Acessibilidade / Reportar erro

The expression patterns of MALAT-1, NEAT-1, THRIL, and miR-155-5p in the acute to the post-acute phase of COVID-19 disease

ABSTRACT

Introduction:

One of the hallmarks of COVID-19 is overwhelming inflammation, which plays a very important role in the pathogenesis of COVID-19. Thus, identification of inflammatory factors that interact with the SARS-CoV-2 can be very important to control and diagnose the severity of COVID-19. The aim of this study was to investigate the expression patterns of inflammation-related non-coding RNAs (ncRNAs) including MALAT-1, NEAT-1, THRIL, and miR-155-5p from the acute phase to the recovery phase of COVID-19.

Methods:

Total RNA was extracted from Peripheral Blood Mononuclear Cell (PBMC) samples of 20 patients with acute COVID-19 infection and 20 healthy individuals and the expression levels of MALAT-1, NEAT-1, THRIL, and miR-155-5p were evaluated by real-time PCR assay. Besides, in order to monitor the expression pattern of selected ncRNAs from the acute phase to the recovery phase of COVID-19 disease, the levels of ncRNAs were re-measured 6–7 weeks after the acute phase.

Result:

The mean expression levels of MALAT-1, THRIL, and miR-155-5p were significantly increased in the acute phase of COVID-19 compared with a healthy control group. In addition, the expression levels of MALAT-1 and THRIL in the post-acute phase of COVID-19 were significantly lower than in the acute phase of COVID-19. According to the ROC curve analysis, these ncRNAs could be considered useful biomarkers for COVID-19 diagnosis and for discriminating between acute and post-acute phase of COVID-19.

Discussion:

Inflammation-related ncRNAs (MALAT-1, THRIL, and miR-150-5p) can act as hopeful biomarkers for the monitoring and diagnosis of COVID-19 disease.

Keywords:
COVID-19; Long non-coding RNAs; MicroRNAs; Inflammation; Biomarker

Introduction

The Coronavirus Disease Pandemic of 2019 (COVID-19) is caused by a novel coronavirus known as Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2). At the moment, 239 million individuals have been infected with SARS-CoV-2, and much more than 4.8 million have died as a result of this infection.11 Sarvepalli SS, Cruz ABV, Chopra T, Salimnia H, Chandrasekar P. Striking absence of “usual suspects” during the winter of the Coronavirus Disease 2019 (COVID-19) pandemic 2020-2021. Infect Control Hospital Epidemiol. 2021: 1–2. In addition, the epidemic has triggered worldwide social and economic instability. SARS-CoV-2 is an enveloped virus with a positive non-segmented single-stranded RNA genome which belongs to Nidovirales order, Coronaviridae family, Betacoronavirus genus, and lineage B.22 Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019;17:181–92. Because of the worldwide severity of the pathogen, the increased infection rate of SARS-CoV-2, and the limitation of effective treatment approaches, more research is needed to properly control their spread and to offer treatment alternatives.

Over than 90% of the human DNA sequence is constantly transcribed, but only 2% of it produces proteins. The vast majority of transcripts are classified as non-coding RNAs (ncRNAs). According to their sequence length, ncRNAs are classified into long non-coding RNA (lncRNA) with size larger than 200 nucleotides (nt) and small non-coding RNA (sncRNA) with length less than 200 nt, such as microRNAs, which are both important epigenetic and sub-cellular regulatory elements that can be involved in complex cellular biological processes.33 Zhao Z, Sun W, Guo Z, Zhang J, Yu H, Liu B. Mechanisms of lncRNA/microRNA interactions in angiogenesis. Life Sci. 2020;254:116900. Reportedly, lncRNAs may play essential regulatory functions in the interaction between virus and host, including regulation of host antiviral responses, direct and indirect roles in viral and host gene transcription, as well as regulation of the stability and translation of mRNAs.44 Liu W, Ding C. Roles of LncRNAs in viral infections. Front Cell Infect Microbiol. 2017;7:205. Also, it has been found that viral proteins can influence the expression level of cellular lncRNAs and microRNAs (miRNAs). As a result, changes in the expression level of these factors, directly and/ or indirectly, can affect viral infection through regulating host innate immune responses, such as inflammation, and by regulating expression of both cellular and viral genes.55 Potenza N, Papa U, Mosca N, Zerbini F, Nobile V, Russo A. Human microRNA hsa-miR-125a-5p interferes with expression of hepatitis B virus surface antigen. Nucl Acids Res. 2011;39:5157–63.,66 Chen L, Zhou Y, Li H. LncRNA, miRNA and lncRNA-miRNA interaction in viral infection. Virus Res. 2018;257:25–32.,77 Yousefpouran S, Mostafaei S, Manesh PV, Iranifar E, Bokharaei-Salim F, Nahand JS, et al. The assessment of selected MiRNAs profile in HIV, HBV, HCV, HIV/HCV, HIV/HBV Co-infection and elite controllers for determination of biomarker. Microb Pathog. 2020;147:104355.

MicroRNA-155-5p (miR-155-5p) has been characterized as an ancient immune cell regulator. MiR-155-5p is remarkable in the immune system because it can change the transcription of activated myeloid and lymphoid cells, regulating a wide range of biological processes from inflammation to immune response.88 Vigorito E, Kohlhaas S, Lu D, Leyland R. miR-155: an ancient regulator of the immune system. Immunol Rev. 2013;253:146–57. Numerous research in recent years have demonstrated that miR-155-5p is evolutionarily conserved. Its expression is continuously increased in diverse cellular systems during viral infections in both animal and human models.99 O’Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA. 2007;104 (5):1604–9.,1010 Goncalves-Alves E, Saferding V, Schliehe C, Benson R, Kurowska-Stolarska M, Brunner JS, et al. MicroRNA-155 controls t helper cell activation during viral infection. Front Immunol. 2019;10:1367. Similarly, in animal models of Acute Respiratory Distress Syndrome (ARDS), increased miR-155-5p is associated with respiratory infections, illness severity, and greater mortality.1111 Wu Z, Hao R, Li P, Zhang X, Liu N, Qiu S, et al. MicroRNA expression profile of mouse lung infected with 2009 pandemic H1N1 influenza virus. PLoS One. 2013;8:e74190.,1212 Woods PS, Doolittle LM, Rosas LE, Nana-Sinkam SP, Tili E, Davis IC. Increased expression of microRNA-155-5p by alveolar type II cells contributes to development of lethal ARDS in H1N1 influenza A virus-infected mice. Virology. 2020;545:40–52. Overall, evidence strongly suggests that miR-155 has a critical role as regulator of inflammation1313 Xiaoyan W, Pais EMA, Lan L, Jingrui C, Lin M, Fordjour PA, et al. MicroRNA-155: a novel armamentarium against inflammatory diseases. Inflammation. 2017;40:708–16. and during most viral infections, since the expression level of miR-155 is upregulated and regulates antiviral immune responses.1414 Jafarzadeh A, Naseri A, Shojaie L, Nemati M, Jafarzadeh S, Baghi HB, et al. MicroRNA-155 and antiviral immune responses. Int Immunopharmacol. 2021;101:108188.

Nuclear Factor-kappa B (NF-kB) is a dimeric transcription factor involved in inflammation and has an important role in pathogenesis of several inflammatory disease such as Chronic Obstructive Pulmonary Disease (COPD) and COVID-19.1515 Alharbi KS, Fuloria NK, Fuloria S, Rahman SB, Al-Malki WH, Shaikh MAJ, et al. Nuclear factor-kappa B and its role in inflammatory lung disease. Chemico-Biolog Interact. 2021:109568. Numerous ncRNAs such as miR-155-5p, MALAT-1, NEAT-1 and THRIL are involved in regulating the NF-kB signaling pathway.1616 Chew CL, Conos SA, Unal B, Tergaonkar V. Noncoding RNAs: master regulators of inflammatory signaling. Trends Molec Med. 2018;24:66–84. It has been reported that lncRNA MALAT-1 can control cytokine secretion in macrophages under inflammatory circumstances and promote inflammatory activity by interacting with the NF-kB signaling pathway.1717 Gong Y-P, Zhang Y-W, Su X-Q, Gao H-B. Inhibition of long noncoding RNA MALAT1 suppresses high glucose-induced apoptosis and inflammation in human umbilical vein endothelial cells by suppressing the NF-κB signaling pathway. Biochem Cell Biol. 2020;98:669–75.,1818 Zhou H-J, Wang L-Q, Wang D-B, Yu J-B, Zhu Y, Xu Q-S, et al. Long noncoding RNA MALAT1 contributes to inflammatory response of microglia following spinal cord injury via the modulation of a miR-199β/IKKb/NF-κB signaling pathway. Am J Physiol-Cell Physiol. 2018;315:C52–61. NEAT-1 is another lncRNA that has been shown to play a role in NF-kB signaling pathway and NEAT-1 inhibition prevented the activation of the NF-kB pathway.1919 Chen Y, Qiu J, Chen B, Lin Y, Chen Y, Xie G, et al. Long non-coding RNA NEAT1 plays an important role in sepsis-induced acute kidney injury by targeting miR-204 and modulating the NF-κB pathway. Inter Immunopharmacol. 2018;59:252–60. and induced expression of inflammatory-related cytokines such as IL-8 and IL-6.2020 Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. 2019;10:1495. Reportedly, NEAT-1 probably can help the inflammation-regulating ncRNA-mRNA network, and some factors linked with this network may be able to regulate inflammation by interacting with essential inflammatory mediators such as IL-6, TNF and muscarinic acetylcholine receptors.2121 Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–62.,2222 Shenhar-Tsarfaty S, Yayon N, Waiskopf N, Shapira I, Toker S, Zaltser D, et al. Fear and C-reactive protein cosynergize annual pulse increases in healthy adults. Proc Natl Acad Sci USA. 2015;112:E467–71.,2323 Meydan C, Madrer N, Soreq H. The neat dance of COVID-19: NEAT1, DANCR, and Co-modulated cholinergic RNAs link to inflammation. Front Immunol. 2020;11:590870. THRIL is a newly described lncRNA that has been confirmed to interact with hnRNPL (Heterogeneous Nuclear Ribonucleoprotein L) and then controlling the expression of TNF-a with an important role in regulation of inflammation and immune response.2424 Xu B, Jin X, Yang T, Zhang Y, Liu S, Wu L, et al. Upregulated lncRNA THRIL/TNF-α signals promote cell growth and predict poor clinical outcomes of osteosarcoma. OncoTargets Therapy. 2020;13:119.,2525 Li Z, Chao T-C, Chang K-Y, Lin N, Patil VS, Shimizu C, et al. The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Nat Acad Sci. 2014;111:1002–7. THRIL induce the upregulation of NRP1 expression and further induce the modulation of the NF-kB signaling pathway.2626 Kuai F, Zhou L, Zhou J, Sun X, Dong W. Long non-coding RNA THRIL inhibits miRNA-24-3p to upregulate neuropilin-1 to aggravate cerebral ischemia-reperfusion injury through regulating the nuclear factor κB p65 signaling. Aging (Albany NY). 2021;13:9071. As a result, the interaction between lncRNAs and targets (e.g., miRNAs, cellular factors and viral genes) has sparked researchers’ interests to investigate the potential biomarkers and/or therapeutic targets. Currently, our understanding of SARS-CoV-2 processes is limited, and there are no particular biomarkers associated with SARS-CoV-2 diagnosis or therapy. Since selected cellular ncRNAs (miR-155-5p,2727 Dickey LL, Hanley TM, Huffaker TB, Ramstead AG, O’Connell RM, Lane TE. MicroRNA 155 and viral-induced neuroinflammation. J Neuroimmunol. 2017;308:17–24.,2828 Tili E, Michaille JJ, Wernicke D, Alder H, Costinean S, Volinia S, et al. Mutator activity induced by microRNA-155 (miR-155) links inflammation and cancer. Proc Natl Acad Sci USA. 2011;108:4908–13.,2929 Bhattacharyya S, Balakathiresan NS, Dalgard C, Gutti U, Armistead D, Jozwik C, et al. Elevated miR-155 promotes inflammation in cystic fibrosis by driving hyperexpression of interleukin-8. J Biol Chem. 2011;286:11604–15.,3030 Aloi MS, Prater KE, Sopher B, Davidson S, Jayadev S, Garden GA. The pro-inflammatory microRNA miR-155 influences fibrillar β-Amyloid(1) (-42) catabolism by microglia. Glia. 2021;69:1736–48.,3131 Zhou Y, Ma XY, Han JY, Yang M, Lv C, Shao Y, et al. Metformin regulates inflammation and fibrosis in diabetic kidney disease through TNC/TLR4/NF-κB/miR-155-5p inflammatory loop. World J Diabetes. 2021;12:19–46. MALAT-1,3232 Puthanveetil P, Chen S, Feng B, Gautam A, Chakrabarti S. Long non-coding RNA MALAT1 regulates hyperglycaemia induced inflammatory process in the endothelial cells. J Cell Mol Med. 2015;19:1418–25.,3333 Zhao G, Su Z, Song D, Mao Y, Mao X. The long noncoding RNA MALAT1 regulates the lipopolysaccharide-induced inflammatory response through its interaction with NF-κB. FEBS Lett. 2016;590:2884–95.,3434 Yang H, Liang N, Wang M, Fei Y, Sun J, Li Z, et al. Long noncoding RNA MALAT-1 is a novel inflammatory regulator in human systemic lupus erythematosus. Oncotarget. 2017;8:77400–6.,3535 Biswas S, Thomas AA, Chen S, Aref-Eshghi E, Feng B, Gonder J, et al. MALAT1: an epigenetic regulator of inflammation in diabetic retinopathy. Sci Rep. 2018;8:6526. NEAT-12020 Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. 2019;10:1495.,3636 Wang SM, Liu GQ, Xian HB, Si JL, Qi SX, Yu YP. LncRNA NEAT1 alleviates sepsis-induced myocardial injury by regulating the TLR2/NF-κB signaling pathway. Eur Rev Med Pharmacol Sci. 2019;23:4898-907.,3737 Dai W, Wang M, Wang P, Wen J, Wang J, Cha S, et al. lncRNA NEAT1 ameliorates LPS-induced inflammation in MG63 cells by activating autophagy and suppressing the NLRP3 inflammasome. Int J Mol Med. 2021;47:607-20. and THRIL3838 Li Z, Chao TC, Chang KY, Lin N, Patil VS, Shimizu C, et al. The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Natl Acad Sci USA. 2014;111:1002-7.,3939 Xu B, Jin X, Yang T, Zhang Y, Liu S, Wu L, et al. Upregulated lncRNA THRIL/TNF-α signals promote cell growth and predict poor clinical outcomes of osteosarcoma. Onco Targets Ther. 2020;13:119-29.) may play critical roles in immune response regulation and inflammation, we evaluated the expression pattern of lncRNAs (MALAT-1, NEAT-1 and THRIL) and miR-155-5p in Peripheral Blood Mononuclear Cells (PBMC) of SARS-CoV-2 infected individuals in both acute and post-acute stages and compared to healthy individuals.

Patients and methods

Patients’ selection

From June 2021 to July 2021, 20 patients with COVID-19 infection were recruited from the West Health Center in Tehran (related to Iran University of Medical Sciences [IUMS]) and enrolled in this cross-sectional survey. A peripheral blood sample of 6 μL was collected from these patients during the acute phase and again in the post-acute phase, and from 20 healthy controls.

It should be noted that the studied participants did not have co-infections with Human Immunodeficiency Virus (HIV), Human Cytomegalovirus (HCMV), Hepatitis B Virus (HBV), and Hepatitis C Virus (HCV), and Mycobacterium tuberculosis. Furthermore, none of the subjects had underlying medical conditions.

Ethical issues

This study was approved by the ethics committee of IUMS (ethical code: IR. IUMS. REC.1400.381), and all of the participants filled written informed consent for blood specimen collection.

Preparation of peripheral blood mononuclear cells (PBMCs)

Collected peripheral blood from each subject was transferred into a tube containing Ethylenediaminetetraacetic Acid (EDTA) as anticoagulant and then separated by centrifugation. PBMCs were isolated based on the ficoll hypaque density gradient centrifugation (Lympholyte-H, Cedarlane, Hornby, Canada) technique according to the manufacturer’s instructions, and then the pellet of PBMCs was washed three times with phosphate-buffered saline (pH: 7.3±0.1), and finally resuspended with 350 μL of RNA maintenance solution (RNA-Later [Ambion, Inc., Austin, TX]), and kept at -80°C until extraction of the total RNA.

Total RNA isolation and complementary DNA (cDNA) synthesis

Total RNA was extracted from PBMC samples according to the manufacturer’s protocols with minor modifications. Briefly, after PBMC lysis with 1 μL QIAzol solution, 250 μL of chloroform was added to the lysate, shaken vigorously for one minute, and after 5‒10 minutes of incubation at room temperature, centrifuged at 12,000 × g for 15 minutes at 4°C. The supernatant was aspirated and approximatel 800 μl iso-propanol was added and placed in the freezer overnight. The samples were centrifuged at 12,000 × g for 45 minutes at 4°C. One μL ethanol (100%) was added to the RNA pellet and the microtubes went up and down several times and centrifuged at 12,000 x g for 15 minutes at 4°C. The pellet of RNA was air-dried and dissolved with RNase/DNase free distilled water. The integrity and purity of the isolated RNA was evaluated using a Nano-Drop spectrophotometer (Thermo Scientific, Wilmington, MA) instrument, and then kept at -80°C until the test.

To determine the expression pattern of lncRNAs (MALAT-1,4040 Zhang K, Mao T, He Z, Wu X, Peng Y, Chen Y, et al. Serum MALAT1 assumes signifying capacity in gastric cancer diagnosis. 2020. NEAT-14141 Galamb O, Barták BK, Kalmár A, Nagy ZB, Szigeti KA, Tulassay Z, et al. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol. 2019;25:5026. and THRIL,4242 Qi H, Shen J, Zhou W. Up-regulation of long non-coding RNA THRIL in coronary heart disease: prediction for disease risk, correlation with inflammation, coronary artery stenosis, and major adverse cardiovascular events. J Clin Lab Anal. 2020;34: e23196.), as well as GAPDH and b-actin (as normalization controls for relative quantification),4242 Qi H, Shen J, Zhou W. Up-regulation of long non-coding RNA THRIL in coronary heart disease: prediction for disease risk, correlation with inflammation, coronary artery stenosis, and major adverse cardiovascular events. J Clin Lab Anal. 2020;34: e23196.,4343 Sun Y, Wen Y, Ruan Q, Yang L, Huang S, Xu X, et al. Exploring the association of long noncoding RNA expression profiles with intracranial aneurysms, based on sequencing and related bioinformatics analysis. BMC Med Genomics. 2020;13:147. cDNA was synthesized using 350 ng of the total RNA as previously described in detail.4444 Dehghani-Dehej F, Hosseini Z, Mortazkar P, Khanaliha K, Esghaei M, Fakhim A, et al. Prevalence of HCV and/or HBV coinfection in Iranian HIV-infected patients. Future Virol. 2020;15:155-63.

Expression analysis of genes using real-time PCR

The expression patterns of lncRNAs (MALAT-1,4040 Zhang K, Mao T, He Z, Wu X, Peng Y, Chen Y, et al. Serum MALAT1 assumes signifying capacity in gastric cancer diagnosis. 2020. NEAT-14141 Galamb O, Barták BK, Kalmár A, Nagy ZB, Szigeti KA, Tulassay Z, et al. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol. 2019;25:5026. and THRIL4242 Qi H, Shen J, Zhou W. Up-regulation of long non-coding RNA THRIL in coronary heart disease: prediction for disease risk, correlation with inflammation, coronary artery stenosis, and major adverse cardiovascular events. J Clin Lab Anal. 2020;34: e23196.), and also GAPDH (the expression level of this housekeeping gene was considered as reference gene)4545 Nakamura R, Ishii H, Endo K, Hotta A, Fujii E, Miyazawa K, et al. Reciprocal expression of Slug and Snail in human oral cancer cells. PLoS One. 2018;13:e0199442. were determined by real-time Polymerase Chain Reaction (PCR) using a Rotorgene Q thermal cycler (Qiagen, Hilden, Germany) instrument. The assays were done on 20 μL reaction mixture including: 10 pmol of each primer (MALAT-1, NEAT-1, THRIL, GAPDH and β-actin), 8 μL nuclease free distilled water, 10 μL 2 × SYBR® Premix Ex Taq (Tli Plus) Master Mix (TaKaRa Bio Inc. Shiga, Japan) (Table 1) and one μL of cDNA as template.

Table 1
Primers used in this study for determining of expression profile of long non-coding RNAs (lncRNAs).

The thermocycling conditions for real-time PCR were defined as follows: initial denaturing at 95°C for 15 minutes, and 40 cycles, including 15 seconds at 95°C, 30 seconds at 60° C, and 20 seconds at 72°C. The 2−ΔΔCT method was used for calculation of the relative expression values. All the specimens were tested in duplicate reactions.

miRNA-155-5p expression analysis

Total RNA was extracted (as described in the previous section) from PBMC samples. Complementary DNA (cDNA) was synthesized using 5 μg of the total RNA as previously described in detail.4646 Donyavi T, Bokharaei-Salim F, Baghi HB, Khanaliha K, Alaei Janat-Makan M, Karimi B, et al. Acute and post-acute phase of COVID-19: analyzing expression patterns of miRNA-29a-3p, 146a-3p, 155-5p, and let-7b-3p in PBMC. Inter Immunopharmacol. 2021;97:107641. In the current study the expression of miR-155-5p was evaluated in PBMC specimens of SARA-CoV-2 infected patients in the two stages of disease (acute and post-acute phase) and of healthy controls based on available information.

The real time PCR assay was carried out in final 20 μL volume, including 0.5 μL of specific forward primer, 0.5 μL of universal reverse primer, 10 μL of SYBR Green PCR Master Mix (TaKaRa, Kusatsu, Japan), 8 μL of nuclease-free water, and one μL of cDNA as template. The thermal profile of this assay (three steps with melt) was set at 95°C for two minutes as hold time, followed by 40 cycles of denaturation at 95°C for 15 seconds, annealing at 60°C for 20 seconds, and extension at 72°C for 25 seconds, and also melting curve analysis was determined at temperatures ranging from 55 to 99°C. This assay was performed using the Rotorgene Q thermal cycler (Qiagen, Hilden, Germany) instrument. The expression levels of miR-155-5p was normalized to Snord47 and 68 as reference RNA and the fold change was calculated by the Livak method.4747 Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2− ΔΔCT method. Methods. 2001;25:402-8. It should be noted that all reactions were done in triplicate.

Statistical analysis

Data were analyzed using SPSS version 16 (SPSS Inc., Chicago, IL, USA) and Prism 6.0 software (GraphPad, San Diego, CA, USA). Clinical and demographics characteristics were presented as n (%) for categorical variables and mean ± Standard Deviations (SD) for age, which were analyzed by Fisher’s exact test and Student t-Test, respectively. The Mann-Whitney U-test or the independent-samples t-test was used to compare the mean expression levels of ncRNAs (MALAT-1, NEAT-1, THRIL and miR-155-5p) between the COVID-19 groups with the healthy control group. The statistical difference of the mean level of ncRNAs between acute and post-acute phases of COVID-19 disease was compared using the paired sample t-test. The Receiver Operating Characteristic (ROC) curve analysis was performed to evaluate the diagnostic value of ncRNA expression level in discriminating between study groups. The Spearman rank correlation was used to compare the association of variables. The Benjamini and Hochberg procedure was used to control for the false discovery rate. All statistical evaluations were two-tailed, and p-values less than 0.05 were considered significant.

Results

Characteristics of participants

Fifty SARS-CoV-2 infected patients (in both acute and post-acute phases of the disease) and 50 healthy individuals were enrolled in this cross-sectional study. These two studied groups were matched for sex and age. The mean age of studied patients with SARS CoV-2 infection was 36.1±10.9 (ranging between 22‒67 years) and for healthy individuals was 36.2±12.1 (ranging between 23‒67 years). The demographic parameters of the studied participants and clinical manifestations of patients with SARS-CoV-2 infection are summarized in Table 2.

Table 2
The demographic parameters of the studied participants and clinical manifestations of patients with SARS-CoV-2 infection.

Expression pattern of ncRNAs was significantly deregulated during COVID-19 disease

The expression level of MALAT-1, NEAT-1, THRIL, and miR-155-5p were examined in PBMC samples during acute and post-acute COVID-19 disease and healthy control subjects. In acute COVID-19 group, expression profiles of MALAT-1, THRIL and miR-155-5P were found significantly deregulated (p-value of < 0.05) when compared with healthy controls. In comparison to the control group, the acute COVID-19 group showed higher expression levels: 3.42-fold for the miR-155-5p, 2.27fold for THRIL, and 2.12-fold for MALAT-1, respectively. Also, there was no significant difference in the mean expression level of NEAT-1 between acute COVID-19 group and control group (Fig. 1A), (p-value > 0.05). The expression pattern of lncRNAs (MALAT-1, NEAT-1 and THRIL) in post-acute COVID-19 group was similar to the healthy control group, as shown in Figure 1A. However, the mean expression level of miR-155-5p in post-acute COVID-19 was higher than that of control subjects (2.49-fold change, p-value < 0.0001).

Fig. 1
Comparison of expression level of ncRNAs between (A) Acute and post-acute COVID-19 groups with healthy controls and between (B) acute COVID-19 groups with post-acute COVID-19 groups (ns, not significant, * p < 0.05; *** p < 0.001; **** p < 0.0001).

In order to identify a PBMC biomarker that could be applicable to distinguish the prognosis of COVID-19 disease, the PBMC level of the selected lncRNAs (MALAT-1, NEAT-1 and THRIL) and miR-155-5p were re-measured 6‒5 weeks after the acute phase of COVID-19. Mean expression level of MALAT-1 and THRIL were significantly down-regulated (-1.8-fold, p-value = 0.037 and -1.98-fold, p-value = 0.022, respectively) in post-acute phase of COVID-19 compared to acute phase of COVID-19 disease (Fig. 1B). However, there was no significant difference in the expression pattern of miR-155-5p, and THRIL between acute COVID-19 group and post-acute COVID-19 group (p-value > 0.05).

In the current study, the potential of ncRNAs (miR-155-5p, MALAT-1, NEAT-1 and THRIL) in discriminating between COVID-19 groups and healthy controls and between acute phase of COVID-19 and post-acute phase of COVID-19 was evaluated by the ROC curve analysis. According to results which are illustrated in Figure 2, miR-155-5p (AUC = 0.89, p-value < 0.0001), MALAT-1 (AUC = 0.81, p-value < 0.0001) and THRIL (AUC = 0.77, p-value = 0.003) are effective in distinguishing acute phase of COVID-19 from healthy controls. In the case of post-acute COVID-19 phase compared with healthy controls, the AUC value for miR-155-5p was 0.83 (p-value < 0.0001). Especially, the miR-155-5p showed an excellent AUC value. Lastly, PBMC THRIL and MALAT-1 were able to distinguish acute phase of COVID-19 from post-acute phase of COVID-19 disease with AUC value of 0.75 (p-value = 0.005) and 0.72 (p-value = 0.021), respectively (Fig. 2).

Fig. 2
ROC analysis for evaluating the diagnostic ability of ncRNAs to discriminate SARS-CoV-2 infected group from uninfected groups (AUC, Area Under the Curve; P, p-value).

The correlation between clinical characteristics and relative expressions of ncRNAs

Hsa-miR-155-5p is one of the cellular miRNAs that maybe play a critical role in regulating inflammation and antiviral cellular defense in SARS-CoV-2 infection.4848 Soni DK, Cabrera-Luque J, Kar S, Sen C, Devaney J, Biswas R. Suppression of miR-155 attenuates lung cytokine storm induced by SARS-CoV-2 infection in human ACE2-transgenic mice. bioRxiv. 2020.,4949 Mahesh G, Biswas R. MicroRNA-155: a master regulator of inflammation. J Interferon Cytokine Res. 2019;39:321–30. Furthermore, it has been documented that some cellular lncRNAs, such as MALAT-1, cause modulating miR-155-5p expression.4949 Mahesh G, Biswas R. MicroRNA-155: a master regulator of inflammation. J Interferon Cytokine Res. 2019;39:321–30. For this reason, to determine the correlation between the expression level of lncRNAs (MALAT-1, NEAT-1 and THRIL) and miR-155-5p, Spearman correlation analysis was performed. According to the result, a negative correlation was found between MALAT-1 level and level of miR-155-5p (r = -0.52, p-value < 0.0001), (Fig. 3A). However, there was no significant correlation between expression level of NEAT-1 and THRIL with miR-155-5p expression level (Fig. 2 B and C).

Fig. 3
Spearman’s correlation coefficient between the expression level of lncRNAs (MALAT-1, NEAT-1 and THRIL) with expression level of miR-155-5p.

To investigate the relationship among expression level of ncRNAs with demographic-clinical characteristics and by SARS-CoV-2 (RdRP and N) genes in the COVID-19 patients during acute COVID-19 disease, Spearman correlation coefficient was carried out (Table 3). According to the result, there was a significant negative correlation between delta Ct of miR-155-5p and delta Ct of RdRp (r = -0.7, p-value < 0.001) and N genes (r = -0.61, p-value < 0.01) of SARS-Cov-2. Besides, a significant positive correlation was found among delta Ct of THRIL and NEAT-1 with dry cough (r = 0.57, and r = 0.46, respectively) and with sputum cough (r = 0.42 and r = 0.58, respectively). As well, a significant positive correlation was found between MALAT-1 and fever (r = 0.61, p-value < 0.001), skeletal pain (r = 0.55, p-value < 0.01) More information is provided in Table 3.

Table 3
Spearman’s correlation coefficient between the expression level of ncRNAs with expression level of SARS-CoV-2 genes (N and RdRp genes), demographic and clinical characteristics.

Discussion

Increasing evidence indicated that ncRNAs play a critical role in inflammation-related disorders by regulation of the diverse biological processes, such as the activation of inflammatory pathway signaling.5050 Onyeagucha BC. The contribution of inflammatory pathway signaling and microRNA changes to colon cancer progression: the University of Arizona; 2013.,5151 Wang W, Yang N, Yang Y-H, Wen R, Liu C-F, Zhang T-N. Non-coding RNAs: master regulators of inflammasomes in inflammatory diseases. J Inflam Res. 2021;14:5023–50. Reportedly, ncRNAs cross-talking with immune cells can also regulate inflammation and immunological response. Further, due to growing understanding of the interactions between SARS-CoV-2 with host ncRNAs,5252 Donyavi T, Bokharaei-Salim F, Baghi HB, Khanaliha K, Janat-Makan MA, Karimi B, et al. Acute and post-acute phase of COVID-19: analyzing expression patterns of miRNA-29a-3p, 146a-3p, 155-5p, and let-7b-3p in PBMC. Inter Immunopharmacol. 2021;97:107641.,5353 Plowman T, Lagos D. Non-coding RNAs in COVID-19: emerging insights and current questions. Non-Coding RNA. 2021;7:54. one could suggest that SARS-CoV-2 can alter the immunological pathway by deregulation of ncRNAs expression. Result of this research point out that the expression level of MALAT-1, NEAT-1, THRIL, and miR-155-5p, which are associated with inflammatory response, were significantly different between COVID-19 patients and healthy control subjects, as well as between acute and post-acute phase of COVID-19 disease. Recently, it was shown that expression level of MALAT-1 was significantly upregulated in SARS-CoV-2-infected bronchial epithelial cells.5454 Vishnubalaji R, Shaath H, Alajez NM. Protein coding and long noncoding RNA (lncRNA) transcriptional landscape in SARS-CoV-2 infected bronchial epithelial cells highlight a role for interferon and inflammatory response. Genes. 2020;11:760. In another study, Huang et al.5555 Huang K, Wang C, Vagts C, Raguveer V, Finn P, Perkins DL. LncRNAs NEAT1 and MALAT1 differentiate inflammation in severe COVID-19 patients. medRxiv. 2021. reported that expressions of NEAT-1 and MALAT-1 were significantly increased in severe COVID-19 patients compared to mild COVID-19 patients and they suggested that NEAT-1 and MALAT-1 promote cellular damage and stress.5555 Huang K, Wang C, Vagts C, Raguveer V, Finn P, Perkins DL. LncRNAs NEAT1 and MALAT1 differentiate inflammation in severe COVID-19 patients. medRxiv. 2021. In addition, an in vivo study reported that silencing MALAT-1 inhibited neutrophil chemotaxis by interleukin-8 and suppresses pulmonary epithelial cells apoptosis.5656 Wei L, Li J, Han Z, Chen Z, Zhang Q. Silencing of lncRNA MALAT1 prevents inflammatory injury after lung transplant ischemia-reperfusion by downregulation of IL-8 via p300. Mol Therapy-Nucl Acids. 2019;18:285–97. All of these findings suggest that MALAT-1 is increased in lung cells of COVID-19 patients, promoting immune cell taxi and subsequent harmful inflammation. MALAT-1 expression is also linked to macrophage activation and maturation into the M1 subtype, which is important in numerous pathological events, including inflammation.5757 Cui H, Banerjee S, Guo S, Xie N, Ge J, Jiang D, et al. Long noncoding RNA Malat1 regulates differential activation of macrophages and response to lung injury. JCI Insight. 2019;4(4). Besides, MALAT-1 can promote the expression of Maf and IL-10 in T-helper (Th) cells and eventually suppresses immunity against infection.5858 Hewitson JP, West KA, James KR, Rani GF, Dey N, Romano A, et al. Malat1 suppresses immunity to infection through promoting expression of maf and IL-10 in Th cells. J Immunol. 2020;204:2949–60. Recently, Rodrigues et al.5959 Rodrigues AC, Adamoski D, Genelhould G, Zhen F, Yamaguto GE, de Araujo Souza PS, et al. NEAT1 and MALAT1 are highly expressed in saliva and nasopharyngeal swab sample of COVID-19 patients. Mol Oral Microbiol. 2021. investigated the expression of miR-3142, MALAT-1, and NEAT-1 in nasopharyngeal swab and saliva specimens of COVID-19 patients. They observed that expression levels of the NEAT-1 and MALAT-1 in SARS-CoV-2 positive samples were higher than those of healthy controls. Further, they suggested that salivary NEAT-1 could act as a potential biomarker for distinguishing between healthy subjects from COVID-19 patients (AUC = 0.80).5959 Rodrigues AC, Adamoski D, Genelhould G, Zhen F, Yamaguto GE, de Araujo Souza PS, et al. NEAT1 and MALAT1 are highly expressed in saliva and nasopharyngeal swab sample of COVID-19 patients. Mol Oral Microbiol. 2021. Similarly, our data reveal that the level of MALAT-1 was significantly over-expressed in the acute phase of COVID-19 disease compared to healthy subjects. However, the expression level of MALAT-1 was significantly decreased from the acute phase to the post-acute phase of COVID-19 disease.

NEAT-1, a pro-inflammatory lncRNA which is comparable genomically to MALAT-1, was found to increase inflammation through enhanced inflammasome assembly and processing.6060 Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. 2019;10:1–17. NEAT-1 promote inflammation by induction of inflammatory cytokines such as Interleukin-6 (IL-6). In response to SARS-CoV-2 infection, IL-6 is one of the key immune components.5959 Rodrigues AC, Adamoski D, Genelhould G, Zhen F, Yamaguto GE, de Araujo Souza PS, et al. NEAT1 and MALAT1 are highly expressed in saliva and nasopharyngeal swab sample of COVID-19 patients. Mol Oral Microbiol. 2021. In nine cell types (M1 and M2 type macrophages, monocytes, CD4+ T cells, and CD8+ memory T cells) identified from severe COVID-19 patient Bronchoalveolar Lavage (BAL) samples, there was overexpression of NEAT-1.6161 Huang K, Wang C, Vagts C, Raguveer V, Finn PW, Perkins DL. LncRNAs NEAT1 and MALAT1 differentiate inflammation in severe COVID-19 patients. medRxiv. 2021:2021.03.26.21254445. According to our findings, no significant difference was observed in PBMC level of NEAT-1 between the acute COVID-19 group and the control group, and also the mean expression level of NEAT-1 during the acute phase of COVID-19 was statistically similar to the post-acute phase of the disease. These results suggest that the immunological effect of NEAT-1 may be specific to the lung, i.e., where the infection and inflammation initiate. Besides, different results from previous studies were possible because of differences in the type of samples.

The lncRNA THRIL can be involved in immune response to viral infection largely through regulating TNF-α, IFN-β, IL8 expression and inflammatory response.6262 Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science. 2013;341:789–92. Tumor Necrosis Factor (TNF), an activator of NF-kB signaling pathway, is a major inflammatory cytokine regulator in host defense against viral infection.6363 Newton K, Manning G. Necroptosis and inflammation. Ann Rev Biochem. 2016;85:743–63. THRIL directly modulates TNF-a, whereas THRIL induces other cytokines and chemokines, but the processes need to be further investigated.2525 Li Z, Chao T-C, Chang K-Y, Lin N, Patil VS, Shimizu C, et al. The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Nat Acad Sci. 2014;111:1002–7. For the first time in this study, we explored the expression pattern of lncRNA THRIL in COVID-19. Similar to the expression level of MALAT-1, THRIL was significantly overexpressed in acute COVID-19 group compared to healthy samples. Comparison between acute and post-acute COVID-19 groups, the mean expression level of THRIL was significantly down-regulated during acute to post-acute phase. As well, the AUC value shows that PBMC THRIL can serve as a biomarker in the discrimination of COVID-19 patients from healthy subjects and those in the acute phase of COVID-19 from those in the post-acute phase of COVID-19 disease.

MiR-155-5p has been known as the ‘master of inflammation’ during COVID-19 disease and constitutes part of an immunopathological picture in COVID-19 disease. In inflammatory responses, miR-155-5p controls NF-kB signaling and plays a critical role in the modulating the immune response.4949 Mahesh G, Biswas R. MicroRNA-155: a master regulator of inflammation. J Interferon Cytokine Res. 2019;39:321–30.,6464 Mann M, Mehta A, Zhao JL, Lee K, Marinov GK, Garcia-Flores Y, et al. An NF-κB-microRNA regulatory network tunes macrophage inflammatory responses. Nat Commun. 2017;8 (1):1–13. The miR-155-5p expression is considered to be the initial step in the NF-kB signaling upregulation of the immune cascade and feeding back through the IKK signalosome complex and PI3K/Akt to further increase NF-kB. It has been reported that regulation of miR-155-5p levels by glucocorticoids, can be considered as one of the effective COVID-19 treatments.6565 Group TRC. Dexamethasone in hospitalized patients with COVID-19 ‒ preliminary report. New Engl J Med. 2020. Although the expression level of miR-155-5p was upregulated in COVID-19 patients, there is no literature so far investigating miRNA-155 mechanism in COVID-19.4848 Soni DK, Cabrera-Luque J, Kar S, Sen C, Devaney J, Biswas R. Suppression of miR-155 attenuates lung cytokine storm induced by SARS-CoV-2 infection in human ACE2-transgenic mice. bioRxiv. 2020.,5252 Donyavi T, Bokharaei-Salim F, Baghi HB, Khanaliha K, Janat-Makan MA, Karimi B, et al. Acute and post-acute phase of COVID-19: analyzing expression patterns of miRNA-29a-3p, 146a-3p, 155-5p, and let-7b-3p in PBMC. Inter Immunopharmacol. 2021;97:107641.,6666 Garg A, Seeliger B, Derda AA, Xiao K, Gietz A, Scherf K, et al. Circulating cardiovascular microRNAs in critically ill COVID-19 patients. Euro J Heart Fail. 2021;23:468–75. However, preceding reports has demonstrated that miRNA-155 has a strong impact in NF-kB signaling.6464 Mann M, Mehta A, Zhao JL, Lee K, Marinov GK, Garcia-Flores Y, et al. An NF-κB-microRNA regulatory network tunes macrophage inflammatory responses. Nat Commun. 2017;8 (1):1–13. Our results are in line with previous studies in which the mean expression level of miR-150-5p in acute-COVID-19 subjects are significantly higher than in the healthy subjects and in post-acute COVID-19 group. In addition, according to ROC curve results for miR-150-5p, this miRNA may be considered a novel biomarker for acute COVID-19 disease diagnosis.

Conclusion

According to the findings of the present study, expression pattern of inflammation-related ncRNAs including MALAT-1, NEAT-1, and miR-150-5p were significantly different between COVID-19 patients and healthy subjects. In addition, the level of miR-150-5p, MALAT-1, and NEAT-1 were significantly down-regulated from the acute phase of COVID-19 to the post-acute phase of COVID-19. Aberrant expression of ncRNAs was found in COVID-19 disease, which maybe associated with the pathogenesis of SARS-CoV-2 and identification of these factors can be helpful in setting a basis for classification of disease conditions and acting as biomarkers and even be considered as a valuable therapeutic target for the treatment of COVID-19 diseases. Finally, the low number of samples in this study was the main limitation of our work. Hence, the assessment of these non-coding RNAs in a large population is needed.

Acknowledgments

The authors of the current survey would like to thank all of the volunteers who have participated in this research. The present study was funded by Research Deputy of Iran University of Medical Sciences, Tehran, Iran with Grant number 20975.

References

  • 1
    Sarvepalli SS, Cruz ABV, Chopra T, Salimnia H, Chandrasekar P. Striking absence of “usual suspects” during the winter of the Coronavirus Disease 2019 (COVID-19) pandemic 2020-2021. Infect Control Hospital Epidemiol. 2021: 1–2.
  • 2
    Cui J, Li F, Shi ZL. Origin and evolution of pathogenic coronaviruses. Nat Rev Microbiol. 2019;17:181–92.
  • 3
    Zhao Z, Sun W, Guo Z, Zhang J, Yu H, Liu B. Mechanisms of lncRNA/microRNA interactions in angiogenesis. Life Sci. 2020;254:116900.
  • 4
    Liu W, Ding C. Roles of LncRNAs in viral infections. Front Cell Infect Microbiol. 2017;7:205.
  • 5
    Potenza N, Papa U, Mosca N, Zerbini F, Nobile V, Russo A. Human microRNA hsa-miR-125a-5p interferes with expression of hepatitis B virus surface antigen. Nucl Acids Res. 2011;39:5157–63.
  • 6
    Chen L, Zhou Y, Li H. LncRNA, miRNA and lncRNA-miRNA interaction in viral infection. Virus Res. 2018;257:25–32.
  • 7
    Yousefpouran S, Mostafaei S, Manesh PV, Iranifar E, Bokharaei-Salim F, Nahand JS, et al. The assessment of selected MiRNAs profile in HIV, HBV, HCV, HIV/HCV, HIV/HBV Co-infection and elite controllers for determination of biomarker. Microb Pathog. 2020;147:104355.
  • 8
    Vigorito E, Kohlhaas S, Lu D, Leyland R. miR-155: an ancient regulator of the immune system. Immunol Rev. 2013;253:146–57.
  • 9
    O’Connell RM, Taganov KD, Boldin MP, Cheng G, Baltimore D. MicroRNA-155 is induced during the macrophage inflammatory response. Proc Natl Acad Sci USA. 2007;104 (5):1604–9.
  • 10
    Goncalves-Alves E, Saferding V, Schliehe C, Benson R, Kurowska-Stolarska M, Brunner JS, et al. MicroRNA-155 controls t helper cell activation during viral infection. Front Immunol. 2019;10:1367.
  • 11
    Wu Z, Hao R, Li P, Zhang X, Liu N, Qiu S, et al. MicroRNA expression profile of mouse lung infected with 2009 pandemic H1N1 influenza virus. PLoS One. 2013;8:e74190.
  • 12
    Woods PS, Doolittle LM, Rosas LE, Nana-Sinkam SP, Tili E, Davis IC. Increased expression of microRNA-155-5p by alveolar type II cells contributes to development of lethal ARDS in H1N1 influenza A virus-infected mice. Virology. 2020;545:40–52.
  • 13
    Xiaoyan W, Pais EMA, Lan L, Jingrui C, Lin M, Fordjour PA, et al. MicroRNA-155: a novel armamentarium against inflammatory diseases. Inflammation. 2017;40:708–16.
  • 14
    Jafarzadeh A, Naseri A, Shojaie L, Nemati M, Jafarzadeh S, Baghi HB, et al. MicroRNA-155 and antiviral immune responses. Int Immunopharmacol. 2021;101:108188.
  • 15
    Alharbi KS, Fuloria NK, Fuloria S, Rahman SB, Al-Malki WH, Shaikh MAJ, et al. Nuclear factor-kappa B and its role in inflammatory lung disease. Chemico-Biolog Interact. 2021:109568.
  • 16
    Chew CL, Conos SA, Unal B, Tergaonkar V. Noncoding RNAs: master regulators of inflammatory signaling. Trends Molec Med. 2018;24:66–84.
  • 17
    Gong Y-P, Zhang Y-W, Su X-Q, Gao H-B. Inhibition of long noncoding RNA MALAT1 suppresses high glucose-induced apoptosis and inflammation in human umbilical vein endothelial cells by suppressing the NF-κB signaling pathway. Biochem Cell Biol. 2020;98:669–75.
  • 18
    Zhou H-J, Wang L-Q, Wang D-B, Yu J-B, Zhu Y, Xu Q-S, et al. Long noncoding RNA MALAT1 contributes to inflammatory response of microglia following spinal cord injury via the modulation of a miR-199β/IKKb/NF-κB signaling pathway. Am J Physiol-Cell Physiol. 2018;315:C52–61.
  • 19
    Chen Y, Qiu J, Chen B, Lin Y, Chen Y, Xie G, et al. Long non-coding RNA NEAT1 plays an important role in sepsis-induced acute kidney injury by targeting miR-204 and modulating the NF-κB pathway. Inter Immunopharmacol. 2018;59:252–60.
  • 20
    Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. 2019;10:1495.
  • 21
    Borovikova LV, Ivanova S, Zhang M, Yang H, Botchkina GI, Watkins LR, et al. Vagus nerve stimulation attenuates the systemic inflammatory response to endotoxin. Nature. 2000;405:458–62.
  • 22
    Shenhar-Tsarfaty S, Yayon N, Waiskopf N, Shapira I, Toker S, Zaltser D, et al. Fear and C-reactive protein cosynergize annual pulse increases in healthy adults. Proc Natl Acad Sci USA. 2015;112:E467–71.
  • 23
    Meydan C, Madrer N, Soreq H. The neat dance of COVID-19: NEAT1, DANCR, and Co-modulated cholinergic RNAs link to inflammation. Front Immunol. 2020;11:590870.
  • 24
    Xu B, Jin X, Yang T, Zhang Y, Liu S, Wu L, et al. Upregulated lncRNA THRIL/TNF-α signals promote cell growth and predict poor clinical outcomes of osteosarcoma. OncoTargets Therapy. 2020;13:119.
  • 25
    Li Z, Chao T-C, Chang K-Y, Lin N, Patil VS, Shimizu C, et al. The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Nat Acad Sci. 2014;111:1002–7.
  • 26
    Kuai F, Zhou L, Zhou J, Sun X, Dong W. Long non-coding RNA THRIL inhibits miRNA-24-3p to upregulate neuropilin-1 to aggravate cerebral ischemia-reperfusion injury through regulating the nuclear factor κB p65 signaling. Aging (Albany NY). 2021;13:9071.
  • 27
    Dickey LL, Hanley TM, Huffaker TB, Ramstead AG, O’Connell RM, Lane TE. MicroRNA 155 and viral-induced neuroinflammation. J Neuroimmunol. 2017;308:17–24.
  • 28
    Tili E, Michaille JJ, Wernicke D, Alder H, Costinean S, Volinia S, et al. Mutator activity induced by microRNA-155 (miR-155) links inflammation and cancer. Proc Natl Acad Sci USA. 2011;108:4908–13.
  • 29
    Bhattacharyya S, Balakathiresan NS, Dalgard C, Gutti U, Armistead D, Jozwik C, et al. Elevated miR-155 promotes inflammation in cystic fibrosis by driving hyperexpression of interleukin-8. J Biol Chem. 2011;286:11604–15.
  • 30
    Aloi MS, Prater KE, Sopher B, Davidson S, Jayadev S, Garden GA. The pro-inflammatory microRNA miR-155 influences fibrillar β-Amyloid(1) (-42) catabolism by microglia. Glia. 2021;69:1736–48.
  • 31
    Zhou Y, Ma XY, Han JY, Yang M, Lv C, Shao Y, et al. Metformin regulates inflammation and fibrosis in diabetic kidney disease through TNC/TLR4/NF-κB/miR-155-5p inflammatory loop. World J Diabetes. 2021;12:19–46.
  • 32
    Puthanveetil P, Chen S, Feng B, Gautam A, Chakrabarti S. Long non-coding RNA MALAT1 regulates hyperglycaemia induced inflammatory process in the endothelial cells. J Cell Mol Med. 2015;19:1418–25.
  • 33
    Zhao G, Su Z, Song D, Mao Y, Mao X. The long noncoding RNA MALAT1 regulates the lipopolysaccharide-induced inflammatory response through its interaction with NF-κB. FEBS Lett. 2016;590:2884–95.
  • 34
    Yang H, Liang N, Wang M, Fei Y, Sun J, Li Z, et al. Long noncoding RNA MALAT-1 is a novel inflammatory regulator in human systemic lupus erythematosus. Oncotarget. 2017;8:77400–6.
  • 35
    Biswas S, Thomas AA, Chen S, Aref-Eshghi E, Feng B, Gonder J, et al. MALAT1: an epigenetic regulator of inflammation in diabetic retinopathy. Sci Rep. 2018;8:6526.
  • 36
    Wang SM, Liu GQ, Xian HB, Si JL, Qi SX, Yu YP. LncRNA NEAT1 alleviates sepsis-induced myocardial injury by regulating the TLR2/NF-κB signaling pathway. Eur Rev Med Pharmacol Sci. 2019;23:4898-907.
  • 37
    Dai W, Wang M, Wang P, Wen J, Wang J, Cha S, et al. lncRNA NEAT1 ameliorates LPS-induced inflammation in MG63 cells by activating autophagy and suppressing the NLRP3 inflammasome. Int J Mol Med. 2021;47:607-20.
  • 38
    Li Z, Chao TC, Chang KY, Lin N, Patil VS, Shimizu C, et al. The long noncoding RNA THRIL regulates TNFα expression through its interaction with hnRNPL. Proc Natl Acad Sci USA. 2014;111:1002-7.
  • 39
    Xu B, Jin X, Yang T, Zhang Y, Liu S, Wu L, et al. Upregulated lncRNA THRIL/TNF-α signals promote cell growth and predict poor clinical outcomes of osteosarcoma. Onco Targets Ther. 2020;13:119-29.
  • 40
    Zhang K, Mao T, He Z, Wu X, Peng Y, Chen Y, et al. Serum MALAT1 assumes signifying capacity in gastric cancer diagnosis. 2020.
  • 41
    Galamb O, Barták BK, Kalmár A, Nagy ZB, Szigeti KA, Tulassay Z, et al. Diagnostic and prognostic potential of tissue and circulating long non-coding RNAs in colorectal tumors. World J Gastroenterol. 2019;25:5026.
  • 42
    Qi H, Shen J, Zhou W. Up-regulation of long non-coding RNA THRIL in coronary heart disease: prediction for disease risk, correlation with inflammation, coronary artery stenosis, and major adverse cardiovascular events. J Clin Lab Anal. 2020;34: e23196.
  • 43
    Sun Y, Wen Y, Ruan Q, Yang L, Huang S, Xu X, et al. Exploring the association of long noncoding RNA expression profiles with intracranial aneurysms, based on sequencing and related bioinformatics analysis. BMC Med Genomics. 2020;13:147.
  • 44
    Dehghani-Dehej F, Hosseini Z, Mortazkar P, Khanaliha K, Esghaei M, Fakhim A, et al. Prevalence of HCV and/or HBV coinfection in Iranian HIV-infected patients. Future Virol. 2020;15:155-63.
  • 45
    Nakamura R, Ishii H, Endo K, Hotta A, Fujii E, Miyazawa K, et al. Reciprocal expression of Slug and Snail in human oral cancer cells. PLoS One. 2018;13:e0199442.
  • 46
    Donyavi T, Bokharaei-Salim F, Baghi HB, Khanaliha K, Alaei Janat-Makan M, Karimi B, et al. Acute and post-acute phase of COVID-19: analyzing expression patterns of miRNA-29a-3p, 146a-3p, 155-5p, and let-7b-3p in PBMC. Inter Immunopharmacol. 2021;97:107641.
  • 47
    Livak KJ, Schmittgen TD. Analysis of relative gene expression data using real-time quantitative PCR and the 2− ΔΔCT method. Methods. 2001;25:402-8.
  • 48
    Soni DK, Cabrera-Luque J, Kar S, Sen C, Devaney J, Biswas R. Suppression of miR-155 attenuates lung cytokine storm induced by SARS-CoV-2 infection in human ACE2-transgenic mice. bioRxiv. 2020.
  • 49
    Mahesh G, Biswas R. MicroRNA-155: a master regulator of inflammation. J Interferon Cytokine Res. 2019;39:321–30.
  • 50
    Onyeagucha BC. The contribution of inflammatory pathway signaling and microRNA changes to colon cancer progression: the University of Arizona; 2013.
  • 51
    Wang W, Yang N, Yang Y-H, Wen R, Liu C-F, Zhang T-N. Non-coding RNAs: master regulators of inflammasomes in inflammatory diseases. J Inflam Res. 2021;14:5023–50.
  • 52
    Donyavi T, Bokharaei-Salim F, Baghi HB, Khanaliha K, Janat-Makan MA, Karimi B, et al. Acute and post-acute phase of COVID-19: analyzing expression patterns of miRNA-29a-3p, 146a-3p, 155-5p, and let-7b-3p in PBMC. Inter Immunopharmacol. 2021;97:107641.
  • 53
    Plowman T, Lagos D. Non-coding RNAs in COVID-19: emerging insights and current questions. Non-Coding RNA. 2021;7:54.
  • 54
    Vishnubalaji R, Shaath H, Alajez NM. Protein coding and long noncoding RNA (lncRNA) transcriptional landscape in SARS-CoV-2 infected bronchial epithelial cells highlight a role for interferon and inflammatory response. Genes. 2020;11:760.
  • 55
    Huang K, Wang C, Vagts C, Raguveer V, Finn P, Perkins DL. LncRNAs NEAT1 and MALAT1 differentiate inflammation in severe COVID-19 patients. medRxiv. 2021.
  • 56
    Wei L, Li J, Han Z, Chen Z, Zhang Q. Silencing of lncRNA MALAT1 prevents inflammatory injury after lung transplant ischemia-reperfusion by downregulation of IL-8 via p300. Mol Therapy-Nucl Acids. 2019;18:285–97.
  • 57
    Cui H, Banerjee S, Guo S, Xie N, Ge J, Jiang D, et al. Long noncoding RNA Malat1 regulates differential activation of macrophages and response to lung injury. JCI Insight. 2019;4(4).
  • 58
    Hewitson JP, West KA, James KR, Rani GF, Dey N, Romano A, et al. Malat1 suppresses immunity to infection through promoting expression of maf and IL-10 in Th cells. J Immunol. 2020;204:2949–60.
  • 59
    Rodrigues AC, Adamoski D, Genelhould G, Zhen F, Yamaguto GE, de Araujo Souza PS, et al. NEAT1 and MALAT1 are highly expressed in saliva and nasopharyngeal swab sample of COVID-19 patients. Mol Oral Microbiol. 2021.
  • 60
    Zhang P, Cao L, Zhou R, Yang X, Wu M. The lncRNA Neat1 promotes activation of inflammasomes in macrophages. Nat Commun. 2019;10:1–17.
  • 61
    Huang K, Wang C, Vagts C, Raguveer V, Finn PW, Perkins DL. LncRNAs NEAT1 and MALAT1 differentiate inflammation in severe COVID-19 patients. medRxiv. 2021:2021.03.26.21254445.
  • 62
    Carpenter S, Aiello D, Atianand MK, Ricci EP, Gandhi P, Hall LL, et al. A long noncoding RNA mediates both activation and repression of immune response genes. Science. 2013;341:789–92.
  • 63
    Newton K, Manning G. Necroptosis and inflammation. Ann Rev Biochem. 2016;85:743–63.
  • 64
    Mann M, Mehta A, Zhao JL, Lee K, Marinov GK, Garcia-Flores Y, et al. An NF-κB-microRNA regulatory network tunes macrophage inflammatory responses. Nat Commun. 2017;8 (1):1–13.
  • 65
    Group TRC. Dexamethasone in hospitalized patients with COVID-19 ‒ preliminary report. New Engl J Med. 2020.
  • 66
    Garg A, Seeliger B, Derda AA, Xiao K, Gietz A, Scherf K, et al. Circulating cardiovascular microRNAs in critically ill COVID-19 patients. Euro J Heart Fail. 2021;23:468–75.

Publication Dates

  • Publication in this collection
    15 July 2022
  • Date of issue
    2022

History

  • Received
    15 Jan 2022
  • Accepted
    11 Apr 2022
  • Published
    25 Apr 2022
Brazilian Society of Infectious Diseases Rua Augusto Viana, SN, 6º., 40110-060 Salvador - Bahia - Brazil, Telefax: (55 71) 3283-8172, Fax: (55 71) 3247-2756 - Salvador - BA - Brazil
E-mail: bjid@bjid.org.br