p, miR-142-3p, miR-146b, miR-155 and downregulated let-7 family, miR-193b and miR-375. Most of the involved miRNAs increase secretion of Th2 cytokines (miR-1248, miR-146b), decrease secretion of Th1 cytokines (miR-513-5p, miR-625-5p) or promote differentiation of T cells towards Th2 (miR-21, miR-19a). In asthma miR-140-3p, miR-708 and miR-142-3p play a role in hyperplasia and hypertrophy of bronchial smooth muscle cells. Some single miRNAs or, more probably, their sets hold the promise for their use as biomarkers of atopic diseases. They are also promising target of future therapies.
Collapse
Affiliation(s)
- Krzysztof Specjalski
- Department of Allergology, Medical University of Gdańsk, Dębinki 7, 80-210, Gdańsk, Poland.
| | - Ewa Jassem
- Department of Allergology, Medical University of Gdańsk, Dębinki 7, 80-210, Gdańsk, Poland
| |
Collapse
|
33
|
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
|
34
|
Wodziński D, Wosiak A, Pietrzak J, Świechowski R, Jeleń A, Balcerczak E. Does the expression of the ACVR2A gene affect the development of colorectal cancer? Genet Mol Biol 2019; 42:32-39. [PMID: 30856244 PMCID: PMC6428132 DOI: 10.1590/1678-4685-gmb-2017-0332] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 06/21/2018] [Indexed: 01/19/2023] Open
Abstract
Colorectal cancer has become a serious problem, especially in highly developed
countries. As reported by the World Health Organization, the number of colon
cancer cases in the world in 2012 amounted to 1.36 million. It is the second
most common cancer in females (614,000 cases, 9.2% of the total) and the third
in males (746,000 cases, 10.0% of the total) worldwide. It is believed that TGFβ
pathway elements are involved in the pathogenesis of colorectal cancer. This
study assessed one of these elements, the ACVR2A gene.
Qualitative and quantitative analyses of the ACVR2A gene in 84
patients with colorectal cancer was performed. There was no statistically
significant association between ACVR2A gene expression and age,
gender, histological type, grading of tumor, vascular invasion, and presence of
lymphocytes in tumor tissue. No association was observed between the
ACVR2A gene expression level and the presence of metastases
in regional lymph nodes and distant metastases. In this study, larger tumors (T3
and T4) were characterized by higher ACVR2A expression compared
to smaller tumors (T1 and T2). This may indicate an association between
ACVR2A expression and the severity of pathological changes
in the tumor growth process.
Collapse
Affiliation(s)
- Damian Wodziński
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Wosiak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
| | - Jacek Pietrzak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
| | - Rafał Świechowski
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
| | - Agnieszka Jeleń
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
| | - Ewa Balcerczak
- Laboratory of Molecular Diagnostics and Pharmacogenomics, Department of Pharmaceutical Biochemistry and Molecular Diagnostics, Interfaculty Cathedral of Laboratory and Molecular Diagnostics, Medical University of Lodz, Lodz, Poland
| |
Collapse
|
35
|
Stolzenburg LR, Harris A. The role of microRNAs in chronic respiratory disease: recent insights. Biol Chem 2018; 399:219-234. [PMID: 29148977 DOI: 10.1515/hsz-2017-0249] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2017] [Accepted: 10/24/2017] [Indexed: 01/16/2023]
Abstract
Chronic respiratory diseases encompass a group of diverse conditions affecting the airways, which all impair lung function over time. They include cystic fibrosis (CF), idiopathic pulmonary fibrosis (IPF), chronic obstructive pulmonary disease (COPD) and asthma, which together affect hundreds of millions of people worldwide. MicroRNAs (miRNAs), a class of small non-coding RNAs involved in post-transcriptional gene repression, are now recognized as major regulators in the development and progression of chronic lung disease. Alterations in miRNA abundance occur in lung tissue, inflammatory cells, and freely circulating in blood and are thought to function both as drivers and modifiers of disease. Their importance in lung pathology has prompted the development of miRNA-based therapies and biomarker tools. Here, we review the current literature on miRNA expression and function in chronic respiratory disease and highlight further research that is needed to propel miRNA treatments for lung disorders towards the clinic.
Collapse
Affiliation(s)
- Lindsay R Stolzenburg
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, IL 60614, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Ann Harris
- Human Molecular Genetics Program, Lurie Children's Research Center, Chicago, IL 60614, USA.,Department of Pediatrics, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA.,Department of Genetics and Genome Sciences, Case Western Reserve University, Cleveland, OH 44016, USA
| |
Collapse
|
36
|
Role of microRNA in severe asthma. Respir Investig 2018; 57:9-19. [PMID: 30455067 DOI: 10.1016/j.resinv.2018.10.005] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Revised: 10/15/2018] [Accepted: 10/18/2018] [Indexed: 12/23/2022]
Abstract
The various roles of microRNAs (miRNAs) in the epigenetic regulation of human disease are gaining importance as areas of research, and a better understanding of these roles may identify targets for development of novel therapies for severe asthma. MiRNAs, a class of small non-coding RNAs that serve as post-transcriptional gene repressors, are recognized as critical components in regulating tissue homeostasis. Alteration in miRNA expression disrupts homeostasis and is an underlying mechanism for development of chronic respiratory diseases, including asthma. Differential profiles of miRNA expression are involved in inflammation and remodeling pathogenicity via activating airway structural cells and immune cells and inducing cytokine releases. miRNA action leads to asthma progression from mild to severe stages. Here, current knowledge of the heterogeneous roles of miRNAs in severe asthma, including biological mechanisms underlying Th2 and macrophage polarization, type 2 innate lymphoid cell (ILC2) biology regulation, steroid-resistant asthma phenotype, airway smooth muscle (ASM) dysfunction, and impaired anti-viral innate immune, are reviewed.
Collapse
|
37
|
Daniel E, Roff A, Hsu MH, Panganiban R, Lambert K, Ishmael F. Effects of allergic stimulation and glucocorticoids on miR-155 in CD4 + T-cells. AMERICAN JOURNAL OF CLINICAL AND EXPERIMENTAL IMMUNOLOGY 2018; 7:57-66. [PMID: 30245919 PMCID: PMC6146153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 07/08/2018] [Indexed: 06/08/2023]
Abstract
RATIONALE MicroRNAs (miRNAs) are emerging as important regulators of allergic inflammation and potential therapeutic targets. We sought to identify which miRNAs are expressed in CD4+ T-cells and determine whether allergic stimuli or glucocorticoids alter their expression. METHODS After IRB approval, blood was collected from dust mite (DM) allergic rhinitis subjects (n=20), non-allergic controls (n=8), and asthmatics (n=16). Peripheral blood mononuclear cells were incubated with dust mite extract (DME), diluent control, or DME + dexamethasone (0.1 µM). CD4+ T-cells were collected by magnetic bead column, and RNA was isolated by guanidinium/phenol-chloroform extraction. MicroRNA expression was measured using Nanostring microarray and quantitative real time PCR (qPCR). RESULTS We identified 196 miRNAs that were stably expressed in circulating CD4+ T-cells. Allergen stimulation of CD4+ T-cells with DME differentially induced miR-155 expression in cells of DM-allergic subjects as compared to non-allergic subjects. Induction of miR-155 expression was also observed with anti-CD3/anti-CD28 simulation and phorbol-12-Myristate-13-Acetate (PMA) treatment, and further augmented by calcium inophore and bromocyclic AMP in the latter treatment. The level of miR-155 expression was positively associated with expression of the TH2 cytokines IL-5 and IL-13. Inhibition of miR-155 in Jurkat T-cells inhibited the production of these cytokines. Glucocorticoids attenuated the effects of dust mite allergen, raising the possibility that inhibition of this miRNA could be a mechanism through which glucocorticoids exhibit their anti-inflammatory effects. The CD4+ T-cells had a higher level of miR-155 expression in asthma compared to in allergic rhinitis and non-asthmatics. The inhibitory effects of glucocorticoids on CD4+ T-cell miR-155 expression were lost in severe asthmatics. CONCLUSION Mir-155 is differentially expressed in allergic T-cells exposed to DM extract compared to in non-allergic cells and it is inhibited by glucocorticoids. MiR-155 may play a role in mediating allergic inflammation in T-cells and could be an anti-inflammatory target of steroids. This pathway may be de-regulated in severe asthma.
Collapse
Affiliation(s)
- Elizabeth Daniel
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University College of MedicineHershey, PA 17033, USA
| | - Alanna Roff
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University College of MedicineHershey, PA 17033, USA
| | - Man-Hsun Hsu
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University College of MedicineHershey, PA 17033, USA
| | - Ronaldo Panganiban
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University College of MedicineHershey, PA 17033, USA
| | - Kristin Lambert
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University College of MedicineHershey, PA 17033, USA
| | - Faoud Ishmael
- Department of Medicine, Division of Pulmonary, Allergy, and Critical Care Medicine, Pennsylvania State University College of MedicineHershey, PA 17033, USA
- Department of Biochemistry and Molecular Biology, Pennsylvania State University College of MedicineHershey, PA 17033, USA
- Allergy and Immunology, Mount Nittany Medical Group, State CollegePA 16803, USA
| |
Collapse
|
38
|
Qiu L, Zhang Y, Do DC, Ke X, Zhang S, Lambert K, Kumar S, Hu C, Zhou Y, Ishmael FT, Gao P. miR-155 Modulates Cockroach Allergen- and Oxidative Stress-Induced Cyclooxygenase-2 in Asthma. THE JOURNAL OF IMMUNOLOGY 2018; 201:916-929. [PMID: 29967100 DOI: 10.4049/jimmunol.1701167] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 06/01/2018] [Indexed: 12/13/2022]
Abstract
Exposure to cockroach allergen is a strong risk factor for developing asthma. Asthma has been associated with allergen-induced airway epithelial damage and heightened oxidant stress. In this study, we investigated cockroach allergen-induced oxidative stress in airway epithelium and its underlying mechanisms. We found that cockroach extract (CRE) could induce reactive oxygen species (ROS) production, particularly mitochondrial-derived ROS, in human bronchial epithelial cells. We then used the RT2 Profiler PCR array and identified that cyclooxygenase-2 (COX-2) was the most significantly upregulated gene related to CRE-induced oxidative stress. miR-155, predicted to target COX-2, was increased in CRE-treated human bronchial epithelial cells, and was showed to regulate COX-2 expression. Moreover, miR-155 can bind COX-2, induce COX-2 reporter activity, and maintain mRNA stability. Furthermore, CRE-treated miR-155-/- mice showed reduced levels of ROS and COX-2 expression in lung tissues and PGE2 in bronchoalveolar lavage fluid compared with wild-type mice. These miR-155-/- mice also showed reduced lung inflammation and Th2/Th17 cytokines. In contrast, when miR-155-/- mice were transfected with adeno-associated virus carrying miR-155, the phenotypic changes in CRE-treated miR-155-/- mice were remarkably reversed, including ROS, COX-2 expression, lung inflammation, and Th2/Th17 cytokines. Importantly, plasma miR-155 levels were elevated in severe asthmatics when compared with nonasthmatics or mild-to-moderate asthmatics. These increased plasma miR-155 levels were also observed in asthmatics with cockroach allergy compared with those without cockroach allergy. Collectively, these findings suggest that COX-2 is a major gene related to cockroach allergen-induced oxidative stress and highlight a novel role of miR-155 in regulating the ROS-COX-2 axis in asthma.
Collapse
Affiliation(s)
- Lipeng Qiu
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224.,Institute of Life Sciences, Jiangsu University, Zhenjiang, Jiangsu 212013, China
| | - Yan Zhang
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224.,Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Danh C Do
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Xia Ke
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Simin Zhang
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033; and
| | - Kristin Lambert
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033; and
| | - Shruthi Kumar
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224
| | - Chengping Hu
- Department of Respiratory Medicine, Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
| | - Yufeng Zhou
- Children's Hospital and Institute of Biomedical Sciences, Fudan University, Key Laboratory of Neonatal Diseases, Ministry of Health, Shanghai 201102, China
| | - Faoud T Ishmael
- Division of Pulmonary, Allergy, and Critical Care Medicine, Department of Medicine, Pennsylvania State University Milton S. Hershey Medical Center, Hershey, PA 17033; and
| | - Peisong Gao
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, MD 21224;
| |
Collapse
|
39
|
Serra MF, Cotias AC, Pão CRR, Daleprane JB, Jurgilas PB, Couto GC, Anjos-Valotta EA, Cordeiro RSB, Carvalho VF, Silva PMR, Martins MA. Repeated Allergen Exposure in A/J Mice Causes Steroid-Insensitive Asthma via a Defect in Glucocorticoid Receptor Bioavailability. THE JOURNAL OF IMMUNOLOGY 2018; 201:851-860. [PMID: 29914889 DOI: 10.4049/jimmunol.1700933] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Received: 06/27/2017] [Accepted: 05/23/2018] [Indexed: 12/21/2022]
Abstract
The importance of developing new animal models to assess the pathogenesis of glucocorticoid (GC)-insensitive asthma has been stressed. Because of the asthma-prone background of A/J mice, we hypothesized that asthma changes in these animals would be or become resistant to GCs under repeated exposures to an allergen. A/J mice were challenged with OVA for 2 or 4 consecutive d, starting on day 19 postsensitization. Oral dexamethasone or inhaled budesonide were given 1 h before challenge, and analyses were done 24 h after the last challenge. Airway hyperreactivity, leukocyte infiltration, tissue remodeling, and cytokine levels as well as phosphorylated GC receptor (p-GCR), p-GATA-3, p-p38, MAPK phosphatase-1 (MKP-1), and GC-induced leucine zipper (GILZ) levels were assessed. A/J mice subjected to two daily consecutive challenges reacted with airway hyperreactivity, subepithelial fibrosis, and marked accumulation of eosinophils in both bronchoalveolar lavage fluid and peribronchial space, all of which were clearly sensitive to dexamethasone and budesonide. Conversely, under four provocations, most of these changes were steroid resistant. A significant reduction in p-GCR/GCR ratio following 4- but not 2-d treatment was observed, as compared with untreated positive control. Accordingly, steroid efficacy to transactivate MKP-1 and GILZ and to downregulate p-p38, p-GATA-3 as well as proinflammatory cytokine levels was also seen after two but not four provocations. In conclusion, we report that repeated allergen exposure causes GC-insensitive asthma in A/J mice in a mechanism associated with decrease in GCR availability and subsequent loss of steroid capacity to modulate pivotal regulatory proteins, such as GATA-3, p-p38, MKP-1, and GILZ.
Collapse
Affiliation(s)
- Magda F Serra
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Amanda C Cotias
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Camila R R Pão
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Julio B Daleprane
- Basic and Experimental Nutrition, State University of Rio de Janeiro, Rio de Janeiro, RJ 20550-900 Brazil
| | - Patricia B Jurgilas
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Gina C Couto
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Edna A Anjos-Valotta
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Renato S B Cordeiro
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Vinicius F Carvalho
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Patricia M R Silva
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| | - Marco A Martins
- Laboratory of Inflammation, Oswaldo Cruz Institute, FIOCRUZ, Rio de Janeiro, RJ 21040-900 Brazil; and
| |
Collapse
|
40
|
Kılıç A, Santolini M, Nakano T, Schiller M, Teranishi M, Gellert P, Ponomareva Y, Braun T, Uchida S, Weiss ST, Sharma A, Renz H. A systems immunology approach identifies the collective impact of 5 miRs in Th2 inflammation. JCI Insight 2018; 3:97503. [PMID: 29875322 DOI: 10.1172/jci.insight.97503] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/29/2018] [Indexed: 12/20/2022] Open
Abstract
Allergic asthma is a chronic inflammatory disease dominated by a CD4+ T helper 2 (Th2) cell signature. The immune response amplifies in self-enforcing loops, promoting Th2-driven cellular immunity and leaving the host unable to terminate inflammation. Posttranscriptional mechanisms, including microRNAs (miRs), are pivotal in maintaining immune homeostasis. Since an altered expression of various miRs has been associated with T cell-driven diseases, including asthma, we hypothesized that miRs control mechanisms ensuring Th2 stability and maintenance in the lung. We isolated murine CD4+ Th2 cells from allergic inflamed lungs and profiled gene and miR expression. Instead of focusing on the magnitude of miR differential expression, here we addressed the secondary consequences for the set of molecular interactions in the cell, the interactome. We developed the Impact of Differential Expression Across Layers, a network-based algorithm to prioritize disease-relevant miRs based on the central role of their targets in the molecular interactome. This method identified 5 Th2-related miRs (mir27b, mir206, mir106b, mir203, and mir23b) whose antagonization led to a sharp reduction of the Th2 phenotype. Overall, a systems biology tool was developed and validated, highlighting the role of miRs in Th2-driven immune response. This result offers potentially novel approaches for therapeutic interventions.
Collapse
Affiliation(s)
- Ayşe Kılıç
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Marburg, Germany
| | - Marc Santolini
- Center for Complex Network Research, Department of Physics, Northeastern University, Boston, Massachusetts, USA.,Brigham and Women's Hospital, Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA.,Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Taiji Nakano
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Marburg, Germany
| | - Matthias Schiller
- Clinic for Dermatology and Venereology, University Medical Center Rostock, Rostock, Germany
| | - Mizue Teranishi
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Pascal Gellert
- Breast Cancer Now Research Centre at The Institute of Cancer Research, London, United Kingdom
| | - Yuliya Ponomareva
- Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe- University Frankfurt, Frankfurt Germany
| | - Thomas Braun
- Max Planck Institute for Heart and Lung Research, Bad Nauheim, Germany
| | - Shizuka Uchida
- Cardiovascular Innovation Institute, University of Louisville, Louisville, Kentucky, USA
| | - Scott T Weiss
- Brigham and Women's Hospital, Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Amitabh Sharma
- Brigham and Women's Hospital, Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Boston, Massachusetts, USA
| | - Harald Renz
- Institute of Laboratory Medicine and Pathobiochemistry, Molecular Diagnostics, Philipps University Marburg, Marburg, Germany
| |
Collapse
|
41
|
Hansbro PM, Kim RY, Starkey MR, Donovan C, Dua K, Mayall JR, Liu G, Hansbro NG, Simpson JL, Wood LG, Hirota JA, Knight DA, Foster PS, Horvat JC. Mechanisms and treatments for severe, steroid-resistant allergic airway disease and asthma. Immunol Rev 2018; 278:41-62. [PMID: 28658552 DOI: 10.1111/imr.12543] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Severe, steroid-resistant asthma is clinically and economically important since affected individuals do not respond to mainstay corticosteroid treatments for asthma. Patients with this disease experience more frequent exacerbations of asthma, are more likely to be hospitalized, and have a poorer quality of life. Effective therapies are urgently required, however, their development has been hampered by a lack of understanding of the pathological processes that underpin disease. A major obstacle to understanding the processes that drive severe, steroid-resistant asthma is that the several endotypes of the disease have been described that are characterized by different inflammatory and immunological phenotypes. This heterogeneity makes pinpointing processes that drive disease difficult in humans. Clinical studies strongly associate specific respiratory infections with severe, steroid-resistant asthma. In this review, we discuss key findings from our studies where we describe the development of representative experimental models to improve our understanding of the links between infection and severe, steroid-resistant forms of this disease. We also discuss their use in elucidating the mechanisms, and their potential for developing effective therapeutic strategies, for severe, steroid-resistant asthma. Finally, we highlight how the immune mechanisms and therapeutic targets we have identified may be applicable to obesity-or pollution-associated asthma.
Collapse
Affiliation(s)
- Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Richard Y Kim
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Chantal Donovan
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jemma R Mayall
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Gang Liu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Nicole G Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jodie L Simpson
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Lisa G Wood
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jeremy A Hirota
- James Hogg Research Centre, University of British Columbia, Vancouver, BC, Canada
| | - Darryl A Knight
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| | - Jay C Horvat
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
42
|
Saradna A, Do DC, Kumar S, Fu QL, Gao P. Macrophage polarization and allergic asthma. Transl Res 2018; 191:1-14. [PMID: 29066321 PMCID: PMC5776696 DOI: 10.1016/j.trsl.2017.09.002] [Citation(s) in RCA: 259] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 09/13/2017] [Accepted: 09/20/2017] [Indexed: 12/17/2022]
Abstract
Allergic asthma is associated with airway inflammation and airway hyperresponsiveness. Macrophage polarization has been shown to have a profound impact on asthma pathogenesis. On exposure to local microenvironments, recruited macrophages can be polarized into either classically activated (or M1) or alternatively activated (or M2) phenotypes. Macrophage polarization has been heavily associated with development of asthma. The process of regulation of macrophage polarization involves an intricate interplay between various cytokines, chemokines, transcriptional factors, and immune-regulatory cells. Different signals from the microenvironment are controlled by different receptors on the macrophages to initiate various macrophage polarization pathways. Most importantly, there is an increased attention on the epigenetic changes (eg, microRNAs, DNA methylation, and histone modification) that impact macrophage functional responses and M1/M2 polarization through modulating cellular signaling and signature gene expression. Thus, modulation of macrophage phenotypes through molecular intervention by targeting some of those potential macrophage regulators may have therapeutic potential in the treatment of allergic asthma and other allergic diseases. In this review, we will discuss the origin of macrophages, characterization of macrophages, macrophage polarization in asthma, and the underlying mechanisms regarding allergen-induced macrophage polarization with emphasis on the regulation of epigenetics, which will provide new insights into the therapeutic strategy for asthma.
Collapse
Affiliation(s)
- Arjun Saradna
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Department of Internal Medicine, Maimonides Medical Center, Brooklyn, NY
| | - Danh C Do
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Shruthi Kumar
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Bangalore Medical College and Research Institute, Bangalore, India
| | - Qing-Ling Fu
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md; Otorhinolaryngology Hospital, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Peisong Gao
- Division Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Md.
| |
Collapse
|
43
|
Heffler E, Allegra A, Pioggia G, Picardi G, Musolino C, Gangemi S. MicroRNA Profiling in Asthma: Potential Biomarkers and Therapeutic Targets. Am J Respir Cell Mol Biol 2017; 57:642-650. [PMID: 28489455 DOI: 10.1165/rcmb.2016-0231tr] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Asthma is a heterogeneous chronic inflammatory disorder in which different endotypes contribute to define clinical inflammatory phenotypes. MicroRNAs (miRNAs) are a group of minute, endogenous 22-25 nt RNA elements that join to particular mRNAs to reduce translation and increase messenger RNA degradation. miRNAs operate in post-transcriptional control and regulate physiological and pathological processes in several illnesses. The purpose of this work is to review and discuss the current knowledge about the function of miRNAs in asthma, focusing particularly on their biological properties, pathophysiologic actions, and possible use as markers and treatments for asthma.
Collapse
Affiliation(s)
- Enrico Heffler
- 1 Personalized Medicine Asthma and Allergy Clinic, Humanitas Research Hospital, and.,2 Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Alessandro Allegra
- 3 Division of Hematology, Department of General Surgery and Oncology, University of Messina
| | - Giovanni Pioggia
- 4 Institute of Applied Sciences and Intelligent Systems-Messina Unit, and
| | - Giuseppe Picardi
- 5 Respiratory Diseases and Allergy, Department of Clinical and Experimental Medicine, University of Catania, Catania, Italy
| | - Caterina Musolino
- 3 Division of Hematology, Department of General Surgery and Oncology, University of Messina
| | - Sebastiano Gangemi
- 4 Institute of Applied Sciences and Intelligent Systems-Messina Unit, and.,6 School and Division of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, University Hospital "G. Martino," Messina, Italy; and
| |
Collapse
|
44
|
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
|
45
|
Sastre B, Cañas JA, Rodrigo-Muñoz JM, Del Pozo V. Novel Modulators of Asthma and Allergy: Exosomes and MicroRNAs. Front Immunol 2017; 8:826. [PMID: 28785260 PMCID: PMC5519536 DOI: 10.3389/fimmu.2017.00826] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Accepted: 06/29/2017] [Indexed: 12/22/2022] Open
Abstract
Intercellular communication is crucial to the immune system response. In the recent years, the discovery of exosomes has changed the way immune response orchestration was understood. Exosomes are able to operate as independent units that act as mediators in both physiological and pathological conditions. These structures contain proteins, lipidic mediators, and nucleic acids and notoriously include microRNAs (miRNAs). miRNAs are short RNA sequences (around 19-22 nucleotides) with a high phylogenetic conservation and can partially or totally regulate multiple mRNAs, inhibiting protein synthesis. In respiratory diseases such as asthma and allergic sensitization, exosomes released by several cell types and their specific content perform crucial functions in the development and continuation of the pathogenic mechanisms. Released exosomes and miRNAs inside them have been found in different types of clinical samples, such as bronchoalveolar lavage fluids and sputum supernatants, providing new data about the environmental factors and mediators that participate in the inflammatory responses that lead to the exacerbation of asthma. In this review, we summarize our current knowledge of the role of exosomes and miRNAs in asthma and allergic sensitization, paying attention to the functions that both exosomes and miRNAs are described to perform through the literature. We review the effect of exosomes and miRNAs in cells implicated in asthma pathology and the genes and pathways that they modify in them, depicting how their behavior is altered in disease status. We also describe their possible repercussion in asthma diagnosis through their possible role as biomarkers. Therefore, both exosomes and miRNAs can be viewed as potential tools to be added to the arsenal of therapeutics to treat this disease.
Collapse
Affiliation(s)
- Beatriz Sastre
- Laboratory of Immunoallergy, Department of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - José A Cañas
- Laboratory of Immunoallergy, Department of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - José M Rodrigo-Muñoz
- Laboratory of Immunoallergy, Department of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| | - Victoria Del Pozo
- Laboratory of Immunoallergy, Department of Immunology, IIS-Fundación Jiménez Díaz, Madrid, Spain.,CIBER de Enfermedades Respiratorias (CIBERES), Madrid, Spain
| |
Collapse
|
46
|
Qiu YY, Zhang YW, Qian XF, Bian T. miR-371, miR-138, miR-544, miR-145, and miR-214 could modulate Th1/Th2 balance in asthma through the combinatorial regulation of Runx3. Am J Transl Res 2017; 9:3184-3199. [PMID: 28804539 PMCID: PMC5553871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/06/2017] [Indexed: 06/07/2023]
Abstract
Asthma is tightly related to the imbalance of Th1/Th2 cells, and Runx3 plays a pivotal role in the differentiation of T helper cells. The present study aimed to investigate dysregulated microRNAs that may target Runx3 in CD4+ T cells from asthmatic patients and reveal Runx3 function in Th1/Th2 balance regulation. We detected the levels of Th1- and Th2-related cytokines by ELISA and analyzed the differentiation marker gene of T helper cells by qRT-PCR. Results indicated that an imbalance of Th1/Th2 cells was present in our asthmatic subject. Runx3 expression was reduced in the CD4+ T cells from asthmatic patients. Overexpression of Runx3 could restore the Th1/Th2 balance. After performing microRNA microarray assay, we found a series of microRNAs that were considerably altered in the CD4+ T cells from asthmatic patients. Among these upregulated microRNAs, eight microRNAs that may target Runx3 were selected by bioinformatics prediction. Five microRNAs, namely miR-371, miR-138, miR-544, miR-145, and miR-214, were confirmed by qRT-PCR and selected as candidate microRNAs. Luciferase reporter assay showed that these five microRNAs could directly target the 3'-UTR of Runx3. However, only simultaneous inhibition of these five microRNAs could alter the expression of Runx3. Most importantly, only simultaneous inhibition could improve the Th1/Th2 balance. Thus, we suggest that miR-371, miR-138, miR-544, miR-145, and miR-214 can modulate the Th1/Th2 balance in asthma by regulating Runx3 in a combinatorial manner.
Collapse
Affiliation(s)
- Yu-Ying Qiu
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNo. 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Ying-Wei Zhang
- Department of Respiratory Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNo. 321 Zhongshan Road, Nanjing 210008, Jiangsu, China
| | - Xiu-Fen Qian
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical UniversityNo. 299 Qingyang Road, Wuxi 214023, Jiangsu, China
| | - Tao Bian
- Department of Respiratory Medicine, Wuxi People’s Hospital Affiliated to Nanjing Medical UniversityNo. 299 Qingyang Road, Wuxi 214023, Jiangsu, China
| |
Collapse
|
47
|
Malmhäll C, Johansson K, Winkler C, Alawieh S, Ekerljung L, Rådinger M. Altered miR-155 Expression in Allergic Asthmatic Airways. Scand J Immunol 2017; 85:300-307. [PMID: 28199728 DOI: 10.1111/sji.12535] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Accepted: 02/09/2017] [Indexed: 01/19/2023]
Abstract
We and others have previously identified microRNAs (miRNAs) with pathological roles in animal models of asthma, where miR-146a and miR-155 have been described to play important roles in inflammatory responses. To date, few studies have investigated miRNA expression in human asthmatics. In the current study, significantly lower levels of miR-155 were detected in cell-free sputum from allergic asthmatics compared to healthy controls. Induced sputum isolated from allergic asthmatics in and out of pollen season revealed that miR-155 expression, but not miR-146a, is reduced in lymphocytes in season compared to post-season. In contrast, miR-155 was found to increase, whereas miR-146a decreased in PBMCs and cell-free PBMC culture media upon T cell receptor stimulation via αCD3/CD28 in both allergic asthmatics and healthy controls. Our findings suggest that miR-155 is differentially expressed ex vivo in airways of allergic asthmatics compared to healthy controls, which may have implications in the local immune response in allergic asthma.
Collapse
Affiliation(s)
- C Malmhäll
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - K Johansson
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | | | - S Alawieh
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - L Ekerljung
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - M Rådinger
- Krefting Research Centre, Department of Internal Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
48
|
Hsu ACY, Dua K, Starkey MR, Haw TJ, Nair PM, Nichol K, Zammit N, Grey ST, Baines KJ, Foster PS, Hansbro PM, Wark PA. MicroRNA-125a and -b inhibit A20 and MAVS to promote inflammation and impair antiviral response in COPD. JCI Insight 2017; 2:e90443. [PMID: 28405612 DOI: 10.1172/jci.insight.90443] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Influenza A virus (IAV) infections lead to severe inflammation in the airways. Patients with chronic obstructive pulmonary disease (COPD) characteristically have exaggerated airway inflammation and are more susceptible to infections with severe symptoms and increased mortality. The mechanisms that control inflammation during IAV infection and the mechanisms of immune dysregulation in COPD are unclear. We found that IAV infections lead to increased inflammatory and antiviral responses in primary bronchial epithelial cells (pBECs) from healthy nonsmoking and smoking subjects. In pBECs from COPD patients, infections resulted in exaggerated inflammatory but deficient antiviral responses. A20 is an important negative regulator of NF-κB-mediated inflammatory but not antiviral responses, and A20 expression was reduced in COPD. IAV infection increased the expression of miR-125a or -b, which directly reduced the expression of A20 and mitochondrial antiviral signaling (MAVS), and caused exaggerated inflammation and impaired antiviral responses. These events were replicated in vivo in a mouse model of experimental COPD. Thus, miR-125a or -b and A20 may be targeted therapeutically to inhibit excessive inflammatory responses and enhance antiviral immunity in IAV infections and in COPD.
Collapse
Affiliation(s)
- Alan C-Y Hsu
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Kamal Dua
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Malcolm R Starkey
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Tatt-Jhong Haw
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Prema M Nair
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Kristy Nichol
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Nathan Zammit
- Transplantation Immunology Group, Immunology Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Shane T Grey
- Transplantation Immunology Group, Immunology Division, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Katherine J Baines
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Paul S Foster
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Philip M Hansbro
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia
| | - Peter A Wark
- Priority Research Centre for Healthy Lungs, Hunter Medical Research Institute and The University of Newcastle, New South Wales, Australia.,Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, New South Wales, Australia
| |
Collapse
|
49
|
Lee HY, Lee HY, Choi JY, Hur J, Kim IK, Kim YK, Kang JY, Lee SY. Inhibition of MicroRNA-21 by an antagomir ameliorates allergic inflammation in a mouse model of asthma. Exp Lung Res 2017; 43:109-119. [PMID: 28379062 DOI: 10.1080/01902148.2017.1304465] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
AIM OF THE STUDY MicroRNA-21 (miR-21) is up-regulated during allergic airway inflammation, reflecting a Th2 immune response. We investigated the effects of an miR-21 antagomir and its mechanism of action in a mouse model of acute bronchial asthma. MATERIALS AND METHODS BALB/c mice were sensitized and challenged with ovalbumin (OVA). The anti-miR-21 antagomir was administered by intranasal inhalation from the day of sensitization. Changes in cell counts, Th2 cytokine levels in bronchoalveolar (BAL) fluid, and airway hyper-responsiveness (AHR) were examined. Histopathological changes and expression levels of miR-21 in lung tissues were analyzed. The mechanism of action of the antagomir was investigated by counting CD4+/CD8- T cells in splenocytes and by measuring the expression levels of transcription factors associated with T cell polarization. RESULTS MiR-21 expression was selectively down-regulated in the lung tissues of mice treated with anti-miR-21. The antagomir suppressed AHR compared with that of the OVA-challenged and scrambled RNA-treated groups. It also reduced the total cell and eosinophil counts in BAL fluid and the levels of Th2 cytokines, including IL-4, IL-5, and IL-13. The direct target of miR-21, IL-12p35, was induced in the antagomir-treated group, decreasing the CD4+/CD8- T cell proportions in splenocytes. The levels of transcription factors involved in the Th2-signaling pathway were reduced in lung tissues on treatment with the antagomir. CONCLUSIONS The miR-21 antagomir suppresses the development of allergic airway inflammation in a mouse model of acute bronchial asthma, inhibiting Th2 activation. These results suggest that this antagomir might be useful for treating bronchial asthma.
Collapse
Affiliation(s)
- Hwa Young Lee
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - Hea Yon Lee
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - Joon Young Choi
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - Jung Hur
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - In Kyoung Kim
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - Young Kyoon Kim
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - Ji Young Kang
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| | - Sook Young Lee
- a Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine , The Catholic University of Korea , Seoul , South Korea
| |
Collapse
|
50
|
Johansson K, Malmhäll C, Ramos-Ramírez P, Rådinger M. MicroRNA-155 is a critical regulator of type 2 innate lymphoid cells and IL-33 signaling in experimental models of allergic airway inflammation. J Allergy Clin Immunol 2017; 139:1007-1016.e9. [DOI: 10.1016/j.jaci.2016.06.035] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2016] [Revised: 05/24/2016] [Accepted: 06/28/2016] [Indexed: 01/01/2023]
|