1
|
Kim CK, Agrawal DK. Glucocorticoid Insensitivity in Severe Asthma: Underlying Molecular Mechanisms, Challenges, and Emerging Therapies. ARCHIVES OF INTERNAL MEDICINE RESEARCH 2025; 8:107-120. [PMID: 40337626 PMCID: PMC12058211 DOI: 10.26502/aimr.0202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Glucocorticoids are the cornerstone of asthma therapy due to their potent anti-inflammatory action. However, a subset of severe asthmatics do not respond to the standard glucocorticoid treatment. Such phenomenon is referred to as glucocorticoid insensitivity (GCI). From a clinical point of view, GCI is characterized by the reduced therapeutic response with improvement of less than 10-15% in lung function parameters, such as FEV1, upon the administration of an adequate glucocorticoid dose. The mechanisms underlying GCI involve disrupted glucocorticoid receptor (GR) signaling, overexpression of the dominant-negative GRβ isoform, increased activity of pro-inflammatory transcription factors such as NF-κB and AP-1, and abnormal GR phosphorylation by kinases such as p38 MAPK. These altered molecular pathways undermine the anti-inflammatory effects of glucocorticoids on immune and structural airway cells, thus maintaining the chronicity of airway inflammation and remodeling. GCI can be of innate genetic origin, as in the case of GR mutations, or acquired through environmental exposures, including viral infections, smoking, and long-term exposure to pollutants in the environment. GCI represents a big challenge in the management of asthma, since a large proportion of cases do not achieve an adequate level of control with the standard treatment options. Recent advances in the understanding of the molecular mechanisms underlying GCI have enabled the development of novel therapeutic strategies, including biologic therapies targeting interleukin-5 and IL-13, Janus kinase inhibitors, and small-molecule drugs aimed at restoring GR function. This article presents a critical discussion on the current state of knowledge regarding the glucocorticoid resistance mechanisms in asthma, identifying the clinical effects of new therapeutic strategies, with special emphasis on the need for personalized treatment regimens to improve outcomes in glucocorticoid insensitivity.
Collapse
Affiliation(s)
- Chang Kon Kim
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| | - Devendra K Agrawal
- Department of Translational Research, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, Pomona, California 91766 USA
| |
Collapse
|
2
|
Ford ML, Reza MI, Ruwanpathirana A, Sathish V, Britt RD. Integrative Roles of Pro-Inflammatory Cytokines on Airway Smooth Muscle Structure and Function in Asthma. Immunol Rev 2025; 330:e70007. [PMID: 39991781 PMCID: PMC11848829 DOI: 10.1111/imr.70007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 02/06/2025] [Indexed: 02/25/2025]
Abstract
Asthma has become more appreciated for its heterogeneity with studies identifying type 2 and non-type 2 phenotypes/endotypes that ultimately lead to airflow obstruction, airway hyperresponsiveness, and remodeling. The pro-inflammatory environment in asthma influences airway smooth muscle (ASM) structure and function. ASM has a vast repertoire of inflammatory receptors that, upon activation, contribute to prominent features in asthma, notably immune cell recruitment and activation, hypercontractility, proliferation, migration, and extracellular matrix protein deposition. These pro-inflammatory responses in ASM can be mediated by both type 2 (e.g., IL-4, IL-13, and TSLP) and non-type 2 (e.g., TNFα, IFNγ, IL-17A, and TGFβ) cytokines, highlighting roles for ASM in type 2 and non-type 2 asthma phenotypes/endotypes. In recent years, there has been considerable advances in understanding how pro-inflammatory cytokines promote ASM dysfunction and impair responsiveness to asthma therapy, corticosteroids and long-acting β2-adrenergic receptor agonists (LABAs). Transcriptomic analyses on human ASM cells and tissues have expanded our knowledge in this area but have also raised new questions regarding ASM and its role in asthma. In this review, we discuss how pro-inflammatory cytokines, corticosteroids, and LABAs affect ASM structure and function, with particular focus on changes in gene expression and transcriptional programs in type 2 and non-type 2 asthma.
Collapse
Affiliation(s)
- Maria L. Ford
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Biomedical Sciences Graduate Program, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Mohammad Irshad Reza
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth DakotaUSA
| | - Anushka Ruwanpathirana
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Biomedical Sciences Graduate Program, College of MedicineThe Ohio State UniversityColumbusOhioUSA
| | - Venkatachalem Sathish
- Department of Pharmaceutical SciencesNorth Dakota State UniversityFargoNorth DakotaUSA
| | - Rodney D. Britt
- Center for Perinatal ResearchAbigail Wexner Research Institute at Nationwide Children's HospitalColumbusOhioUSA
- Department of PediatricsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
3
|
Ma X, Luo Y, Xu J, Liu F, Xu C, Tang H. Long non-coding RNA AK007111 mediates mast cells apoptosis via targeting of protein MOAP1. J Asthma 2025:1-10. [PMID: 39969102 DOI: 10.1080/02770903.2025.2463974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 09/14/2024] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) are emerging regulators of pathophysiological processes in a variety of diseases including asthma. In this study, we reported the identification of lncRNA-AK007111 as an essential modulator of mast cell apoptosis and investigated its potential mechanism. METHODS RNA-seq profiling and transcriptome sequencing technology were adopted to screen for differentially expressed genes. Transfection was done by small interfering RNAs (siRNAs) to down-regulate lncRNA-AK007111 and Modulator of apoptosis 1 (MOAP1). Starvation was used to induce apoptosis. The apoptotic rate was measured by flow cytometry. Western Blot was conducted to detect the expression of apoptosis-related proteins. RESULTS LncRNA-AK007111 was highly expressed in IgE/Ag-mediated activation of mast cells. Down-regulation of LncRNA-AK007111 promoted apoptosis of mast cells. Down-regulation of MOAP1 attenuated apoptosis in mast cells induced by the down-regulation of lncRNA-AK007111. CONCLUSION LncRNA-AK007111 may be a potential regulator of mast cell apoptosis by interaction with MOAP1.
Collapse
Affiliation(s)
- Xiao Ma
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
- Department of Pediatrics, Yangzhou Maternal and Child Health Care Hospital Affiliated to Yangzhou University, Yangzhou, Jiangsu, China
| | - Yingying Luo
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiejing Xu
- Department of Pediatrics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Feng Liu
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Changdi Xu
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Heng Tang
- Department of Respiratory Medicine, Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
4
|
Hu Y, Wang M, Luo Y, Xie J, Jiao L, Tang W, Deng Y, Yao B. Therapeutic potency and possible mechanism of Wuhu decoction underlying asthmatic progression via Th1/Th2 imbalance. J Thorac Dis 2025; 17:265-277. [PMID: 39975754 PMCID: PMC11833591 DOI: 10.21037/jtd-24-804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 11/22/2024] [Indexed: 02/21/2025]
Abstract
Background Cough variant asthma (CVA) is a disease with no definitive diagnosis or pathogenic causes, and still lacks effective and safe treatment. Wuhu decoction is a traditional Chinese medicine with potential effects against CVA, of which underlying mechanism remains elusive. The aim of this study is to explore the therapeutic potential of Wuhu decoction against CVA. Methods The CVA mice model was established by ovalbumin (OVA) treatment. The airway hyperresponsiveness and remodeling were assessed. The pulmonary inflammatory injury was determined by counting of inflammatory cells and serum OVA-specific immunoglobulin E (IgE) in bronchoalveolar lavage fluid, as well as the amount of CD4+, CD8+ or CD25+ T cells. The Th1/Th2 balance was evaluated by type specific cytokines. The level of antisense long non-coding RNA TRPM2 (lncTRPM2-AS) and its downstream targets were determined by mRNA and protein detection, respectively. Results Wuhu decoction could improve airway hyperresponsiveness and remodeling in OVA-induced asthmatic mice model by regulating Th1/Th2 balance and affecting pulmonary inflammatory injury. On the molecular level, Wuhu decoction significantly inhibited the expression of lncTRPM2-AS, which was found to be a critical modulator of Th1/Th2 balance. Meanwhile, overexpression of lncTRPM2-AS could abolish Wuhu decoction-mediated protection effects on OVA-induced asthmatic mice model. Conclusions This study discovered the protective function of Wuhu decoction against CVA and illustrated its molecular mechanism, highlighting the therapeutic application of Wuhu decoction for asthma treatment.
Collapse
Affiliation(s)
- Yan Hu
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Mengqing Wang
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yinhe Luo
- Teaching and Research Office of Chinese and Western Combination, Hunan University of Chinese Medicine, Changsha, China
| | - Jing Xie
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Luojia Jiao
- Department of Organizational Personnel Department, Hunan University of Chinese Medicine, Changsha, China
| | - Wei Tang
- Department of Acupuncture and Massage, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Yijue Deng
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| | - Bing Yao
- Department of Pediatrics, The First Affiliated Hospital of Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
5
|
Hou L, Zuo H, Xiao B, Yao D. Toll-like receptor 4 mediated autophagy regulates airway smooth muscle cells behavior. J Asthma 2024; 61:976-987. [PMID: 38349366 DOI: 10.1080/02770903.2024.2316728] [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: 11/29/2023] [Revised: 02/02/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024]
Abstract
OBJECTIVES Airway remodeling, a prominent feature of asthma, involves aberrant proliferation, apoptosis, and migration of airway smooth muscle cells (ASMCs). Toll-like receptors (TLRs) are implicated in the regulation of the autophagy pathway. In this study, we aimed to investigate the influence of Toll-like receptor 4 (TLR4) on autophagy and its underlying mechanism in ASMC proliferation, apoptosis, and migration. METHODS Histopathological changes in the lungs of asthmatic mice assessed by Hematoxylin-Eosin (HE) and Masson staining. Cell proliferation, apoptosis and migration were evaluated utilizing CCK8, Edu, Flow cytometry and wound heading assays. The effectiveness of siRNA transfection and the expression of TLR4, autophagy, and proliferation-related proteins after siRNA treatment were examined through RT-PCR and Western blot (WB). CONCLUSION We observed an increase in TLR4 expression and autophagy in a mouse model of OVA-induced asthma. In vitro experiments showed that siRNA-mediated inhibition of TLR4 suppressed autophagy, proliferation, and migration of ASMCs, whereas TLR4 activation by lipopolysaccharide (LPS) had the opposite effect. Furthermore, the autophagy inhibitor 3-Methyladenine (3MA) inhibited ASMCs proliferation and migration while promoting apoptosis. Significantly, our study demonstrated that autophagy inhibition reversed the promotion effect of LPS on ASMC proliferation and migration. These findings suggest that TLR4 may modulate ASMC behavior through the regulation of autophagy.
Collapse
Affiliation(s)
- Lixia Hou
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Key Laboratory of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, China
- Key Laboratory of Basic Research on Respiratory Diseases, Guangxi Health Commission, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Huiqing Zuo
- Key Laboratory of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Bo Xiao
- Key Laboratory of Respiratory Diseases, Affiliated Hospital of Guilin Medical University, Guilin, China
| | - Dong Yao
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Clinical Research Center for Diabetes and Metabolic Diseases, Guangxi Health Commission Key Laboratory of Glucose and Lipid Metabolism Disorders, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Guangxi Key Laboratory of Metabolic Reprogramming and Intelligent Medical Engineering for Chronic Diseases, The Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin Medical University, Guilin, China
| |
Collapse
|
6
|
Li S, Cui H, Lu H, Zheng S, Yuan C. Advances in noncoding RNA in children allergic rhinitis. Int Forum Allergy Rhinol 2024; 14:1350-1362. [PMID: 38946149 DOI: 10.1002/alr.23393] [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: 02/22/2024] [Revised: 06/02/2024] [Accepted: 06/08/2024] [Indexed: 07/02/2024]
Abstract
BACKGROUND A chronic condition that significantly reduces a child's quality of life is allergic rhinitis (AR). The environment and allergens that the body is regularly exposed to can cause inflammatory and immunological reactions, which can change the expression of certain genes Epigenetic changes are closely linked to the onset and severity of allergy disorders according to mounting amounts of data. Noncoding RNAs (ncRNAs) are a group of RNA molecules that cannot be converted into polypeptides. The three main categories of ncRNAs include microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs). NcRNAs play a crucial role in controlling gene expression and contribute to the development of numerous human diseases. METHODS Articles are selected based on Pubmed's literature review and the author's personal knowledge. The largest and highest quality studies were included. The search selection is not standardized. RESULTS Recent findings indicate that various categories of ncRNAs play distinct yet interconnected roles and actively contribute to intricate gene regulatory networks. CONCLUSION This article demonstrates the significance and progress of ncRNAs in children's AR. The database covers three key areas: miRNAs, lncRNAs, and circRNAs. Additionally, potential avenues for future research to facilitate the practical application of ncRNAs as therapeutic targets and biomarkers will be explore.
Collapse
Affiliation(s)
- Shuman Li
- Chongqing Medical University, Chongqing, China
| | - Hongtao Cui
- Department of Pediatrics, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Huina Lu
- Department of Pediatrics, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Shan Zheng
- Department of Pediatrics, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Chao Yuan
- Department of Pediatrics, Chongqing Hospital of Traditional Chinese Medicine, Chongqing, China
| |
Collapse
|
7
|
Miyachi H, Ooka T, Pérez-Losada M, Camargo CA, Hasegawa K, Zhu Z. Nasopharyngeal airway long noncoding RNAs of infants with bronchiolitis and subsequent risk of developing childhood asthma. J Allergy Clin Immunol 2024; 153:1729-1735.e7. [PMID: 38272372 PMCID: PMC11162336 DOI: 10.1016/j.jaci.2024.01.010] [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: 08/29/2023] [Revised: 01/12/2024] [Accepted: 01/18/2024] [Indexed: 01/27/2024]
Abstract
BACKGROUND Severe bronchiolitis (ie, bronchiolitis requiring hospitalization) during infancy is a major risk factor for developing childhood asthma. However, the biological mechanisms linking these 2 conditions remain unclear. OBJECTIVE We sought to investigate the longitudinal relationship between nasopharyngeal airway long noncoding RNA (lncRNA) in infants with severe bronchiolitis and subsequent asthma development. METHODS In this multicenter prospective cohort study of infants with severe bronchiolitis, we performed RNA sequencing of nasopharyngeal airway lncRNAs at index hospitalization. First, we identified differentially expressed lncRNAs (DE-lncRNAs) associated with asthma development by age 6 years. Second, we investigated the associations of DE-lncRNAs with asthma-related clinical characteristics. Third, to characterize the function of DE-lncRNAs, we performed pathway analysis for mRNA targeted by DE-lncRNAs. Finally, we examined the associations of DE-lncRNAs with nasal cytokines at index hospitalization. RESULTS Among 343 infants with severe bronchiolitis (median age, 3 months), we identified 190 DE-lncRNAs (false-discovery rate [FDR] < 0.05) associated with asthma development (eg, LINC02145, RAMP2-AS1, and PVT1). These DE-lncRNAs were associated with asthma-related clinical characteristics (FDR < 0.05), for example, respiratory syncytial virus or rhinovirus infection, infant eczema, and IgE sensitization. Furthermore, DE-lncRNAs were characterized by asthma-related pathways, including mitogen-activated protein kinase, FcɛR, and phosphatidylinositol 3-kinase (PI3K)-protein kinase B signaling pathways (FDR < 0.05). These DE-lncRNAs were also associated with nasal cytokines (eg, IL-1β, IL-4, and IL-13; FDR < 0.05). CONCLUSIONS In a multicenter cohort study of infants with severe bronchiolitis, we identified nasopharyngeal airway lncRNAs associated with childhood asthma development, characterized by asthma-related clinical characteristics, asthma-related pathways, and nasal cytokines. Our approach identifies lncRNAs underlying the bronchiolitis-asthma link and facilitates the early identification of infants at high risk of subsequent asthma development.
Collapse
Affiliation(s)
- Hideaki Miyachi
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Tadao Ooka
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass; Department of Health Sciences, University of Yamanashi, Chuo, Yamanashi, Japan
| | - Marcos Pérez-Losada
- Department of Biostatistics and Bioinformatics, Computational Biology Institute, The George Washington University, Washington, DC
| | - Carlos A Camargo
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Kohei Hasegawa
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Zhaozhong Zhu
- Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Mass.
| |
Collapse
|
8
|
Bao T, Liu X, Hu J, Ma M, Li J, Cao L, Yu B, Cheng H, Zhao S, Tian Z. Recruitment of PVT1 Enhances YTHDC1-Mediated m6A Modification of IL-33 in Hyperoxia-Induced Lung Injury During Bronchopulmonary Dysplasia. Inflammation 2024; 47:469-482. [PMID: 37917328 PMCID: PMC11074042 DOI: 10.1007/s10753-023-01923-1] [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: 07/06/2023] [Revised: 09/26/2023] [Accepted: 10/17/2023] [Indexed: 11/04/2023]
Abstract
Bronchopulmonary dysplasia (BPD) is a chronic lung disease that specifically affects preterm infants. Oxygen therapy administered to treat BPD can lead to hyperoxia-induced lung injury, characterized by apoptosis of lung alveolar epithelial cells. Our epitranscriptomic microarray analysis of normal mice lungs and hyperoxia-stimulated mice lungs revealed elevated RNA expression levels of IL-33, as well as increased m6A RNA methylation levels of IL-33 and PVT1 in the hyperoxia-stimulated lungs. This study aimed to investigate the role of the PVT1/IL-33 axis in BPD. A mouse model of BPD was established through hyperoxia induction, and lung histological changes were assessed by hematoxylin-eosin staining. Parameters such as radial alveolar count and mean chord length were measured to assess lung function. Mouse and human lung alveolar epithelial cells (MLE12 and A549, respectively) were stimulated with hyperoxia to create an in vitro BPD model. Cell apoptosis was detected using Western blotting and flow cytometry analysis. Our results demonstrated that silencing PVT1 suppressed apoptosis in MLE12 and A549 cells and improved lung function in hyperoxia-stimulated lungs. Additionally, IL-33 reversed the effects of PVT1 both in vivo and in vitro. Through online bioinformatics analysis and RNA-binding protein immunoprecipitation assays, YTHDC1 was identified as a RNA-binding protein (RBP) for both PVT1 and IL-33. We found that PVT1 positively regulated IL-33 expression by recruiting YTHDC1 to mediate m6A modification of IL-33. In conclusion, silencing PVT1 demonstrated beneficial effects in alleviating BPD by facilitating YTHDC1-mediated m6A modification of IL-33. Inhibition of the PVT1/IL-33 axis to suppress apoptosis in lung alveolar epithelial cells may hold promise as a therapeutic approach for managing hyperoxia-induced lung injury in BPD.
Collapse
Affiliation(s)
- Tianping Bao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Xiangye Liu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Jian Hu
- Department of Pediatrics, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, Huai'an, Jiangsu, China
| | - Mengmeng Ma
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Jingyan Li
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Linxia Cao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Bingrui Yu
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Huaiping Cheng
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China
| | - Sai Zhao
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China.
| | - Zhaofang Tian
- Department of Neonatology, The Affiliated Huaian No.1 People's Hospital of Nanjing Medical University, No. 1 Western Huanghe Road, Huai'an, 223300, Jiangsu, China.
| |
Collapse
|
9
|
Ji N, Chen Z, Wang Z, Sun W, Yuan Q, Zhang X, Jia X, Wu J, Jiang J, Song M, Xu T, Liu Y, Ma Q, Sun Z, Bao Y, Zhang M, Huang M. LincR-PPP2R5C Promotes Th2 Cell Differentiation Through PPP2R5C/PP2A by Forming an RNA-DNA Triplex in Allergic Asthma. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2024; 16:71-90. [PMID: 38262392 PMCID: PMC10823138 DOI: 10.4168/aair.2024.16.1.71] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 08/13/2023] [Accepted: 08/22/2023] [Indexed: 01/25/2024]
Abstract
PURPOSE The roles and mechanisms of long noncoding RNAs (lncRNAs) in T helper 2 (Th2) differentiation from allergic asthma are poorly understood. We aimed to explore a novel lncRNA, LincR-protein phosphatase 2 regulatory subunit B' gamma (PPP2R5C), in Th2 differentiation in a mouse model of asthma. METHODS LincR-PPP2R5C from RNA-seq data of CD4+ T cells of asthma-like mice were validated and confirmed by quantitative reverse transcription polymerase chain reaction, northern blotting, nuclear and cytoplasmic separation, and fluorescence in situ hybridization (FISH). Lentiviruses encoding LincR-PPP2R5C or shRNA were used to overexpress or silence LincR-PPP2R5C in CD4+ T cells. The interactions between LincR-PPP2R5C and PPP2R5C were explored with western blotting, chromatin isolation by RNA purification assay, and fluorescence resonance energy transfer. An ovalbumin-induced acute asthma model in knockout (KO) mice (LincR-PPP2R5C KO, CD4 conditional LincR-PPP2R5C KO) was established to explore the roles of LincR-PPP2R5C in Th2 differentiation. RESULTS LncR-PPP2R5C was significantly higher in CD4+ T cells from asthmatic mice ex vivo and Th2 cells in vitro. The lentivirus encoding LincR-PPP2R5C suppressed Th1 differentiation; in contrast, the short hairpin RNA (shRNA) lentivirus decreased LincR-PPP2R5C and Th2 differentiation. Mechanistically, LincR-PPP2R5C deficiency suppressed the phosphatase activity of the protein phosphatase 2A (PP2A) holocomplex, resulting in a decline in Th2 differentiation. The formation of an RNA-DNA triplex between LincR-PPP2R5C and the PPP2R5C promoter enhanced PPP2R5C expression and activated PP2A. LincR-PPP2R5C KO and CD4 conditional KO decreased Th2 differentiation, airway hyperresponsiveness and inflammatory responses. CONCLUSIONS LincR-PPP2R5C regulated PPP2R5C expression and PP2A activity by forming an RNA-DNA triplex with the PPP2R5C promoter, leading to Th2 polarization in a mouse model of acute asthma. Our data presented the first definitive evidence of lncRNAs in the regulation of Th2 cells in asthma.
Collapse
Affiliation(s)
- Ningfei Ji
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhongqi Chen
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhengxia Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Wei Sun
- Department of Respiratory and Critical Care Medicine, Xishan People's Hospital of Wuxi City, Wuxi Branch of Zhongda Hospital Affiliate to Southeast University, Wuxi, China
| | - Qi Yuan
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xijie Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xinyu Jia
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingjing Wu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Jingxian Jiang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Meijuan Song
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Tingting Xu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanan Liu
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Qiyun Ma
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhixiao Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yanmin Bao
- Department of Respiratory Medicine, Shenzhen Children's Hospital, Shenzhen, China
| | - Mingshun Zhang
- Jiangsu Province Engineering Research Center of Antibody Drugs, NHC Key Laboratory of Antibody Technique, Department of Immunology, Nanjing Medical University, Nanjing, China.
| | - Mao Huang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China.
| |
Collapse
|
10
|
Zhu H, Shi J, Li W. Bioinformatics analysis of ceRNA network of autophagy-related genes in pediatric asthma. Medicine (Baltimore) 2023; 102:e36343. [PMID: 38050261 PMCID: PMC10695615 DOI: 10.1097/md.0000000000036343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 11/06/2023] [Indexed: 12/06/2023] Open
Abstract
The molecular underpinnings of pediatric asthma present avenues for targeted therapies. A deeper exploration into the significance of differentially expressed autophagy-related genes (DE-ARGs) and their interactions with the long noncoding RNA (lncRNA)-microRNA (miRNA)-mRNA network may offer insights into the pathogenesis of pediatric asthma. DE-ARGs were retrieved from the Gene Expression Omnibus and the Human Autophagy Database. These DE-ARGs were subjected to comprehensive analyses, including Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway, Gene Set Enrichment Analysis, and protein-protein interaction networks. The identified DE-ARGs were further verified for core gene expression. The miRDB and ENCORI databases were used for inverse miRNA predictions. Furthermore, miRNA-lncRNA interactions were predicted using LncBase and ENCORI platforms. Following the exclusion of lncRNAs exclusively localized in the nucleus and extracellular space, a competitive endogenous RNA (ceRNA) network was established and subsequently subjected to detailed analysis. The mRNA expression patterns in the ceRNA network were validated using quantitative real-time PCR. In total, 31 DE-ARGs were obtained, of which 29 were up-regulated and 2 were down-regulated. Notably, the autophagy, regulation of apoptotic signaling pathways, interferon-α/β signaling, interferon γ signaling, autophagy-animal, and apoptosis pathways were predominantly enriched in pediatric asthma. Five hub genes (VEGFA, CFLAR, RELA, FAS, and ATF6) were further analyzed using the Gene Expression Omnibus dataset to verify their expression patterns and diagnostic efficacy. Four hub genes (VEGFA, CFLAR, RELA, and FAS) were obtained. Finally, a ceRNA network of 4 mRNAs (VEGFA, CFLAR, RELA, and FAS), 3 miRNAs (hsa-miR-320b, hsa-miR-22-3p, and hsa-miR-625-5p), and 35 lncRNAs was constructed by integrating data from literature review and analyzing the predicted miRNAs and lncRNAs. Moreover, the quantitative real-time PCR data revealed a pronounced upregulation of Fas cell surface death receptor. The identification of 4 DE-ARGs, especially Fas cell surface death receptor, has shed light on their potential pivotal role in the pathogenesis of pediatric asthma. The established ceRNA network provides novel insights into the autophagy mechanism in asthma and suggests promising avenues for the development of potential therapeutic strategies.
Collapse
Affiliation(s)
- Hao Zhu
- Department of Pediatrics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Jiao Shi
- Clinical Laboratory, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| | - Wen Li
- Department of Pediatrics, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning, China
| |
Collapse
|
11
|
Xie J, Wu Y, Tao Q, Liu H, Wang J, Zhang C, Zhou Y, Wei C, Chang Y, Jin Y, Ding Z. The role of lncRNA in the pathogenesis of chronic obstructive pulmonary disease. Heliyon 2023; 9:e22460. [PMID: 38034626 PMCID: PMC10687241 DOI: 10.1016/j.heliyon.2023.e22460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by progressive and irreversible airflow obstruction with abnormal lung function. Because its pathogenesis involves multiple aspects of oxidative stress, immunity and inflammation, apoptosis, airway and lung repair and destruction, the clinical approach to COPD treatment is not further updated. Therefore, it is crucial to discover a new means of COPD diagnosis and treatment. COPD etiology is associated with complex interactions between environmental and genetic determinants. Numerous genes are involved in the pathogenic process of this illness in research samples exposed to hazardous environmental conditions. Among them, Long non-coding RNAs (lncRNAs) have been reported to be involved in the molecular mechanisms of COPD development induced by different environmental exposures and genetic susceptibility encounters, and some potential lncRNA biomarkers have been identified as early diagnostic, disease course determination, and therapeutic targets for COPD. In this review, we summarize the expression profiles of the reported lncRNAs that have been reported in COPD studies related to environmental risk factors such as smoking and air pollution exposure and provided an overview of the roles of those lncRNAs in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Jing Xie
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yongkang Wu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Qing Tao
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hua Liu
- Anhui Institute for Food and Drug Control, Hefei, Anhui, China
| | - Jingjing Wang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chunwei Zhang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yuanzhi Zhou
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chengyan Wei
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, China
| | - Yong Jin
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Zhen Ding
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, China
| |
Collapse
|
12
|
Xiao B, Li L, Yao D, Mo B. Noncoding RNAs in asthmatic airway smooth muscle cells. Eur Respir Rev 2023; 32:32/168/220184. [PMID: 37076176 PMCID: PMC10113956 DOI: 10.1183/16000617.0184-2022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 02/07/2023] [Indexed: 04/21/2023] Open
Abstract
Asthma is a complex and heterogeneous airway disease caused by genetic, environmental and epigenetic factors treated with hormones and biologics. Irreversible pathological changes to airway smooth muscle cells (ASMCs) such as hyperplasia and hypertrophy can occur in asthmatic patients. Determining the mechanisms responsible is vital for preventing such changes. In recent years, noncoding RNAs (ncRNAs), especially microRNAs, long noncoding RNAs and circular RNAs, have been found to be associated with abnormalities of the ASMCs. This review highlights recent ncRNA research into ASMC pathologies. We present a schematic that illustrates the role of ncRNAs in pathophysiological changes to ASMCs that may be useful in future research in diagnostic and treatment strategies for patients with asthma.
Collapse
Affiliation(s)
- Bo Xiao
- Laboratory of Respiratory Disease, Affiliated Hospital of Guilin Medical University, Guilin, China
- Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- These authors contributed equally to this work
| | - Liangxian Li
- Guangxi Key Laboratory of Brain and Cognitive Neuroscience, Guilin Medical University, Guilin, China
- These authors contributed equally to this work
| | - Dong Yao
- Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- These authors contributed equally to this work
| | - Biwen Mo
- Key Laboratory of Respiratory Diseases, Education Department of Guangxi Zhuang Autonomous Region, Guilin, China
- Department of Respiratory and Critical Care Medicine, The Second Affiliated Hospital of Guilin Medical University, Guilin, China
- Key Laboratory of Glucose and Lipid Metabolism Disorders, Guangxi Health Commission, Guilin, China
| |
Collapse
|
13
|
Xu J, Zhang S, Li H, Bao Y, Du Y, Zhou Y, Zhao D, Liu F. LncRNA-AK007111 affects airway inflammation in asthma via the regulation of mast cell function. Int Immunopharmacol 2023; 121:110341. [PMID: 37301118 DOI: 10.1016/j.intimp.2023.110341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 05/05/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
Long noncoding RNAs (lncRNAs) are involved in gene transcription and pathophysiological processes of human diseases. Multiple lncRNAs have been shown to play important roles in the occurrence and development of asthma. This study aimed to explore the role of a novel lncRNA, lncRNA-AK007111, in asthma. Overexpression of lncRNA-AK007111 was induced in a mouse model of asthma via viral transfection, followed by the collection of alveolar lavage fluid and lung tissue for the detection of relevant inflammatory factors and pathological analysis of lung sections. Pulmonary resistance and respiratory dynamic compliance were measured using an animal pulmonary function analyzer. The number of mast cells sensitized by immunofluorescence was detected at the cellular level. The degree of degranulation of lncRNA-AK007111 after its knockdown was determined by detecting the level of β-hexosaminidase that was released and quantifying IL-6 and TNF-α using ELISA in a model of RBL-2H3 cells activated by immunoglobulin E plus antigen. Finally, we observed the migration ability of mast cells under a microscope. The results showed that in ovalbumin-sensitized mice, the upregulation of lncRNA-AK007111 promoted the infiltration of inflammatory cells in lung tissue, increased the number of total cells, eosinophils, and mast cells, upregulated IL-5 and IL-6 levels, and increased airway hyper-reactivity. Downregulation of lncRNA-AK007111 decreased the degranulation ability of IgE/Ag-activated mast cells and inhibited the expression of IL-6 and TNF-α; moreover, the migration ability of mast cells was significantly weakened. In conclusion, our study revealed that lncRNA-AK007111 plays an important role in asthma by modulating mast cell-related functions.
Collapse
Affiliation(s)
- Jiejing Xu
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Pediatrics, The Second People's Hospital of Changzhou, Affiliate Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Siqing Zhang
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China; Department of Pediatrics, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Huilin Li
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yaqing Bao
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yi Du
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yao Zhou
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Deyu Zhao
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Feng Liu
- Children's Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
14
|
Wang AL, Lahousse L, Dahlin A, Edris A, McGeachie M, Lutz SM, Sordillo JE, Brusselle G, Lasky-Su J, Weiss ST, Iribarren C, Lu MX, Tantisira KG, Wu AC. Novel genetic variants associated with inhaled corticosteroid treatment response in older adults with asthma. Thorax 2023; 78:432-441. [PMID: 35501119 PMCID: PMC9810110 DOI: 10.1136/thoraxjnl-2021-217674] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Accepted: 04/01/2022] [Indexed: 01/07/2023]
Abstract
INTRODUCTION Older adults have the greatest burden of asthma and poorest outcomes. The pharmacogenetics of inhaled corticosteroid (ICS) treatment response is not well studied in older adults. METHODS A genome-wide association study of ICS response was performed in asthmatics of European ancestry in Genetic Epidemiology Research on Adult Health and Aging (GERA) by fitting Cox proportional hazards regression models, followed by validation in the Mass General Brigham (MGB) Biobank and Rotterdam Study. ICS response was measured using two definitions in asthmatics on ICS treatment: (1) absence of oral corticosteroid (OCS) bursts using prescription records and (2) absence of asthma-related exacerbations using diagnosis codes. A fixed-effect meta-analysis was performed for each outcome. The validated single-nucleotide polymorphisms (SNPs) were functionally annotated to standard databases. RESULTS In 5710 subjects in GERA, 676 subjects in MGB Biobank, and 465 subjects in the Rotterdam Study, four novel SNPs on chromosome six near PTCHD4 validated across all cohorts and met genome-wide significance on meta-analysis for the OCS burst outcome. In 4541 subjects in GERA and 505 subjects in MGB Biobank, 152 SNPs with p<5 × 10-5 were validated across these two cohorts for the asthma-related exacerbation outcome. The validated SNPs included methylation and expression quantitative trait loci for CPED1, CRADD and DST for the OCS burst outcome and GM2A, SNW1, CACNA1C, DPH1, and RPS10 for the asthma-related exacerbation outcome. CONCLUSIONS Multiple novel SNPs associated with ICS response were identified in older adult asthmatics. Several SNPs annotated to genes previously associated with asthma and other airway or allergic diseases, including PTCHD4.
Collapse
Affiliation(s)
- Alberta L Wang
- Division of Allergy and Clinical Immunology, Brigham and Women's Hospital, Boston, Massachusetts, USA
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Lies Lahousse
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Amber Dahlin
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Ahmed Edris
- Department of Bioanalysis, Faculty of Pharmaceutical Sciences, Ghent University, Ghent, Belgium
| | - Michael McGeachie
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Sharon M Lutz
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Joanne E Sordillo
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| | - Guy Brusselle
- Department of Epidemiology, Erasmus Medical Center, Rotterdam, The Netherlands
- Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Respiratory Medicine, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jessica Lasky-Su
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Scott T Weiss
- Channing Division of Network Medicine, Brigham and Women's Hospital, Boston, Massachusetts, USA
| | - Carlos Iribarren
- Kaiser Permanente Division of Research, Kaiser Permanente, Oakland, California, USA
| | - Meng X Lu
- Kaiser Permanente Division of Research, Kaiser Permanente, Oakland, California, USA
| | - Kelan G Tantisira
- Division of Pediatric Respiratory Medicine, Rady's Children's Hospital-San Diego, University of California San Diego School of Medicine, San Diego, California, USA
| | - Ann C Wu
- PRecisiOn Medicine Translational Research (PROMoTeR) Center, Department of Population Medicine, Harvard Pilgrim Health Care Institute and Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
15
|
Long Non-Coding RNAs Expressed in the Peanut Allergy for Understanding the Pathophysiology of Peanut Allergy Rat Model. Foods 2022; 11:foods11233760. [PMID: 36496569 PMCID: PMC9740276 DOI: 10.3390/foods11233760] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 11/11/2022] [Accepted: 11/18/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Peanut allergy (PA) has become a clinical and public health problem, which is mainly regulated by genetics, immune responses, and environmental factors. Diagnosis and treatment for PA have always remained huge challenges due to its multiple triggers. Studies have shown that long non-coding RNAs (lncRNAs) play a critical role in the development of allergic diseases. METHOD AND RESULTS In the current study, we examined the plasma lncRNA expression profiles of peanut allergy Brown Norway rats and healthy controls and 496 differently expressed lncRNAs were identified, including 411 up-regulated genes and 85 down-regulated genes. We screened 8 lncRNAs based on the candidate principle and the candidates were verified in individual samples by quantitative real-time PCR. Then, the four lncRNA-based diagnostic model was established by least absolute shrinkage and selection operator (LASSO) and logistic regression, which was proved by area under the receiver operating characteristic curve (AUC). CONCLUSIONS In summary, we assessed the correlation between lncRNA expression levels and the diagnosis of peanut allergy, which may perform a vital role in guiding the management of peanut allergy.
Collapse
|
16
|
The Role of Noncoding RNA in Airway Allergic Diseases through Regulation of T Cell Subsets. Mediators Inflamm 2022; 2022:6125698. [PMID: 36248190 PMCID: PMC9553461 DOI: 10.1155/2022/6125698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 09/23/2022] [Indexed: 11/18/2022] Open
Abstract
Allergic rhinitis and asthma are common airway allergic diseases, the incidence of which has increased annually in recent years. The human body is frequently exposed to allergens and environmental irritants that trigger immune and inflammatory responses, resulting in altered gene expression. Mounting evidence suggested that epigenetic alterations were strongly associated with the progression and severity of allergic diseases. Noncoding RNAs (ncRNAs) are a class of transcribed RNA molecules that cannot be translated into polypeptides and consist of three major categories, microRNAs (miRNAs), long noncoding RNAs (lncRNAs), and circular RNAs (circRNAs). Previous studies showed that ncRNAs were involved in the physiopathological mechanisms of airway allergic diseases and contributed to their occurrence and development. This article reviews the current state of understanding of the role of noncoding RNAs in airway allergic diseases, highlights the limitations of recent studies, and outlines the prospects for further research to facilitate the clinical translation of noncoding RNAs as therapeutic targets and biomarkers.
Collapse
|
17
|
Liu X, Lin Y. Long non-coding RNA plasmacytoma variant translocation 1 correlates with higher inflammation, multiple organ injury and mortality risk in acute pancreatitis, especially in severe acute pancreatitis. Clin Res Hepatol Gastroenterol 2022; 46:101870. [PMID: 35108655 DOI: 10.1016/j.clinre.2022.101870] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 01/06/2022] [Accepted: 01/20/2022] [Indexed: 02/04/2023]
Abstract
BACKGROUND Long non-coding RNA plasmacytoma variant translocation 1 (lnc-PVT1) possesses a good ability to regulate inflammation as well as multiple organ injury via multiple pathways, and clinically exacerbates severe acute pancreatitis (SAP) via autophagy. This study aimed to further assess the correlation of lnc-PVT1 with inflammation, multiple disease assessment scales, and prognostication in acute pancreatitis (AP) patients. METHODS Peripheral blood mononuclear cell (PBMC) samples were collected from 98 AP patients (within 24 h after admission) and 50 healthy controls (HCs). lnc-PVT1 in PBMC samples was examined by reverse transcription-quantitive polymerase chain reaction. Multiple AP assessments, C-reactive protein (CRP) level, and in-hospital deaths were evaluated or recorded. RESULTS lnc-PVT1 was overexpressed in AP patients compared with HCs; it was also positively correlated with Ranson's score, acute pathologic and chronic health evaluation II (APACHE II) score, sequential organ failure assessment (SOFA) score, and CRP level in AP patients. Besides, lnc-PVT1 disclosed a good predictive value for higher in-hospital mortality in AP patients (the area under the curve: 0.838, 95% confidence interval: 0.708-0.968). Lastly, lnc-PVT1 was generally correlated with CRP level as well as SOFA score among mild AP, moderate-severe AP, and SAP subgroups, especially in SAP subgroup; it was also correlated with higher mortality risk in SAP subgroup, but not in mild AP or moderate-severe AP subgroup. CONCLUSION lnc-PVT1 is associated with CRP level, SOFA score, and higher mortality risk in AP patients, especially in SAP patients, indicating its potential as a biomarker for AP.
Collapse
Affiliation(s)
- Xue Liu
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou 341000, Jiangxi, China
| | - Ye Lin
- Department of Gastroenterology, The Affiliated Ganzhou Hospital of Nanchang University, Ganzhou 341000, Jiangxi, China.
| |
Collapse
|
18
|
The Effect of lncRNA-PVT1 on Liver Cancer Rats by Regulating the Expression of MMP9. BIOMED RESEARCH INTERNATIONAL 2022; 2022:4126839. [PMID: 36164442 PMCID: PMC9509214 DOI: 10.1155/2022/4126839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 07/12/2022] [Accepted: 08/13/2022] [Indexed: 11/26/2022]
Abstract
As a malignant tumor, liver cancer has a high lethality rate. The research on the pathogenesis of liver cancer is the key to the treatment of liver cancer. The latest research claims that lncRNA-PVT1 as a tumor gene participates in the generation and development of tumors by regulating matrix metalloproteinase MMP9. The purpose of this article is to explore the specific effect and mechanism of lncRNA-PVT1 on liver cancer rats by regulating the expression of MMP9. In this article, 50 rats are used as experimental subjects, the rats are divided into control group and observation group, and the liver cancer cell line HepG2 is cultured. The transplanted tumor liver cancer model was constructed by transfection of hair, the expression of lncRNA-PVT1 in the observation group was reduced by knockdown method, the expression levels and changes of lncRNA-PVT1 and MMP9 in the two groups were detected by PCR fluorescence method, and the difference between lncRNA-PVT1 and MMP9 was analyzed. The MTT method was used to detect the proliferation, migration, and invasion capabilities of the two groups of liver cancer cells (LCC) and to explore the effect of lncRNA-PVT1 on rat LCC by regulating the expression of MMP9. The results of the study showed that after knocking down the expression of lncRNA-PVT1 in the observation group, the expression of MMP9 also decreased. At the same time, the migration rate of LCC HepG2 decreased by 27.4%, the level of invasion ability decreased by 21.6%, and the proliferation rate of LCC decreased by 17.8%. Therefore, it can be seen that lncRNA-PVT1 plays a positive regulatory role on the expression of MMP9, and the expression of MMP9 promotes the proliferation, migration, and invasion of rat LCC.
Collapse
|
19
|
Xiang LL, Wan QQ, Wang YM, He SJ, Xu WJ, Ding M, Zhang JJ, Sun YL, Dong X, Zhou Y, Cui YB, Gao YD. IL-13 Regulates Orai1 Expression in Human Bronchial Smooth Muscle Cells and Airway Remodeling in Asthma Mice Model via LncRNA H19. J Asthma Allergy 2022; 15:1245-1261. [PMID: 36101840 PMCID: PMC9464454 DOI: 10.2147/jaa.s360381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 08/20/2022] [Indexed: 11/23/2022] Open
Abstract
Background Increased proliferation and hypertrophy of airway smooth muscle cells (ASMCs) contribute substantially to airway remodeling in asthma. Interleukin (IL)-13 regulates ASMC proliferation by increasing Orai1 expression, the pore-forming subunit of store-operated Ca2+ entry (SOCE). The underlying mechanisms of this effect are not fully understood. Methods Bioinformatic analysis identified an interaction between microRNA 93-5p (miR-93-5p) and long non-coding RNA (lncRNA) H19, and between miR-93-5p and Orai1. RNA interference was used to investigate H19 knockdown on IL-13-induced proliferation and migration of in vitro cultured human bronchial smooth muscle cells (hBSMCs). Functional relevance of H19 in airway inflammation and airway remodeling was investigated in murine models of acute and chronic asthma. Results IL-13 concentration-dependently increased the expression of H19 and Orai1 and decreased the expression of miR-93-5p in hBSMCs. H19 knockdown partly reversed the effects of IL-13 on the expression of miR-93-5p and Orai1 and attenuated the proliferation and migration of hBSMCs promoted by IL-13. IL-13-promoted expression of Orai1 was attenuated by miR-93-5p mimic and increased by miR-93-5p inhibitor. IL-13-promoted proliferation of hBSMCs was increased by miR-93-5p inhibitor but not affected by miR-93-5p mimic, whereas IL-13-promoted migration of hBSMCs was increased by miR-93-5p inhibitor and attenuated by miR-93-5p mimic. The inhibiting effect of H19 knockdown on IL-13-induced Orai1 expression and the proliferation and migration of hBSMCs was counteracted by miR-93-5p inhibitor but only marginally or not impacted by miR-93-5p mimic. The expression of H19 and Orai1 was higher in the lungs of asthmatic mice than in control mice. In asthmatic mice, H19 siRNA reduced Orai1 expression, inflammatory cell infiltration, goblet cell hyperplasia, collagen deposition and smooth muscle mass in the lungs. Conclusion H19 may mediate the effects of IL-13 on Orai1 expression by inhibition of miR-93-5p in hBSMCs. H19 may be a therapeutic target for airway inflammation and airway remodeling.
Collapse
Affiliation(s)
- Lin-Li Xiang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Qian-Qian Wan
- Department of Rheumatology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Yi-Min Wang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Shao-Jun He
- Department of Respiratory and Critical Care Medicine Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Wen-Juan Xu
- Department of Respiratory and Critical Care Medicine Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China
| | - Mei Ding
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, People's Republic of China
| | - Jin-Jin Zhang
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, People's Republic of China
| | - Yuan-Li Sun
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, People's Republic of China
| | - Xiang Dong
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, People's Republic of China
| | - Ying Zhou
- Department of Pediatrics Laboratory, The Affiliated Wuxi Children's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Yu-Bao Cui
- Department of Clinical Laboratory, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi, People's Republic of China
| | - Ya-Dong Gao
- Department of Allergology, Zhongnan Hospital of Wuhan University, Wuhan, People's Republic of China.,Hubei Province Key Laboratory of Allergy and Immunology, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
20
|
Huang Y, Qiu C. Research advances in airway remodeling in asthma: a narrative review. ANNALS OF TRANSLATIONAL MEDICINE 2022; 10:1023. [PMID: 36267708 PMCID: PMC9577744 DOI: 10.21037/atm-22-2835] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 09/06/2022] [Indexed: 12/04/2022]
Abstract
Background and Objective Asthma is a common chronic disorder of the airway, and its disability and mortality rates continue to increase each year. Due to the lack of an ideal treatment, asthma control in China remains unsatisfactory. Airway remodeling is the pathological basis for the eventual development of the fixed airflow limitation in asthmatic patients. Early diagnosis and the prevention of airway remodeling has the potential to decrease disease severity, to improve control, and to prevent disease expression. Methods This article presents an overview. The literature was combed through via CNKi and PubMed according to the listed keywords. We considered Chinese and English original publications (basic science and clinical), reviews and abstracts of 21th Century. Key Content and Findings We review the pathological features and pathogenesis of, and the interventional treatment options for airway remodeling in asthmatic patients, emphasizing the importance of airway remodeling in asthma and providing novel insights into the prevention and control of asthma. Conclusions Thus, there have been research advances in airway remodeling, especially in the areas of slowing down or reversing airway remodeling. As growing studies showed, treating airway remodeling is a promising strategy in preventing the occurrence and progression of asthma. Breakthroughs in these difficulties airway remodeling still facing will open up new avenues in the research and treatment of asthma.
Collapse
Affiliation(s)
- Yanmei Huang
- Department of Respiratory and Critical Care Medicine, The Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen, China
| | - Chen Qiu
- Department of Respiratory and Critical Care Medicine, Shenzhen People’s Hospital, Shenzhen, China
| |
Collapse
|
21
|
Li Y, Li S, Li R, Xu H. LncRNA PVT1 upregulates FBN1 by sponging miR-30b-5p to aggravate pulpitis. Mol Cell Toxicol 2022. [DOI: 10.1007/s13273-022-00264-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
22
|
Qiao X, Hou G, He YL, Song DF, An Y, Altawil A, Zhou XM, Wang QY, Kang J, Yin Y. The Novel Regulatory Role of the lncRNA–miRNA–mRNA Axis in Chronic Inflammatory Airway Diseases. Front Mol Biosci 2022; 9:927549. [PMID: 35769905 PMCID: PMC9234692 DOI: 10.3389/fmolb.2022.927549] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2022] [Accepted: 05/19/2022] [Indexed: 12/28/2022] Open
Abstract
Chronic inflammatory airway diseases, characterized by airway inflammation and airway remodelling, are increasing as a cause of morbidity and mortality for all age groups and races across the world. The underlying molecular mechanisms involved in chronic inflammatory airway diseases have not been fully explored. MicroRNAs (miRNAs) and long noncoding RNAs (lncRNAs) have recently attracted much attention for their roles in the regulation of a variety of biological processes. A number of studies have confirmed that both lncRNAs and miRNAs can regulate the initiation and progression of chronic airway diseases by targeting mRNAs and regulating different cellular processes, such as proliferation, apoptosis, inflammation, migration, and epithelial–mesenchymal transition (EMT). Recently, accumulative evidence has shown that the novel regulatory mechanism underlying the interaction among lncRNAs, miRNAs and messenger RNAs (mRNAs) plays a critical role in the pathophysiological processes of chronic inflammatory airway diseases. In this review, we comprehensively summarized the regulatory roles of the lncRNA–miRNA–mRNA network in different cell types and their potential roles as biomarkers, indicators of comorbidities or therapeutic targets for chronic inflammatory airway diseases, particularly chronic obstructive pulmonary disease (COPD) and asthma.
Collapse
Affiliation(s)
- Xin Qiao
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Gang Hou
- Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, China
| | - Yu-Lin He
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Dong-Fang Song
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yi An
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Abdullah Altawil
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xiao-Ming Zhou
- Respiratory Department, Center for Pulmonary Vascular Diseases, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- *Correspondence: Xiao-Ming Zhou, ; Yan Yin,
| | - Qiu-Yue Wang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Jian Kang
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
| | - Yan Yin
- Department of Pulmonary and Critical Care Medicine, First Affiliated Hospital of China Medical University, Shenyang, China
- *Correspondence: Xiao-Ming Zhou, ; Yan Yin,
| |
Collapse
|
23
|
Ding L, Liu GL, Lu L, Ge L, Wang JY. circ_CSNK1E modulates airway smooth muscle cells proliferation and migration via miR-34a-5p/VAMP2 axis in asthma. Cell Signal 2022; 95:110340. [PMID: 35483563 DOI: 10.1016/j.cellsig.2022.110340] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Accepted: 04/21/2022] [Indexed: 12/22/2022]
Abstract
BACKGROUND Excessive proliferation and migration of airway smooth muscle cells (ASMCs) directly lead to airway remodeling in asthma. However, the role of circular RNAs (circRNAs) in airway remodeling remains unclear. This study aimed to investigate the regulatory role and mechanism of circ_CSNK1E in ASMCs proliferation and migration. METHODS In this study, RNA-sequencing was used to analyze cicRNAs expression in asthma samples. ASMCs were treated with 25 ng/mL PDGF-BB to establish a model of asthma in vitro. Then, we used RT-qPCR to assess circRNAs, microRNAs (miRNAs) and messenger RNAs (mRNAs) expression. Besides, CCK-8, colony formation, wound healing and transwell chamber assays were carried out to explore cell proliferation and migration. Subcellular localization assay was used to detect the location of circRNA. Next, bioinformatics, luciferase reporter and RIP assays were performed to evaluate the relationship among circ_CSNK1E, miRNA-34a-5p and VAMP2. RESULTS circ_CSNK1E expression was found to be significantly up-regulated in asthma samples and PDGF-BB-induced ASMCs. Functional experiments revealed that inhibition of circRNA_CSNK1E suppressed proliferation and migration of ASMCs stimulated by PDGF-BB. Next, we found that circRNA_CSNK1E served as a sponge for miR-34a-5p in ASMCs, and miR-34a-5p mimic suppressed proliferation and migration of ASMCs. Moreover, VAMP2 was confirmed as a direct target of miR-34a-5p. At last, inhibition of circRNA_CSNK1E suppressed proliferation and migration of ASMCs stimulated by PDGF-BB through miR-34a-5p/VAMP2 axis. CONCLUSION Collectively, these findings clarified the importance of circ_CSNK1E/miRNA-34a-5p/VAMP2 axis for the proliferation and migration of ASMCs. These indicated that inhibition of circ_CSNK1E might be a potential target for the treatment of airway remodeling in asthma.
Collapse
Affiliation(s)
- Ling Ding
- Department of Pediatrics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321. Zhongshan Road, 210008 Nanjing, Jiangsu, China; Department of Pediatrics, The Fourth Affiliated Hospital of Nanjing Medical University, No. 298, Nanpu Road, 210031 Nanjing, Jiangsu, China
| | - Guang-Ling Liu
- Department of Pediatrics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321. Zhongshan Road, 210008 Nanjing, Jiangsu, China
| | - Lu Lu
- Department of Pediatrics, The Second Affiliated Hospital of Nanjing Medical University, No. 262. Zhongshan North Road, 210003 Nanjing, Jiangsu, China
| | - Lei Ge
- Department of Pediatrics, The Second Affiliated Hospital of Nanjing Medical University, No. 262. Zhongshan North Road, 210003 Nanjing, Jiangsu, China
| | - Jin-Ya Wang
- Department of Pediatrics, Nanjing Drum Tower Hospital, the Affiliated Hospital of Nanjing University Medical School, No.321. Zhongshan Road, 210008 Nanjing, Jiangsu, China.
| |
Collapse
|
24
|
Multi-omics research in sarcopenia: Current progress and future prospects. Ageing Res Rev 2022; 76:101576. [PMID: 35104630 DOI: 10.1016/j.arr.2022.101576] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 12/13/2021] [Accepted: 01/26/2022] [Indexed: 12/17/2022]
Abstract
Sarcopenia is a systemic disease with progressive and generalized skeletal muscle dysfunction defined by age-related low muscle mass, high content of muscle slow fibers, and low muscle function. Muscle phenotypes and sarcopenia risk are heritable; however, the genetic architecture and molecular mechanisms underlying sarcopenia remain largely unclear. In recent years, significant progress has been made in determining susceptibility loci using genome-wide association studies. In addition, recent advances in omics techniques, including genomics, epigenomics, transcriptomics, proteomics, and metabolomics, offer new opportunities to identify novel targets to help us understand the pathophysiology of sarcopenia. However, each individual technology cannot capture the entire view of the biological complexity of this disorder, while integrative multi-omics analyses may be able to reveal new insights. Here, we review the latest findings of multi-omics studies for sarcopenia and provide an in-depth summary of our current understanding of sarcopenia pathogenesis. Leveraging multi-omics data could give us a holistic understanding of sarcopenia etiology that may lead to new clinical applications. This review offers guidance and recommendations for fundamental research, innovative perspectives, and preventative and therapeutic interventions for sarcopenia.
Collapse
|
25
|
Ma C, Wang S, Cao Y, Tang W, Wuniqiemu T, Teng F, Zhu X, Wei Y, Dong J. Screening and Verification of Differentially Expressed Long Non-coding RNAs in the Peripheral Blood of Patients With asthma. Front Pharmacol 2022; 13:834009. [PMID: 35273507 PMCID: PMC8902465 DOI: 10.3389/fphar.2022.834009] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2021] [Accepted: 01/19/2022] [Indexed: 12/21/2022] Open
Abstract
Growing evidence suggests that long non-coding RNAs (lncRNAs) play a key role in the pathogenesis of asthma. Although some differentially expressed lncRNAs have been identified in asthmatic patients, many asthma-related lncRNAs have not been annotated. In the present study, six patients and three healthy subjects were randomly selected from 34 asthmatic patients and 17 healthy subjects. Second-generation high-throughput sequencing was performed on their peripheral blood samples. There were 1,137 differentially expressed lncRNAs in the asthma patients compared to in the healthy controls, of which 485 were upregulated and 652 were downregulated. The top 30 enriched GO and KEGG terms were identified, and the cytosolic ribosome (GO:0022626) and ribosome (hsa03010) were associated with the most differentially expressed lncRNAs. The top 10 differentially expressed lncRNAs associated with asthma were verified by an lncRNA-mRNA co-expression network and RT-qPCR. Seven of the these (NONHSAT015495.2, MSTRG.71212.2, NONHSAT163272.1, NONHSAT181891.1, NONHSAT190964.1, ENST00000564809, and NONHSAT076890.2) were down-regulated in the peripheral blood of asthmatic patients, which was consistent with the sequencing results. Three patients and three healthy subjects were randomly selected from the remaining subjects to verify these seven lncRNAs by RT-qPCR, which further confirmed the sequencing results. Public database GSE106230 was also in agreement with the FPKM (Fragments Per kilobase of exon model per Million mapped reads) trends of ENST00000564809, NONHSAT015495.2, NONHSAT181891.1, and NONHSAT190964.1. In conclusion, the present study identified seven lncRNAs that may serve as potential biological markers for asthma.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Shiyuan Wang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Yuxue Cao
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Weifeng Tang
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Tulake Wuniqiemu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Fangzhou Teng
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Xueyi Zhu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Ying Wei
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| | - Jingcheng Dong
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai, China.,Institutes of Integrative Medicine, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Gysens F, Mestdagh P, de Bony de Lavergne E, Maes T. Unlocking the secrets of long non-coding RNAs in asthma. Thorax 2022; 77:514-522. [PMID: 35246486 PMCID: PMC9016255 DOI: 10.1136/thoraxjnl-2021-218359] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 01/21/2022] [Indexed: 12/15/2022]
Abstract
Asthma is a very heterozygous disease, divided in subtypes, such as eosinophilic and neutrophilic asthma. Phenotyping and endotyping of patients, especially patients with severe asthma who are refractory to standard treatment, are crucial in asthma management and are based on a combination of clinical and biological features. Nevertheless, the quest remains to find better biomarkers that distinguish asthma subtypes in a more clear and objective manner and to find new therapeutic targets to treat people with therapy-resistant asthma. In the past, research to identify asthma subtypes mainly focused on expression profiles of protein-coding genes. However, advances in RNA-sequencing technologies and the discovery of non-coding RNAs as important post-transcriptional regulators have provided an entire new field of research opportunities in asthma. This review focusses on long non-coding RNAs (lncRNAs) in asthma; these are non-coding RNAs with a length of more than 200 nucleotides. Many lncRNAs are differentially expressed in asthma, and several have been associated with asthma severity or inflammatory phenotype. Moreover, in vivo and in vitro functional studies have identified the mechanisms of action of specific lncRNAs. Although lncRNAs remain not widely studied in asthma, the current studies show the potential of lncRNAs as biomarkers and therapeutic targets as well as the need for further research.
Collapse
Affiliation(s)
- Fien Gysens
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | - Pieter Mestdagh
- OncoRNALab, Department of Biomolecular Medicine, Ghent University, Ghent, Belgium
| | | | - Tania Maes
- Laboratory for Translational Research in Obstructive Pulmonary Diseases, Department of Respiratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
27
|
王 冲, 黄 海, 王 宏, 李 椿, 刘 晓. [Research progress on the relationship between lncRNA and the pathogenesis of allergic rhinitis]. LIN CHUANG ER BI YAN HOU TOU JING WAI KE ZA ZHI = JOURNAL OF CLINICAL OTORHINOLARYNGOLOGY, HEAD, AND NECK SURGERY 2022; 36:233-238. [PMID: 35193349 PMCID: PMC10128298 DOI: 10.13201/j.issn.2096-7993.2022.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 10/11/2021] [Indexed: 04/30/2023]
Abstract
Long non-coding RNA(lncRNA) belongs to the category of non-coding RNA, which length exceeds 200 nucleotides and can hardly encode the expression of proteins. Based on the data from several clinical researches, it is considered that lncRNA not only plays the biological role in epigenetics, transcriptional and post-transcriptional levels, but also abnormally expresses in inflammatory response and the related diseases. In recent years, with the intensive study of gene expression regulation of allergic rhinitis(AR), it has been found that a variety of non-coding RNA, including lncRNA, have close relationship with the occurrence and development of AR. This review mainly summarized the biological function, immunomodulatory effect of lncRNA and the relationship between the lncRNA and the pathogenesis of AR, providing new thoughts and strategies for the further research, prevention as well as the treatment of AR.
Collapse
|
28
|
Chen J, Ren H, Liu B. Evaluating the potency of blood long noncoding RNA PVT1 as candidate biomarker reflecting inflammation, multiple organ dysfunction, and mortality risk in sepsis patients. J Clin Lab Anal 2022; 36:e24268. [PMID: 35119126 PMCID: PMC8906045 DOI: 10.1002/jcla.24268] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Revised: 01/17/2022] [Accepted: 01/21/2022] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE Long noncoding RNA plasmacytoma variant translocation 1 (lnc-PVT1) promotes septic inflammation and organ injuries via multiple ways, while its clinical engagement in sepsis management is indistinct. This study aimed to investigate its relationship with inflammation, multiple organ dysfunction, and mortality risk in sepsis patients. METHODS Sepsis patients and age-/gender-matched healthy controls were enrolled; their lnc-PVT1 expression in plasma were detected by RT-qPCR. For sepsis patients only, the inflammatory cytokine levels (tumor necrosis factor (TNF)-α, interleukin (IL)-1β, and IL-17A) in plasma were detected by ELISA. According to the survival data during 28-day follow-up, sepsis patients were divided into sepsis survivors and sepsis deaths. RESULTS Lnc-PVT1 expression was increased in sepsis patients (N = 157) compared with healthy controls (N = 80) (p < 0.001). In sepsis patients, lnc-PVT1 was linked with higher acute physiology and chronic health evaluation II (APACHEII) score (p = 0.001), total sequential organ failure assessment (SOFA) score, and its most subitems (SOFA-respiratory system, SOFA-coagulation, SOFA-liver, SOFA-cardiovascular system, and SOFA-renal system scores) (all p < 0.01), but not SOFA-nervous system score (p = 0.091); it did not relate to primary infection sites either (p = 0.204). Furthermore, lnc-PVT1 correlated with increased C-reactive protein, TNF-α, IL-1β, and IL-17 in sepsis patients (all p < 0.01). Additionally, lnc-PVT1 expression was higher in sepsis deaths than that in sepsis survivors (p < 0.001), following receiver-operating characteristic curve disclosed that lnc-PVT1 predicted 28-day septic mortality risk (area under the curve: 0.789, 95% confidence interval: 0.702-0.875). CONCLUSION Circulating lnc-PVT1 exhibits the potential as a biomarker in sepsis patients to inform inflammation, multiple organ dysfunction, and mortality risk.
Collapse
Affiliation(s)
- Jing Chen
- Department of Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, China
| | - Haibo Ren
- Department of Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, China
| | - Bo Liu
- Department of Critical Care Medicine, Wuhan Asia General Hospital, Wuhan, China
| |
Collapse
|
29
|
Panganiban RA, Lu Q. A Long Non-Coding RNA "lnc"ed to Asthma Genetics. Am J Respir Cell Mol Biol 2022; 66:243-244. [PMID: 35030310 PMCID: PMC8937238 DOI: 10.1165/rcmb.2021-0534ed] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Affiliation(s)
- Ronald A Panganiban
- Harvard University T H Chan School of Public Health, 1857, Boston, Massachusetts, United States
| | - Quan Lu
- Harvard University T H Chan School of Public Health, 1857, Boston, Massachusetts, United States;
| |
Collapse
|
30
|
Liang J, Liu XH, Chen XM, Song XL, Li W, Huang Y. Emerging Roles of Non-Coding RNAs in Childhood Asthma. Front Pharmacol 2022; 13:856104. [PMID: 35656293 PMCID: PMC9152219 DOI: 10.3389/fphar.2022.856104] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease in children characterized by airway inflammation, airway hyperresponsiveness and airway remodeling. Childhood asthma is usually associated with allergy and atopy, unlike adult asthma, which is commonly associated with obesity, smoking, etc. The pathogenesis and diagnosis of childhood asthma also remains more challenging than adult asthma, such as many diseases showing similar symptoms may coexist and be confused with asthma. In terms of the treatment, although most childhood asthma can potentially be self-managed and controlled with drugs, approximately 5-10% of children suffer from severe uncontrolled asthma, which carries significant health and socioeconomic burdens. Therefore, it is necessary to explore the pathogenesis of childhood asthma from a new perspective. Studies have revealed that non-coding RNAs (ncRNAs) are involved in the regulation of respiratory diseases. In addition, altered expression of ncRNAs in blood, and in condensate of sputum or exhalation affects the progression of asthma via regulating immune response. In this review, we outline the regulation and pathogenesis of asthma and summarize the role of ncRNAs in childhood asthma. We also hold promise that ncRNAs may be used for the development of biomarkers and support a new therapeutic strategy for childhood asthma.
Collapse
Affiliation(s)
- Juan Liang
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Xiao-Hua Liu
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Xue-Mei Chen
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Xiu-Ling Song
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China.,Graduate School of Guangdong Medical University, Zhanjiang, China
| | - Wen Li
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| | - Yuge Huang
- Department of Pediatrics, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
| |
Collapse
|
31
|
Ran MY, Yuan Z, Fan CT, Ke Z, Wang XX, Sun JY, Su DJ. Multiplex-Heterogeneous Network-Based Capturing Potential SNP "Switches" of Pathways Associating With Diverse Disease Characteristics of Asthma. Front Cell Dev Biol 2022; 9:744932. [PMID: 34970542 PMCID: PMC8712737 DOI: 10.3389/fcell.2021.744932] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2021] [Accepted: 11/17/2021] [Indexed: 11/25/2022] Open
Abstract
Asthma is a complex heterogeneous respiratory disorder. In recent years nubbly regions of the role of genetic variants and transcriptome including mRNAs, microRNAs, and long non-coding RNAs in the pathogenesis of asthma have been separately excavated and reported. However, how to systematically integrate and decode this scattered information remains unclear. Further exploration would improve understanding of the internal communication of asthma. To excavate new insights into the pathogenesis of asthma, we ascertained three asthma characteristics according to reviews, airway inflammation, airway hyperresponsiveness, and airway remodeling. We manually created a contemporary catalog of corresponding risk transcriptome, including mRNAs, miRNAs, and lncRNAs. MIMP is a multiplex-heterogeneous networks-based approach, measuring the relevance of disease characteristics to the pathway by examining the similarity between the determined vectors of risk transcriptome and pathways in the same low-dimensional vector space. It was developed to enable a more concentrated and in-depth exploration of potential pathways. We integrated experimentally validated competing endogenous RNA regulatory information and the SNPs with significant pathways into the ceRNA-mediated SNP switching pathway network (CSSPN) to analyze ceRNA regulation of pathways and the role of SNP in these dysfunctions. We discovered 11 crucial ceRNA regulations concerning asthma disease feature pathway and propose a potential mechanism of ceRNA regulatory SNP → gene → pathway → disease feature effecting asthma pathogenesis, especially for MALAT1 (rs765499057/rs764699354/rs189435941) → hsa-miR-155 → IL13 (rs201185816/rs1000978586/rs202101165) → Interleukin-4 and Interleukin-13 signaling → inflammation/airway remodeling and MALAT1 (rs765499057/rs764699354/rs189435941) → hsa-miR-155 → IL17RB (rs948046241) → Interleukin-17 signaling (airway remodeling)/Cytokine-cytokine receptor interaction (inflammation). This study showed a systematic and propagable workflow for capturing the potential SNP “switch” of asthma through text and database mining and provides further information on the pathogenesis of asthma.
Collapse
Affiliation(s)
- Ming-Yu Ran
- Department of College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Zhang Yuan
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Chui-Ting Fan
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhou Ke
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin-Xing Wang
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jia-Yuan Sun
- Shanghai Chest Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Dong-Ju Su
- Department of Respiratory, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| |
Collapse
|
32
|
Prognostic Value of lncRNA PVT1 for Patients with Gastric Cancer: A Meta-Analysis. DISEASE MARKERS 2021; 2021:5595965. [PMID: 34900027 PMCID: PMC8660201 DOI: 10.1155/2021/5595965] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 09/24/2021] [Accepted: 10/15/2021] [Indexed: 02/07/2023]
Abstract
Objective To evaluate the prognostic value of lncRNA PVT1 for patients with gastric cancer. Methods A comprehensive literature searching was performed in PubMed, Cochrane Library, Web of Science, Embase, CNKI, CBM, and Wanfang Database to identify published studies on the expression level of lncRNA PVT1 in human gastric cancer. STATA 12.0 was conducted to perform the meta-analysis. Clinical outcomes including patients' age, genders, TNM stage, OS, and DFS were assessed in the study. Results A total of 8 studies involving 747 patients were included in this meta-analysis. The results of meta-analysis showed that higher expression level of lncRNA PVT1 was associated with GC patients' gender (for male: OR = 2.27, 95% CI: 1.67~3.07, P = 0.000), invasion depth (for T3~4: OR = 3.98, 95% CI: 2.85~5.56, P = 0.000), poorer OS (HR = 1.68, 95% CI: 1.43~1.97, P = 0.000), and DFS (HR = 1.74, 95% CI: 1.44~2.08, P = 0.000). Conclusion Higher expression level of lncRNA PVT1 is significantly associated with GC patients' gender, invasion depth, poorer OS, and worse DFS. lncRNA PVT1 might act as a novel predictive biomarker of poor prognosis and clinicopathological characteristics for gastric cancer.
Collapse
|
33
|
Gedefaw L, Ullah S, Lee TMH, Yip SP, Huang CL. Targeting Inflammasome Activation in COVID-19: Delivery of RNA Interference-Based Therapeutic Molecules. Biomedicines 2021; 9:1823. [PMID: 34944639 PMCID: PMC8698532 DOI: 10.3390/biomedicines9121823] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 11/22/2021] [Accepted: 11/30/2021] [Indexed: 02/07/2023] Open
Abstract
Mortality and morbidity associated with COVID-19 continue to be significantly high worldwide, owing to the absence of effective treatment strategies. The emergence of different variants of SARS-CoV-2 is also a considerable source of concern and has led to challenges in the development of better prevention and treatment strategies, including vaccines. Immune dysregulation due to pro-inflammatory mediators has worsened the situation in COVID-19 patients. Inflammasomes play a critical role in modulating pro-inflammatory cytokines in the pathogenesis of COVID-19 and their activation is associated with poor clinical outcomes. Numerous preclinical and clinical trials for COVID-19 treatment using different approaches are currently underway. Targeting different inflammasomes to reduce the cytokine storm, and its associated complications, in COVID-19 patients is a new area of research. Non-coding RNAs, targeting inflammasome activation, may serve as an effective treatment strategy. However, the efficacy of these therapeutic agents is highly dependent on the delivery system. MicroRNAs and long non-coding RNAs, in conjunction with an efficient delivery vehicle, present a potential strategy for regulating NLRP3 activity through various RNA interference (RNAi) mechanisms. In this regard, the use of nanomaterials and other vehicle types for the delivery of RNAi-based therapeutic molecules for COVID-19 may serve as a novel approach for enhancing drug efficacy. The present review briefly summarizes immune dysregulation and its consequences, the roles of different non-coding RNAs in regulating the NLRP3 inflammasome, distinct types of vectors for their delivery, and potential therapeutic targets of microRNA for treatment of COVID-19.
Collapse
Affiliation(s)
- Lealem Gedefaw
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (S.U.)
| | - Sami Ullah
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (S.U.)
| | - Thomas M. H. Lee
- Department of Biomedical Engineering, The Hong Kong Polytechnic University, Hong Kong, China;
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (S.U.)
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (S.U.)
- Research Institute for Future Food, The Hong Kong Polytechnic University, Hong Kong, China
| |
Collapse
|
34
|
Soni DK, Biswas R. Role of Non-Coding RNAs in Post-Transcriptional Regulation of Lung Diseases. Front Genet 2021; 12:767348. [PMID: 34819948 PMCID: PMC8606426 DOI: 10.3389/fgene.2021.767348] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 10/25/2021] [Indexed: 12/16/2022] Open
Abstract
Non-coding RNAs (ncRNAs), notably microRNAs (miRNAs) and long noncoding RNAs (lncRNAs), have recently gained increasing consideration because of their versatile role as key regulators of gene expression. They adopt diverse mechanisms to regulate transcription and translation, and thereby, the function of the protein, which is associated with several major biological processes. For example, proliferation, differentiation, apoptosis, and metabolic pathways demand fine-tuning for the precise development of a specific tissue or organ. The deregulation of ncRNA expression is concomitant with multiple diseases, including lung diseases. This review highlights recent advances in the post-transcriptional regulation of miRNAs and lncRNAs in lung diseases such as asthma, chronic obstructive pulmonary disease, cystic fibrosis, and idiopathic pulmonary fibrosis. Further, we also discuss the emerging role of ncRNAs as biomarkers as well as therapeutic targets for lung diseases. However, more investigations are required to explore miRNAs and lncRNAs interaction, and their function in the regulation of mRNA expression. Understanding these mechanisms might lead to early diagnosis and the development of novel therapeutics for lung diseases.
Collapse
Affiliation(s)
- Dharmendra Kumar Soni
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| | - Roopa Biswas
- Department of Anatomy, Physiology and Genetics, School of Medicine, Uniformed Services University of the Health Sciences, Bethesda, MD, United States
| |
Collapse
|
35
|
Wang X, Chen J. Long non-coding RNA TUG1 promotes proliferation and migration in PDGF-BB-stimulated HASMCs by regulating miR-216a-3p/SMURF2 axis. BMC Mol Cell Biol 2021; 22:56. [PMID: 34749662 PMCID: PMC8573901 DOI: 10.1186/s12860-021-00396-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Accepted: 10/15/2021] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND Abnormal proliferation and migration of human airway smooth muscle cells (HASMCs) play an important role in the development of childhood asthma. Long non-coding RNAs (lncRNAs) have been demonstrated to participate in HASMC proliferation and migration. We aimed to explore more effects and molecular mechanism of taurine upregulated gene 1 (TUG1) in childhood asthma. RESULTS TUG1 and SMURF2 were overexpressed and miR-216a-3p was downregulated in childhood asthma patients and PDGF-BB-stimulated HASMCs. TUG1 knockdown attenuated PDGF-BB-triggered proliferation and migration of HASMCs. MiR-216a-3p was targeted by TUG1, and miR-216a-3p suppression counteracted the repressive effects of TUG1 interference on proliferation and migration in PDGF-BB-treated HASMCs. SMURF2 was a downstream target of miR-216a-3p, and SMURF2 upregulation abated the inhibiting effects of miR-216a-3p on migration and proliferation in PDGF-BB-exposed HASMCs. TUG1 sponged miR-216a-3p to positively regulate SMURF2 expression. CONCLUSION TUG1 downregulation inhibited PDGF-BB-induced HASMC proliferation and migration by regulating miR-216a-3p/SMURF2 axis, offering novel insight into the potential application of TUG1 for childhood asthma treatment.
Collapse
Affiliation(s)
- Xinfang Wang
- Department of Pediatrics, Hangzhou First People's Hospital Affiliated to Zhejiang University, Zhejiang, Hangzhou, China
| | - Junsong Chen
- Respiratory Department, Hangzhou Children's Hospital, 195 Wenhui Road, Zhejiang, 310003, Hangzhou, China.
| |
Collapse
|
36
|
Dai B, Sun F, Cai X, Li C, Liu F, Shang Y. Long noncoding RNA PTTG3P/miR-192-3p/CCNB1 axis is a potential biomarker of childhood asthma. Int Immunopharmacol 2021; 101:108229. [PMID: 34717195 DOI: 10.1016/j.intimp.2021.108229] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 08/25/2021] [Accepted: 10/03/2021] [Indexed: 12/22/2022]
Abstract
BACKGROUND Increasing evidence suggests that long non-coding RNAs (lncRNAs) affect the regulation of immune responses, airway inflammation, and other pathological processes involved in asthma. LncRNA PTTG3P is associated with the development of various tumors, but its role in childhood asthma remains unknown. In this study, we investigated the functions of the lncRNA PTTG3P in the progression of childhood asthma. METHODS Twenty-six healthy children and 26 asthmatic children were monitored for disease progression for 2 years. We obtained blood samples during the chronic phase of disease for lncRNA/mRNA expression microarray analysis. A competitive endogenous RNA network (PTTG3P/miR-192-3p/CCNB1) was identified using bioinformatics analyses. Real-time qPCR and western blot were used to quantify gene and protein expression levels, respectively. Cell counting kit‑8 and transwell assays were used to evaluate the proliferation and migration of bronchial epithelial (16HBE) cells. Double luciferase reporter gene assay was used to validate the predictive targets in PTTG3P, miR-192-3p, and CCNB1. RESULTS PTTG3P was highly expressed in the peripheral blood of asthmatic children. Knocking down PTTG3P inhibited epithelial-mesenchymal transition, proliferation, and migration of 16HBE cells. PTTG3P promoted progression of childhood asthma by targeting the miR-192-3p/CCNB1 axis. CONCLUSIONS Childhood asthma was associated with the PTTG3P/miR-192-3p/CCNB1 axis. This study provides potential diagnostic and treatment biomarkers for childhood asthma.
Collapse
Affiliation(s)
- Bing Dai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Feifei Sun
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuxu Cai
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Chunlu Li
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Fen Liu
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunxiao Shang
- Department of Pediatrics, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
37
|
Zhou H, Long C, Liu P, Chen Y, Luo L, Xiao Z. Long non-coding RNA TUG1 accelerates abnormal growth of airway smooth muscle cells in asthma by targeting the miR-138-5p/E2F3 axis. Exp Ther Med 2021; 22:1229. [PMID: 34539825 DOI: 10.3892/etm.2021.10663] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 06/28/2021] [Indexed: 12/14/2022] Open
Abstract
Asthma is a chronic airway inflammatory disease. The present study aimed to explore the effect of the long non-coding RNA taurine-upregulated gene 1 (TUG1) on the viability and migration of airway smooth muscle cells (ASMCs) in asthma. Rat asthma models were constructed with ovalbumin sensitization and challenge and the level of serum immunoglobulin E (IgE) and the rates of inspiratory and expiratory resistance were measured. Reverse transcription-quantitative PCR was also performed to determine the expression levels of TUG1. Platelet-derived growth factor-BB (PDGF-BB)-treated ASMCs were then used as a cell model of asthma. The viability and migratory abilities of ASMCs were analysed with the MTT and Transwell assays. Additionally, a dual-luciferase reporter assay was used to confirm the relationship between TUG1 and microRNA (miR)-138-5p and between transcription factor E2F3 and miR-138-5p. The expression of TUG1, level of serum IgE, inspiratory resistance and expiratory resistance were clearly increased in the rat asthma model in comparison with controls. Knockdown of TUG1 the viability and migration of PDGF-BB-induced ASMCs and reduced the inspiratory and expiratory resistances. In addition, TUG1 functioned as a bait of miR-138-5p, and miR-138-5p modulated E2F3 expression. Knockdown of E2F3 hindered the abnormal growth of ASMCs. Moreover, miR-138-5p inhibition or E2F3 overexpression reversed the inhibitory effects of TUG1 knockdown on viability and migration of PDGF-BB-induced ASMCs. The TUG1/miR-138-5p/E2F3 regulatory axis appeared to play a critical role in accelerating the viability and migration of ASMCs and may therefore have a role in asthma.
Collapse
Affiliation(s)
- Haiyin Zhou
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Caixia Long
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Pingping Liu
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Yanying Chen
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Lan Luo
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| | - Zhenghui Xiao
- Intensive Care Unit, Hunan Children's Hospital, Changsha, Hunan 410007, P.R. China
| |
Collapse
|
38
|
Nociti V, Santoro M. What do we know about the role of lncRNAs in multiple sclerosis? Neural Regen Res 2021; 16:1715-1722. [PMID: 33510060 PMCID: PMC8328773 DOI: 10.4103/1673-5374.306061] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 08/21/2020] [Accepted: 11/11/2020] [Indexed: 12/24/2022] Open
Abstract
Multiple sclerosis is a chronic, inflammatory and degenerative disease of the central nervous system of unknown aetiology although well-defined evidence supports an autoimmune pathogenesis. So far, the exact mechanisms leading to autoimmune diseases are still only partially understood. We know that genetic, epigenetic, molecular, and cellular factors resulting in pathogenic inflammatory responses are certainly involved. Long non-coding RNAs (lncRNAs) are non-protein coding transcripts longer than 200 nucleotides that play an important role in both innate and acquired immunity, so there is great interest in lncRNAs involved in autoimmune diseases. The research on multiple sclerosis has been enriched with many studies on the molecular role of lncRNAs in the pathogenesis of the disease and their potential application as diagnostic and prognostic biomarkers. In particular, many multiple sclerosis fields of research are based on the identification of lncRNAs as possible biomarkers able to predict the onset of the disease, its activity degree, its progression phase and the response to disease-modifying drugs. Last but not least, studies on lncRNAs can provide a new molecular target for new therapies, missing, so far, a cure for multiple sclerosis. While our knowledge on the role of lncRNA in multiple sclerosis has recently improved, further studies are required to better understand the specific role of lncRNAs in this neurological disease. In this review, we present the most recent studies on molecular characterization of lncRNAs in multiple sclerosis disorder discussing their clinical relevance as biomarkers for diagnosis and treatments.
Collapse
Affiliation(s)
- Viviana Nociti
- Institute of Neurology, Fondazione Policlinico Universitario 'A. Gemelli' IRCCS, Università Cattolica del Sacro Cuore, Rome, Italy
| | | |
Collapse
|
39
|
Chen X, Yang J, Shen H, Zhang X, Wang H, Wu G, Qi Y, Wang L, Xu W. Muc5ac Production Inhibited by Decreased lncRNA H19 via PI3K/Akt/NF-kB in Asthma. J Asthma Allergy 2021; 14:1033-1043. [PMID: 34421304 PMCID: PMC8373259 DOI: 10.2147/jaa.s316250] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 07/27/2021] [Indexed: 12/27/2022] Open
Abstract
Introduction LncRNAs play important roles in multiple diseases including asthma, while there are a few reports on the role of lncRNA H19 about asthma. This study aimed to investigate the roles and mechanisms of lncRNA H19 in asthma. Methods We detected lncRNA H19 and Muc5ac mRNA by establishing a murine asthma model and an in vitro inflammation model. Regulatory roles of lncRNA H19 in asthma were explored by lncRNA H19 overexpression or knockdown in vitro. To study its mechanisms, we detect p-NF-κB and p-Akt expression, and treated 16-HBE cells with inhibitors of PI3K. To study regulatory effects of miR-675-3p on Muc5ac, miR-675-3p mimics and inhibitors were respectively transfected into 16-HBE cells. Results Firstly, we established a murine asthma model and an in vitro inflammation model. We found that lncRNA H19 expression was decreased, while Muc5ac mRNA was increased in lung tissues of murine asthma model and in the in vitro inflammation model. lncRNA H19 overexpression increased Muc5ac mRNA expression and lncRNA H19 knockdown decreased Muc5ac mRNA expression in 16-HBE cells. Moreover, lncRNA H19 overexpression further increased Muc5ac expression in TNFα-induced in vitro inflammation model. lncRNA H19 knockdown decreased p-Akt and p-NF-κB expression. Inhibitors of PI3K abolished Muc5ac induced by lncRNA H19 overexpression. Although miR-675-3p was increased by lncRNA H19 overexpression, it had no regulatory effects on Muc5ac expression. Discussion These results demonstrated that lncRNA H19 positively regulates Muc5ac expression through PI3K/Akt /NF-κB pathway in the in vitro inflammation model. Therefore, this study indicated that decreased lncRNA H19 in asthma might play a protective role relieving mucus overproduction, and lncRNA H19 might be a potential target for asthma treatment.
Collapse
Affiliation(s)
- Xu Chen
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China.,Department of Clinical Laboratory, First People's Hospital of Liangshan Yi Autonomous Prefecture, Xichang, Sichuan, People's Republic of China
| | - Jing Yang
- Department of Clinical Laboratory, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 611731, People's Republic of China
| | - Hailan Shen
- Department of Laboratory Medicine, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Xuemei Zhang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Hong Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Guangying Wu
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Yuhong Qi
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Ling Wang
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| | - Wenchun Xu
- Key Laboratory of Laboratory Medical Diagnostics Designated by the Ministry of Education, School of Laboratory Medicine, Chongqing Medical University, Chongqing, People's Republic of China
| |
Collapse
|
40
|
Hao M, Zan J. The Identification of Childhood Asthma Progression-Related lncRNAs and mRNAs Suitable as Biomarkers Using Weighted Gene Coexpression Network Analysis. Genet Res (Camb) 2021; 2021:5511507. [PMID: 34456632 PMCID: PMC8371738 DOI: 10.1155/2021/5511507] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 07/16/2021] [Indexed: 12/14/2022] Open
Abstract
Background Asthma is a common chronic respiratory disease in children, seriously affecting children's health and growth. This bioinformatics study aimed to identify potential RNA candidates closely associated with childhood asthma development within current gene databases. Methods GSE65204 and GSE19187 datasets were screened and downloaded from the NCBI GEO database. Differentially expressed long noncoding RNAs (DE-lncRNAs) and mRNAs (DE-mRNAs) were identified using the Bioconductor limma package in R, and these DE-mRNAs were used to perform biological process (BP) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analyses. Thereafter, weighted gene coexpression network analysis (WGCNA) was utilized to screen the modules directly related to childhood asthma, and a coexpression network of DE-lncRNAs and DE-mRNAs was built. Finally, principal component analysis (PCA) was performed. Results In total, 7 DE-lncRNAs and 1060 DE-mRNAs, as well as 7 DE-lncRNAs and 1027 DE-mRNAs, were identified in GSE65204 and GSE19187, respectively. After comparison, 336 overlapping genes had the same trend of expression, including 2 overlapped DE-lncRNAs and 334 overlapped DE-mRNAs. These overlapped DE-mRNAs were enriched in 28 BP and 12 KEGG pathways. Eleven modules were obtained in GSE65204, and it was found that the purple, black, and yellow modules were significantly positively correlated with asthma development. Subsequently, a coexpression network including 63 DE-mRNAs and 2 DE-lncRNAs was built, and five KEGG pathways, containing 8 genes, were found to be directly associated with childhood asthma. The PCA further verified these results. Conclusion LncRNAs LINC01559 and SNHG8 and mRNAs VWF, LAMB3, LAMA4, CAV1, ALDH1A3, SMOX, GNG4, and PPARG were identified as biomarkers associated with the progression of childhood asthma.
Collapse
Affiliation(s)
- Min Hao
- Department of Pediatrics, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| | - Jinling Zan
- Department of Intensive Care Unit, Zaozhuang Municipal Hospital, Zaozhuang, Shandong 277100, China
| |
Collapse
|
41
|
Devadoss D, Daly G, Manevski M, Houserova D, Hussain SS, Baumlin N, Salathe M, Borchert GM, Langley RJ, Chand HS. A long noncoding RNA antisense to ICAM-1 is involved in allergic asthma associated hyperreactive response of airway epithelial cells. Mucosal Immunol 2021; 14:630-639. [PMID: 33122732 PMCID: PMC8081750 DOI: 10.1038/s41385-020-00352-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 10/11/2020] [Accepted: 10/14/2020] [Indexed: 02/04/2023]
Abstract
Epithelial cells of the conducting airways are a pivotal first line of defense against airborne pathogens and allergens that orchestrate inflammatory responses and mucociliary clearance. Nonetheless, the molecular mechanisms responsible for epithelial hyperreactivity associated with allergic asthma are not completely understood. Transcriptomic analysis of human airway epithelial cells (HAECs), differentiated in-vitro at air-liquid interface (ALI), showed 725 differentially expressed immediate-early transcripts, including putative long noncoding RNAs (lncRNAs). A novel lncRNA on the antisense strand of ICAM-1 or LASI was identified, which was induced in LPS-primed HAECs along with mucin MUC5AC and its transcriptional regulator SPDEF. LPS-primed expression of LASI, MUC5AC, and SPDEF transcripts were higher in ex-vivo cultured asthmatic HAECs that were further augmented by LPS treatment. Airway sections from asthmatics with increased mucus load showed higher LASI expression in MUC5AC+ goblet cells following multi-fluorescent in-situ hybridization and immunostaining. LPS- or IL-13-induced LASI transcripts were mostly enriched in the nuclear/perinuclear region and were associated with increased ICAM-1, IL-6, and CXCL-8 expression. Blocking LASI expression reduced the LPS or IL-13-induced epithelial inflammatory factors and MUC5AC expression, suggesting that the novel lncRNA LASI could play a key role in LPS-primed trained airway epithelial responses that are dysregulated in allergic asthma.
Collapse
Affiliation(s)
- Dinesh Devadoss
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Grant Daly
- Department of Pharmacology, University of South Alabama, Mobile, AL, 36688, USA
| | - Marko Manevski
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA
| | - Dominika Houserova
- Department of Pharmacology, University of South Alabama, Mobile, AL, 36688, USA
| | - Shah S Hussain
- Medicine-Pulmonary/Allergy/Critical Care, University of Alabama at Birmingham, Birmingham, AL, 35233, USA
| | - Nathalie Baumlin
- Division of Pulmonary, Critical Care, and Sleep Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Matthias Salathe
- Division of Pulmonary, Critical Care, and Sleep Medicine, Miller School of Medicine, University of Miami, Miami, FL, 33136, USA
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS, 66160, USA
| | - Glen M Borchert
- Department of Pharmacology, University of South Alabama, Mobile, AL, 36688, USA
| | - Raymond J Langley
- Department of Pharmacology, University of South Alabama, Mobile, AL, 36688, USA
| | - Hitendra S Chand
- Department of Immunology and Nano-Medicine, Herbert Wertheim College of Medicine, Florida International University, Miami, FL, 33199, USA.
| |
Collapse
|
42
|
Wang WL, Luo XM, Zhang Q, Zhu HQ, Chen GQ, Zhou Q. The lncRNA PVT1/miR-590-5p/FSTL1 axis modulates the proliferation and migration of airway smooth muscle cells in asthma. Autoimmunity 2021; 54:138-147. [PMID: 33825599 DOI: 10.1080/08916934.2021.1897977] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE Asthma is a prevalent chronic inflammatory airway disease that is characterised by airway remodelling and airway hyperresponsiveness. Abnormal proliferation and migration of airway smooth muscle cells (ASMCs) contribute to airway remodelling in asthma. However, the molecular mechanism underlying an increased ASMC mass in asthma remains elusive. Herein, we aimed at investigating the regulation of lncRNA PVT1 on ASMCs and focussing on the mechanism in the proliferation and migration. METHODS Expression levels of lncRNA PVT1 and miR-590-5p in the serum collected from 24 children with asthma and 10 control children were determined by qRT-PCR. ASMCs proliferation and migration prior to and post platelet-derived growth factor subunit B (PDGF-BB) stimulation were examined by CCK-8 test and transwell assay. Dual-luciferase reporter assay was performed to determine miR-590-5p interaction with lncRNA PVT1 and follistatin-like 1 (FSTL1). Expression of lncRNA PVT1, miR-590-5p, FSTL1, C-Myc, cyclin D1, and cyclin-dependent kinase 1 (CDK1) was tested by quantitative real-time PCR (qRT-PCR) and immunoblotting analysis. RESULTS The expression level of lncRNA PVT1 was higher but the expression level of miR-590-5p was lower in the serum of children with asthma than in control children. The expression level of lncRNA PVT1 was negatively correlated with the expression level of miR-590-5p in asthma. LncRNA PVT1 was upregulated upon PDGF-BB stimulation. LncRNA PVT1 knockdown by its specific shRNA repressed PDGF-BB-induced promotion of proliferation and migration in ASMCs and triggered an elevated miR-590-5p along with declined C-Myc, cyclin D1, and CDK1. The effects of lncRNA PVT1 knockdown on PDGF-BB-induced ASMCs were lost upon miR-590-5p inhibition. MiR-590-5p targeted FSTL1 gene and declined its expression, thus suppressing ASMC proliferation and migration following PDGF-BB stimulation and downregulating C-Myc, cyclin D1, and CDK1 expressions. The effects of miR-590-5p on PDGF-BB-induced ASMCs were lost upon FSTL1 overexpression. CONCLUSION These results support the notion that the lncRNA PVT1/miR-590-5p/FSTL1 axis modulates ASMCs proliferation and migration following PDGF-BB stimulation, providing a potential therapeutic target to attenuate airway remodelling in asthma.
Collapse
Affiliation(s)
- Wen-Lan Wang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Xiao-Ming Luo
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhang
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Hai-Qiao Zhu
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Guo-Qing Chen
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| | - Qin Zhou
- Department of Pediatrics, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, P.R. China
| |
Collapse
|
43
|
Liu H, Yin Y, Liu T, Gao Y, Ye Q, Yan J, Ha F. Long non-coding RNA PVT1 regulates the migration of hepatocellular carcinoma HepG2 cells via miR-3619-5p/MKL1 axis. Bosn J Basic Med Sci 2021; 21:187-197. [PMID: 32156248 PMCID: PMC7982070 DOI: 10.17305/bjbms.2020.4641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 02/22/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the third most common malignant tumor of the digestive system. Plasma cell tumor heterotopic gene 1 (PVT1) is an intergenic long non-coding RNA that is aberrantly expressed in different cancers. Myocardin-related transcription factor A or megakaryoblastic leukemia 1 (MKL1) is a transcriptional coactivator of serum response factor that has been shown to promote cancer cell migration and invasion. In this study, we investigated the relationship between PVT1 and MKL1 as a novel regulatory mechanism underlying HCC progression. We used HepG2 and Cos-7 cell lines. Transfection experiments with miR-3619-5p mimics/inhibitor, PVT1, siRNA-PVT1, MKL1, or siRNA-MKL1 were performed. RNA and protein levels were analyzed by quantitative reverse transcription PCR and Western blot, respectively. Cell migration was assessed by transwell assay. Luciferase assays, RNA-FISH, RNA immunoprecipitation, and chromatin immunoprecipitation assays were performed to confirm the interaction between PVT1, miR-3619-5p, and MKL1 in HCC cells. Overexpression of PVT1 was positively correlated with MKL1 upregulation, which promoted HepG2 cell migration. miR-3619-5p inhibited MKL1 expression in HCC cells by acting on its 3′-UTR. Furthermore, PVT1 promoted MKL1 expression and migration in HCC cells by directly binding to miR-3619-5p. In a positive feedback loop, MKL1 could activate PVT1 transcription by binding to the CArG box in the promoter region. Our findings may provide a basis for the development of novel targeted therapies in HCC.
Collapse
Affiliation(s)
- Hua Liu
- The Third Central Hospital of Tianjin, Tianjin, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Yan Yin
- Respiratory and Critical Care Medicine of Tianjin Chest Hospital, Tianjin, China
| | - Ting Liu
- Tianjin Institute of Cardiovascular Disease, Tianjin Chest Hospital, Tianjin, China
| | - Yanying Gao
- The Third Central Hospital of Tianjin, Tianjin, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Qing Ye
- The Third Central Hospital of Tianjin, Tianjin, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Junqing Yan
- The Third Central Hospital of Tianjin, Tianjin, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| | - Fushuang Ha
- The Third Central Hospital of Tianjin, Tianjin, China; Tianjin Key Laboratory of Extracorporeal Life Support for Critical Diseases, Tianjin, China; Artificial Cell Engineering Technology Research Center, Tianjin, China; Tianjin Institute of Hepatobiliary Disease, Tianjin, China
| |
Collapse
|
44
|
Yang M, Wang L. MALAT1 knockdown protects from bronchial/tracheal smooth muscle cell injury via regulation of microRNA-133a/ryanodine receptor 2 axis. J Biosci 2021. [DOI: 10.1007/s12038-021-00149-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
45
|
Liu Y, Peng H, Gui F. Long noncoding plasmacytoma variant translocation 1 facilitates the surveillance of acute respiratory distress syndrome and mortality prediction in sepsis. Biomark Med 2021; 15:401-412. [PMID: 33733809 DOI: 10.2217/bmm-2020-0506] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Aim: We aimed to investigate the association of long noncoding RNA plasmacytoma variant translocation 1 (lncRNA PVT1) expression with acute respiratory distress syndrome (ARDS) risk and its prognostic value for 28-day mortality in sepsis patients. Materials & methods: LncRNA PVT1 expression from 109 sepsis patients and 100 health controls was detected. General sepsis severity was assessed using acute physiology and chronic health evaluation II score and sequential organ failure assessment score. Results: LncRNA PVT1 had an acceptable predictive value for higher ARDS risk, then was identified as an independent risk factor for sepsis ARDS; LncRNA PVT1 expression positively correlated with general disease severity in sepsis patients; LncRNA PVT1 was overexpressed in 28-day deaths compared with 28-day survivors in sepsis patients. Conclusion: LncRNA PVT1 may facilitate the surveillance of ARDS, general disease severity and the prediction of mortality in sepsis patients.
Collapse
Affiliation(s)
- Yijue Liu
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Huan Peng
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| | - Feng Gui
- Department of Emergency, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, China
| |
Collapse
|
46
|
Zhu MX, Huang LH, Zhu YK, Cai XJ. LncRNA NEAT1 promotes airway smooth muscle cell inflammation by activating the JAK3/STAT5 pathway through targeting of miR-139. Exp Lung Res 2021; 47:161-172. [PMID: 33590796 DOI: 10.1080/01902148.2021.1876792] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Background Asthma is a chronic inflammatory heterogeneous respiratory disease. Previous studies showed that the lncRNA NEAT1 (nuclear paraspeckle assembly transcript 1) might play an important role in the pathogenesis of asthma, but its potential mechanism in airway smooth muscle cell (ASMC) inflammation remains largely unknown and needs further investigation.Methods We performed cellular immunofluorescence to identify the features of ASMCs and detected the expression levels of lncRNA NEAT1, miR-139, TNF-α, IL-6, IL-8 and IL-1β by quantitative real-time PCR (Q-PCR) and ELISA. Western blotting (WB) was used to measure the protein expression of the related genes, and bioinformatics as well as dual luciferase assays were used to validate the interaction between lncRNA NEAT1 and miR-139 and the interaction between miR-139 and the 3'-UTR of JAK3.Results The expression of lncRNA NEAT1 was increased in the ASMCs of asthma patients, but miR-139 was decreased. Overexpression of lncRNA NEAT1 promoted the expression of the inflammatory cytokines such as TNF-α, IL-6, IL-8 and IL-1β in ASMCs. LncRNA NEAT1 was able to target miR-139 to activate the JAK3/STAT5 signaling pathway and induced the expression of these inflammatory cytokines in ASMCs. Overexpression of miR-139 or suppression of the JAK3/STAT5 signaling pathway reversed the inflammatory effect of lncRNA NEAT1.Conclusion LncRNA NEAT1 played a pivotal role in ASMC inflammation and exerted its function through the miR-139/JAK3/STAT5 signaling network.
Collapse
Affiliation(s)
- Meng-Xia Zhu
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan Province, China
| | - Lin-Hui Huang
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan Province, China
| | - Yi-Ke Zhu
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan Province, China
| | - Xing-Jun Cai
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan Province, China
| |
Collapse
|
47
|
Yu H, Qi N, Zhou Q. LncRNA H19 Inhibits Proliferation and Migration of Airway Smooth Muscle Cells Induced by PDGF-BB Through miR-21/PTEN/Akt Axis. J Asthma Allergy 2021; 14:71-80. [PMID: 33536765 PMCID: PMC7850386 DOI: 10.2147/jaa.s291333] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Accepted: 01/18/2021] [Indexed: 12/21/2022] Open
Abstract
Background LncRNA H19 expression is down-regulated in patients with asthma. The hyperplasia of airway smooth muscle cells (ASMCs) promotes the development of airway remodeling in asthma. Therefore, we attempted to evaluate the regulatory function of H19 in the proliferation and migration of ASMCs. Methods The expressions of H19 and miR-21 were detected using qRT-PCR. PDGF-BB-induced abnormal proliferation and migration of ASMCs was used as the airway remodeling model in vitro. The expressions of H19 and miR-21 were modified by transfection with pcDNA3.1-H19 and miR-21 mimic, respectively. CCK-8 assay, flow cytometry-based cell cycle analysis was conducted to examine the proliferation ability of ASMCs. The migration ability was measured by transwell assay. Dual-luciferase reporter system was carried out to find the potential relationship between miR-21 and H19 or PTEN. Western blot was conducted to detect the expressions of PCNA, MMP-9, α-SMA, PTEN, and the phosphorylation level of Akt. Results LncRNA-H19 expression was decreased and microRNA-21 expression was increased in serum samples of children with asthma and PDGF-BB-stimulated ASMCs. Overexpression of H19 reduced the proliferation and migration ability of ASMCs with PDGF-BB treatment and these changes were reversed by miR-21 mimic. H19 promoted the protein level of PTEN via sponging miR-21. Overexpression of H19 suppressed miR-21-induced phosphorylation of Akt, and the suppression effect of H19 on phosphorylation of Akt was significantly reduced after transfecting shPTEN in ASMCs. Conclusion In this study, overexpression of H19 suppressed the proliferation and migration of ASMCs induced by PDGF-BB via miR-21/PTEN/Akt axis, which could be a potential biomarker and target for treating hyperplasia of airway smooth muscle cells.
Collapse
Affiliation(s)
- Haiying Yu
- Department of Pediatrics, Weifang People's Hospital, Weifang, Shandong 261041, People's Republic of China
| | - Ningning Qi
- Department of Pediatrics, Weifang People's Hospital, Weifang, Shandong 261041, People's Republic of China
| | - Qingxia Zhou
- Department of Pediatrics, Weifang People's Hospital, Weifang, Shandong 261041, People's Republic of China
| |
Collapse
|
48
|
Cai XJ, Huang LH, Zhu YK, Huang YJ. LncRNA OIP5‑AS1 aggravates house dust mite‑induced inflammatory responses in human bronchial epithelial cells via the miR‑143‑3p/HMGB1 axis. Mol Med Rep 2020; 22:4509-4518. [PMID: 33174035 PMCID: PMC7646745 DOI: 10.3892/mmr.2020.11536] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Accepted: 06/23/2020] [Indexed: 12/21/2022] Open
Abstract
Bronchial asthma poses a serious threat to human health. Previous studies have documented the role of long non‑coding RNAs (lncRNAs) in asthma. However, the molecular mechanism underlying bronchial asthma remains unclear. The aim of the present study was to evaluate the role of the lncRNA Opa‑interacting protein 5 antisense RNA1 (OIP5‑AS1) in the house dust mite‑induced inflammatory response in human bronchial epithelial cells. BEAS‑2B cells were treated with Dermatophagoides pteronyssinus peptidase 1 (Der p1) to establish an in vitro model of asthma. OIP5‑AS1 expression levels increased in BEAS‑2B cells following Der p1 treatment, while microRNA (miR)‑143‑3p was downregulated. Additionally, the levels of the pro‑inflammatory factors tumor necrosis factor‑α, interleukin (IL)‑6 and IL‑8 were measured, and apoptosis was evaluated following OIP5 silencing. OIP5‑AS1 knockdown reduced the inflammatory response and apoptosis in BEAS‑2B cells. Furthermore, using dual luciferase reporter assays and co‑transfection experiments, it was demonstrated that the function of OIP5‑AS1 was mediated by miR‑143‑3p. miR‑143‑3p overexpression attenuated the Der p1‑induced inflammatory response and apoptosis of BEAS‑2B cells by targeting high mobility group box 1 (HMGB1). In summary, OIP5‑AS1 exacerbated Der p1‑induced inflammation and apoptosis in BEAS‑2B cells by targeting miR‑143‑3p via HMGB1.
Collapse
Affiliation(s)
- Xing-Jun Cai
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Lin-Hui Huang
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Yi-Ke Zhu
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| | - Yi-Jiang Huang
- Department of Respiratory and Critical Care Medicine, Hainan General Hospital, Haikou, Hainan 570311, P.R. China
| |
Collapse
|
49
|
Wei Y, Han B, Dai W, Guo S, Zhang C, Zhao L, Gao Y, Jiang Y, Kong X. Exposure to ozone impacted Th1/Th2 imbalance of CD 4+ T cells and apoptosis of ASMCs underlying asthmatic progression by activating lncRNA PVT1-miR-15a-5p/miR-29c-3p signaling. Aging (Albany NY) 2020; 12:25229-25255. [PMID: 33223504 PMCID: PMC7803560 DOI: 10.18632/aging.104124] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 09/14/2020] [Indexed: 02/07/2023]
Abstract
This investigation attempted to elucidate whether lncRNA PVT1-led miRNA axes participated in aggravating ozone-triggered asthma progression. One hundred and sixty-eight BALB/c mice were evenly divided into saline+air group, ovalbumin+air group, saline+ozone group and ovalbumin+ozone group. Correlations were evaluated between PVT1 expression and airway smooth muscle function/inflammatory cytokine release among the mice models. Furthermore, pcDNA3.1-PVT1 and si-PVT1 were, respectively, transfected into CD4+T cells and airway smooth muscle cells (ASMCs), and activities of the cells were observed. Ultimately, a cohort of asthma patients was recruited to estimate the diagnostic performance of PVT1. It was demonstrated that mice of ovalbumin+ozone group were associated with higher PVT1 expression, thicker trachea/airway smooth muscle and smaller ratio of Th1/Th2-like cytokines than mice of ovalbumin+air group and saline+ozone group (P<0.05). Moreover, pcDNA3.1-PVT1 significantly brought down Th1/Th2 ratio in CD4+ T cells by depressing miR-15a-5p expression and activating PI3K-Akt-mTOR signaling (P<0.05). The PVT1 also facilitated ASMC proliferation by sponging miR-29c-3p and motivating PI3K-Akt-mTOR signaling (P<0.05). Additionally, PVT1 seemed promising in diagnosis of asthma, with favorable sensitivity (i.e. 0.844) and specificity (i.e. 0.978). Conclusively, lncRNA PVT1-miR-15a-5p/miR-29c-3p-PI3K-Akt-mTOR axis was implicated in ozone-induced asthma development by promoting ASMC proliferation and Th1/Th2 imbalance.
Collapse
Affiliation(s)
- Yangyang Wei
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Baofen Han
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Wenjuan Dai
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Shufang Guo
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Caiping Zhang
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Lixuan Zhao
- Department of Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yan Gao
- Department of Medicine, Shanxi Medical University, Taiyuan 030001, China
| | - Yi Jiang
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| | - Xiaomei Kong
- Department of Respiratory and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan 030001, China
| |
Collapse
|
50
|
Ogunwobi OO, Segura MF. Editorial: PVT1 in Cancer. Front Oncol 2020; 10:588786. [PMID: 33194746 PMCID: PMC7606904 DOI: 10.3389/fonc.2020.588786] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 09/08/2020] [Indexed: 12/03/2022] Open
Affiliation(s)
- Olorunseun O Ogunwobi
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY, United States
| | - Miguel F Segura
- Laboratory of Translational Research in Child and Adolescent Cancer, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, VHIR, Universitat Autònoma de Barcelona, Barcelona, Spain
| |
Collapse
|