1
|
Xu W, Yang J, Yu H, Li S. Diagnostic value of lncRNAs LINC00152 and LARS2-AS1 and their regulatory roles in macrophage immune response in tuberculosis. Tuberculosis (Edinb) 2024; 148:102530. [PMID: 38857553 DOI: 10.1016/j.tube.2024.102530] [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: 12/28/2023] [Revised: 06/04/2024] [Accepted: 06/05/2024] [Indexed: 06/12/2024]
Abstract
OBJECTIVES To determine the usefulness of LINC00152 and LARS2-AS1 as potential biomarkers for latent tuberculosis (LTB) and active tuberculosis (ATB), as well as their effect on Mycobacterium (Mtb) infection. METHODS The expression levels of LINC00152 and LARS2-AS1 in the health, patients with LTB and ATB were detected by qRT-PCR. The ROC curves were constructed to show their potential as biomarkers. The intracellular survival assays for Mtb and the levels of immune-related cytokines were determined to discover the effect of LINC00152 and LARS2-AS1 on Mtb infection. The relationships of miR-485-5p with LINC00152 and LARS2-AS1 were explored. RESULTS LINC00152 and LARS2-AS1 levels were significantly elevated in patients with ATB and LTB, and Mtb-infected macrophages. LINC00152 and LARS2-AS1 can distinguish the LTB from the health and ATB from LTB. LARS2-AS1 and LINC00152 knock-down reduced the intracellular Mtb survival and induced cellular immune response after Mtb challenge. miR-485-5p was a targeting miRNA for LINC00152 and LARS2-AS1. CONCLUSIONS LINC00152 and LARS2-AS1 can be considered as potential biomarkers for tuberculosis disease. LINC00152 and LARS2-AS1 have anti-Mtb effects.
Collapse
Affiliation(s)
- Wenlong Xu
- Department of Clinical Laboratory, Shanghai Yangsi Hospital, Shanghai, 200126, China
| | - Jihua Yang
- Department of Ultrasound, Central Hospital Affiliated to Shandong First Medical University, Jinan, 250013, China
| | - Haizhen Yu
- Department of Clinical Laboratory, Zhucheng People's Hospital, Zhucheng, 262299, China
| | - Shizhen Li
- Department of Clinical Laboratory, Zhucheng People's Hospital, Zhucheng, 262299, China.
| |
Collapse
|
2
|
Hu R, Molibeli KM, Zhu L, Li H, Chen C, Wang Y, Xiong D, Liu J, Tang L. Long non-coding RNA-XLOC_002383 enhances the inhibitory effects of THP-1 macrophages on Mycobacterium avium and functions as a competing endogenous RNA by sponging miR-146a-5p to target TRAF6. Microbes Infect 2023; 25:105175. [PMID: 37392988 DOI: 10.1016/j.micinf.2023.105175] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 05/30/2023] [Accepted: 06/22/2023] [Indexed: 07/03/2023]
Abstract
The morbidity associated with infection by Mycobacterium avium (M. avium), a type of non-tuberculous mycobacteria (NTM), has increased in recent years due to infections that are easily missed, and thus, difficult to diagnose and treat. Here, we reported that miR-146a-5p was highly expressed, and XLOC_002383 and TRAF6 were downregulated in a time- and MOI-dependent manner in THP-1 macrophages infected with M. avium. In macrophages obtained from peripheral blood mononuclear cells, the expression levels of XLOC_002383 and TRAF6 were also decreased, and miR-146a-5p expression was increased following 24 h of infection with M. avium. miR-146a-5p was a target of XLOC_002383 and TRAF6 mRNA was a target of miR-146a-5p, and XLOC_002383 regulated TRAF6 expression by adsorbing miR-146a-5p, and further increased IL-6, TNF-α, IL-1β and iNOS levels in THP-1 macrophages. The results of qPCR and CFU assays indicated that XLOC_002383 decreased the intracellular M. avium loads. Overall, the present study demonstrated that XLOC_002383 may function as a competing endogenous RNA and interacts with miR-146a-5p to increase THP-1 macrophage inflammatory factors and microbicidal mediators iNOS. This enhanced the inhibitory effects of THP-1 macrophages on M. avium, which improved the understanding of the pathogenesis and host defenses in the process of NTM infectious diseases.
Collapse
Affiliation(s)
- Rong Hu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Kearabetsoe Matseliso Molibeli
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Lin Zhu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Hui Li
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Cai Chen
- Changsha KingMed Center for Clinical Laboratory, Changsha, Hunan, 410100, China.
| | - Yang Wang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Dehui Xiong
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Jing Liu
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| | - Lijun Tang
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China; Xiangya School of Medicine, Central South University, 172 Tongzipo Road, Yuelu, Changsha, Hunan, 410013, China.
| |
Collapse
|
3
|
Li Z, Gao J, Sun D, Jiao Q, Ma J, Cui W, Lou Y, Xu F, Li S, Li H. LncRNA MEG3: Potential stock for precision treatment of cardiovascular diseases. Front Pharmacol 2022; 13:1045501. [PMID: 36523500 PMCID: PMC9744949 DOI: 10.3389/fphar.2022.1045501] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/11/2022] [Indexed: 10/13/2023] Open
Abstract
The prevalence and mortality rates of cardiovascular diseases are increasing, and new treatment strategies are urgently needed. From the perspective of basic pathogenesis, the occurrence and development of cardiovascular diseases are related to inflammation, apoptosis, fibrosis and autophagy of cardiomyocytes, endothelial cells and other related cells. The involvement of maternally expressed gene 3 (MEG3) in human disease processes has been increasingly reported. P53 and PI3K/Akt are important pathways by which MEG3 participates in regulating cell apoptosis. MEG3 directly or competitively binds with miRNA to participate in apoptosis, inflammation, oxidative stress, endoplasmic reticulum stress, EMT and other processes. LncRNA MEG3 is mainly involved in malignant tumors, metabolic diseases, immune system diseases, cardiovascular and cerebrovascular diseases, etc., LncRNA MEG3 has a variety of pathological effects in cardiomyocytes, fibroblasts and endothelial cells and has great clinical application potential in the prevention and treatment of AS, MIRI, hypertension and HF. This paper will review the research progress of MEG3 in the aspects of mechanism of action, other systemic diseases and cardiovascular diseases, and point out its great potential in the prevention and treatment of cardiovascular diseases. lncRNAs also play a role in endothelial cells. In addition, lncRNA MEG3 has shown biomarker value, prognostic value and therapeutic response measurement in tumor diseases. We boldly speculate that MEG3 will play a role in the emerging discipline of tumor heart disease.
Collapse
Affiliation(s)
- Zining Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Jialiang Gao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Division, Beijing, China
- Deputy Chief Physician, Beijing, China
| | - Di Sun
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Qian Jiao
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Jing Ma
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Weilu Cui
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Yuqing Lou
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Fan Xu
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Shanshan Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Master’s Degree Student, Beijing, China
- Cardiovascular Division, Beijing, China
| | - Haixia Li
- Guang’anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
- Cardiovascular Division, Beijing, China
- Chief Physician, Beijing, China
| |
Collapse
|
4
|
Yao Q, Xie Y, Xu D, Qu Z, Wu J, Zhou Y, Wei Y, Xiong H, Zhang XL. Lnc-EST12, which is negatively regulated by mycobacterial EST12, suppresses antimycobacterial innate immunity through its interaction with FUBP3. Cell Mol Immunol 2022; 19:883-897. [PMID: 35637281 PMCID: PMC9149337 DOI: 10.1038/s41423-022-00878-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 05/02/2022] [Indexed: 02/07/2023] Open
Abstract
Long noncoding RNAs (lncRNAs) have been implicated in the pathogenesis of intracellular pathogens. However, the role and mechanism of the important lncRNAs in Mycobacterium tuberculosis (M.tb) infection remain largely unexplored. Recently, we found that a secreted M.tb Rv1579c (an early secreted target with a molecular weight of 12 kDa, named EST12) protein activates NLRP3-gasdermin D (GSDMD)-mediated pyroptosis and plays a pivotal role in M.tb-induced immunity. In the present study, M.tb and the EST12 protein negatively regulated the expression of a key lncRNA (named lnc-EST12) in mouse macrophages by activating the JAK2-STAT5a signaling pathway. Lnc-EST12, with a size of 1583 bp, is mainly expressed in immune-related organs (liver, lung and spleen). Lnc-EST12 not only reduces the expression of the proinflammatory cytokines IL-1β, IL-6, and CCL5/8 but also suppresses the NLRP3 inflammasome and GSDMD pyroptosis-IL-1β immune pathway through its interaction with the transcription factor far upstream element-binding protein 3 (FUBP3). The KH3 and KH4 domains of FUBP3 are the critical sites for binding to lnc-EST12. Deficiency of mouse lnc-EST12 or FUBP3 in macrophages increased M.tb clearance and inflammation in mouse macrophages or mice. In conclusion, we report a new immunoregulatory mechanism in which mouse lnc-EST12 negatively regulates anti-M.tb innate immunity through FUBP3.
Collapse
Affiliation(s)
- Qili Yao
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Yan Xie
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Dandan Xu
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Zilu Qu
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Jian Wu
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Yuanyuan Zhou
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Yuying Wei
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Huan Xiong
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China
| | - Xiao-Lian Zhang
- Hubei Province Key Laboratory of Allergy and Immunology, Department of Immunology, Wuhan University TaiKang Medical School (School of Basic Medical Sciences), Wuhan, China.
- State Key Laboratory of Virology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan, China.
- Department of Allergy, Zhongnan Hospital, Wuhan University, Wuhan, China.
- Wuhan Research Center for Infectious Diseases and Cancer, Chinese Academy of Medical Sciences, Wuhan, China.
| |
Collapse
|
5
|
Sun W, He X, Zhang X, Wang X, Lin W, Wang X, Liang Y. Diagnostic value of Long non-coding Ribonucleic Acid non-coding activated by Deoxyribonucleic Acid damage in pulmonary tuberculosis and its regulatory role in Mycobacterium tuberculosis infection of macrophages. Microbiol Immunol 2022; 66:433-441. [PMID: 35568971 DOI: 10.1111/1348-0421.12986] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Revised: 05/11/2022] [Accepted: 05/11/2022] [Indexed: 11/29/2022]
Abstract
Pulmonary tuberculosis (PTB) infection is a chronic inflammatory response caused by Mycobacterium tuberculosis (Mtb). The purpose of this study was to confirm the value of Long non-coding RNA (LncRNA) non-coding activated by DNA damage (NORAD) in the diagnosis of PTB and to explore its mechanism in Mtb-infected macrophages. NORAD serum levels were estimated by qRT-PCR in 90 PTB patients and 85 healthy individuals. ROC curves were employed to assess the diagnostic value of NORAD for PTB. Human and murine macrophages were infected with Mtb strain H37Rv. CCK-8 and ELISA detected macrophages viability and inflammatory cytokine secretion. A dual-luciferase reporter assay was performed to analyze the targeting relationship between NORAD and microRNA (miR)-618. NORAD was significantly elevated in patients with PTB, and its positivity was correlated with inflammatory cytokines IL-1 β (r = 0.854), TNF-α (r = 0.617), IL-6 (r = 0.585). With an AUC of 0.918, and sensitivity and specificity of 80.0% and 89.4%, respectively, NORAD remarkedly identified PTB patients from healthy individuals. Furthermore, Mtb infection significantly increased NORAD levels in THP-1 and RAW264.7 and increased their viability and inflammation (P <0.001). However, this increased effect was weakened by reduced NORAD. Dual-luciferase reporter assay confirmed that miR-618 in macrophages was a target miRNA for NORAD and can be negatively regulated by it. Moreover, elevated miR-618 suppressed macrophage viability and inflammation in Mtb infection. NORAD is a potential diagnostic biomarker for PTB and is involved in Mtb infected macrophage activity and inflammation by targeting miR-618. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Wenna Sun
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Xiong He
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Xiushuang Zhang
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Xiaomeng Wang
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Wen Lin
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Xiaofeng Wang
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| | - Yan Liang
- Senior Department of Tuberculosis, The 8th Medical Center of PLA General Hospital, Beijing, China
| |
Collapse
|
6
|
Expression and Clinical Significance of lncRNA NEAT1 in Patients with Spinal Tuberculosis. DISEASE MARKERS 2022; 2022:5748756. [PMID: 35465262 PMCID: PMC9023229 DOI: 10.1155/2022/5748756] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 03/22/2022] [Indexed: 12/22/2022]
Abstract
Background Spinal tuberculosis (STB) often leads to irreversible neurological injury, resulting in serious social and economic problems. With the emergence of drug resistance, the management becomes even more challenging, given the treatment courses are generally longer for skeletal than pulmonary tuberculosis (PTB). The development and validation of nonsputum biomarkers for diagnosis and tailoring of treatment duration to enable personalized and evidence-based management of such diseases to improve treatment outcomes is being called for globally. Studies have demonstrated that lncRNA NEAT1 was highly expressed in pulmonary tuberculosis (TB) and was related to its progression and recovery. However, the expression and clinical significance of lncRNA NEAT1 in STB remains unclear. Methods The relative expression of lncRNA NEAT1 was quantified by relative real-time reverse transcription PCR (RT-PCR). The prognostic value was assessed by receiver-operating characteristic (ROC) curve analysis. Pearson and Spearman correlation coefficient and chi-square test were used to analyze the correlation between the lncRNA NEAT1 expression and the clinical characteristics. Univariate and multivariate logistic regression analyses were used to analyze independent predictors of STB recurrence. Results Compared with normal healthy individuals, the expression level of lncRNA NEAT1 in peripheral blood and granulomatous tissues of STB patients was significantly increased. The results of the in vitro Mycobacterium tuberculosis- (Mtb-) infected cell model showed that the expression level of lncRNA NEAT1 was significantly upregulated in macrophages infected with Mtb, and the difference was statistically significant compared with Mtb-uninfected group. The expression level of lncRNA NEAT1 in granulomatous tissue of STB was significantly higher than that in peripheral blood. The expression of lncRNA NEAT1 was related to segments of the lesions, paraspinal abscesses, anti-TB treatment, drug resistance, interleukin-6 (IL-6), C-reactive protein (CRP), and erythrocyte sedimentation rate (ESR). Multivariate analysis results showed that relatively high expression of lncRNA NEAT1_1, the shorter transcript of the NEAT1 gene, was an independent prognostic factor of STB outcome. Conclusion LncRNA NEAT1 was highly expressed in peripheral blood mononuclear cells (PBMCs) and granulomatous tissue from patients with STB, as well as in Mtb-infected THP-1 cell lines. LncRNA NEAT1 expression was significantly associated with clinical characteristics (paraspinal abscesses, segments of the lesions and anti-TB treatment, IL-6, CRP, and ESR) of patients in STB. Increased expression of lncRNA NEAT1_1 predicted good prognosis of STB and might become a prognostic biomarker for STB.
Collapse
|
7
|
Angiotensin II-Treated Cardiac Myocytes Regulate M1 Macrophage Polarization via Transferring Exosomal PVT1. J Immunol Res 2021; 2021:1994328. [PMID: 34514000 PMCID: PMC8427676 DOI: 10.1155/2021/1994328] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 07/12/2021] [Accepted: 08/10/2021] [Indexed: 12/27/2022] Open
Abstract
Atrial fibrillation (AF) seriously reduces the health and life quality of patients. It is necessary to explore the pathogenesis of AF and provide a new target for the treatment. Here, exosomes were identified using transmission electron microscopy and nanoparticle tracing analysis. Western blotting assay was performed to detect the expression of exosomal surface markers, extracellular matrix-related proteins, and IL-16. The expression of genes was measured using qRT-PCR. Flow cytometry was performed to examine the percentages of CD86- and CD163-positive macrophages. Besides, luciferase activity assay was performed to explore the combination between PVT1 and miR-145-5p and the combination between miR-145-5p and IL-16 3'UTR. The combination between PVT1 and miR-145-5p also was examined using RIP assay. In our study, we isolated human cardiac myocyte- (HCM-) derived exosomes successfully. Ang-II-treated HCM-derived exosomes (Ang-II-Exo) promoted M1 macrophage polarization. PVT1 was highly expressed in Ang-II-Exo. Ang-II-Exo induced macrophage to M1 polarization through transferring PVT1. Furthermore, our data showed that PVT1 increased the expression of IL-16 via sponging miR-145-5p. Finally, we proved that exosomal PVT1 could boost the extracellular matrix remodeling of atrial fibroblasts. Overall, our data demonstrated that Ang-II-Exo promoted the extracellular matrix remodeling of atrial fibroblasts via inducing M1 macrophage polarization by transferring PVT1. PVT1 facilitated M1 polarization macrophage via increasing IL-16 expression by sponging miR-145-5p. Our results provided a new evidence for PVT1 which might be a treatment target of AF.
Collapse
|
8
|
Kundu M, Basu J. The Role of microRNAs and Long Non-Coding RNAs in the Regulation of the Immune Response to Mycobacterium tuberculosis Infection. Front Immunol 2021; 12:687962. [PMID: 34248974 PMCID: PMC8264550 DOI: 10.3389/fimmu.2021.687962] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 06/09/2021] [Indexed: 12/16/2022] Open
Abstract
Non-coding RNAs have emerged as critical regulators of the immune response to infection. MicroRNAs (miRNAs) are small non-coding RNAs which regulate host defense mechanisms against viruses, bacteria and fungi. They are involved in the delicate interplay between Mycobacterium tuberculosis, the causative agent of tuberculosis (TB), and its host, which dictates the course of infection. Differential expression of miRNAs upon infection with M. tuberculosis, regulates host signaling pathways linked to inflammation, autophagy, apoptosis and polarization of macrophages. Experimental evidence suggests that virulent M. tuberculosis often utilize host miRNAs to promote pathogenicity by restricting host-mediated antibacterial signaling pathways. At the same time, host- induced miRNAs augment antibacterial processes such as autophagy, to limit bacterial proliferation. Targeting miRNAs is an emerging option for host-directed therapies. Recent studies have explored the role of long non-coding RNA (lncRNAs) in the regulation of the host response to mycobacterial infection. Among other functions, lncRNAs interact with chromatin remodelers to regulate gene expression and also function as miRNA sponges. In this review we attempt to summarize recent literature on how miRNAs and lncRNAs are differentially expressed during the course of M. tuberculosis infection, and how they influence the outcome of infection. We also discuss the potential use of non-coding RNAs as biomarkers of active and latent tuberculosis. Comprehensive understanding of the role of these non-coding RNAs is the first step towards developing RNA-based therapeutics and diagnostic tools for the treatment of TB.
Collapse
Affiliation(s)
| | - Joyoti Basu
- Department of Chemistry, Bose Institute, Kolkata, India
| |
Collapse
|
9
|
Wei L, Liu K, Jia Q, Zhang H, Bie Q, Zhang B. The Roles of Host Noncoding RNAs in Mycobacterium tuberculosis Infection. Front Immunol 2021; 12:664787. [PMID: 34093557 PMCID: PMC8170620 DOI: 10.3389/fimmu.2021.664787] [Citation(s) in RCA: 9] [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: 02/06/2021] [Accepted: 04/23/2021] [Indexed: 12/21/2022] Open
Abstract
Tuberculosis remains a major health problem. Mycobacterium tuberculosis, the causative agent of tuberculosis, can replicate and persist in host cells. Noncoding RNAs (ncRNAs) widely participate in various biological processes, including Mycobacterium tuberculosis infection, and play critical roles in gene regulation. In this review, we summarize the latest reports on ncRNAs (microRNAs, piRNAs, circRNAs and lncRNAs) that regulate the host response against Mycobacterium tuberculosis infection. In the context of host-Mycobacterium tuberculosis interactions, a broad and in-depth understanding of host ncRNA regulatory mechanisms may lead to potential clinical prospects for tuberculosis diagnosis and the development of new anti-tuberculosis therapies.
Collapse
Affiliation(s)
- Li Wei
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Kai Liu
- Nursing Department, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Qingzhi Jia
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Hui Zhang
- Institute of Immunology and Molecular Medicine, Jining Medical University, Jining, China
| | - Qingli Bie
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| | - Bin Zhang
- Department of Laboratory Medicine, Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
- Institute of Forensic Medicine and Laboratory Medicine, Jining Medical University, Jining, China
| |
Collapse
|
10
|
Fan K, Shen Y, Xu X, Tao L, Bao T, Li J. LncRNA-WAS and lncRNA-C8807 interact with miR-142a-3p to regulate the inflammatory response in grass carp. FISH & SHELLFISH IMMUNOLOGY 2021; 111:201-207. [PMID: 33582280 DOI: 10.1016/j.fsi.2021.02.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/30/2021] [Accepted: 02/02/2021] [Indexed: 06/12/2023]
Abstract
Septicemia of grass carp is a systemic inflammatory reaction caused by bacterial infection. More and more evidences show that long non-coding RNAs (lncRNAs) can participate in the regulation of inflammatory response. In the present study, lncRNA-WAS and lncRNA-C8807 were confirmed to be involved in the inflammatory response following infection with Aeromonas hydrophila. LncRNA-WAS and lncRNA-C8807 could interact with miR-142a-3p. LncRNA-WAS and lncRNA-C8807 interact with miR-142a-3p to effect pro-inflammatory genes and NF-κB pathway. Our results provide a theoretical basis for studying the molecular mechanism underlying the regulation of inflammation by lncRNA in grass carp.
Collapse
Affiliation(s)
- Kun Fan
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China
| | - Yubang Shen
- National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China
| | - Xiaoyan Xu
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China.
| | - Lizhu Tao
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China
| | - Tianjie Bao
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China
| | - Jiale Li
- Key Laboratory of Freshwater Aquatic Genetic Resources Ministry of Agriculture and Rural Affairs, Shanghai Ocean University, Shanghai, China; Shanghai Engineering Research Center of Aquaculture, Shanghai Ocean University, Shanghai, China; National Demonstration Center for Experimental Fisheries Science Education, Shanghai Ocean University, Shanghai, China.
| |
Collapse
|
11
|
Zhang S, Li L, Wang J, Zhang T, Ye T, Wang S, Xing D, Chen W. Recent advances in the regulation of ABCA1 and ABCG1 by lncRNAs. Clin Chim Acta 2021; 516:100-110. [PMID: 33545111 DOI: 10.1016/j.cca.2021.01.019] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Revised: 01/24/2021] [Accepted: 01/26/2021] [Indexed: 02/07/2023]
Abstract
Coronary heart disease (CHD) with atherosclerosis is the leading cause of death worldwide. ABCA1 and ABCG1 promote cholesterol efflux to suppress foam cell generation and reduce atherosclerosis development. Long noncoding RNAs (lncRNAs) are emerging as a unique group of RNA transcripts that longer than 200 nucleotides and have no protein-coding potential. Many studies have found that lncRNAs regulate cholesterol efflux to influence atherosclerosis development. ABCA1 is regulated by different lncRNAs, including MeXis, GAS5, TUG1, MEG3, MALAT1, Lnc-HC, RP5-833A20.1, LOXL1-AS1, CHROME, DAPK1-IT1, SIRT1 AS lncRNA, DYNLRB2-2, DANCR, LeXis, LOC286367, and LncOR13C9. ABCG1 is also regulated by different lncRNAs, including TUG1, GAS5, RP5-833A20.1, DYNLRB2-2, ENST00000602558.1, and AC096664.3. Thus, various lncRNAs are associated with the roles of ABCA1 and ABCG1 on cholesterol efflux in atherosclerosis regulation. However, some lncRNAs play dual roles in ABCA1 expression and atherosclerosis, and the functions of some lncRNAs in atherosclerosis have not been investigated in vivo. In this article, we review the roles of lncRNAs in atherosclerosis and focus on new insights into lncRNAs associated with the roles of ABCA1 and ABCG1 on cholesterol efflux and the potential of these lncRNAs as novel therapeutic targets in atherosclerosis.
Collapse
Affiliation(s)
- Shun Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Lu Li
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Jie Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Tingting Zhang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Ting Ye
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Shuai Wang
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Medical Imaging, Radiotherapy Department of Affiliated Hospital, Weifang Medical University, Weifang, Shandong 261053, China
| | - Dongming Xing
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Wujun Chen
- Cancer Institute, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China.
| |
Collapse
|