key: cord-0723787-hktwk386 authors: Jafarinejad-Farsangi, Saeideh; Jazi, Maryam Moazzam; Rostamzadeh, Farzaneh; Hadizadeh, Morteza title: High affinity of host human microRNAs to SARS-CoV-2 genome: An in silico analysis date: 2020-11-21 journal: Noncoding RNA Res DOI: 10.1016/j.ncrna.2020.11.005 sha: 77c52f3719930183f6bc0ba4e3dccb1ce49406d4 doc_id: 723787 cord_uid: hktwk386 BACKGROUND: Coronavirus disease 2019 (COVID-19) caused by a novel betacoronavirus named severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has attracted top health concerns worldwide within a few months after its appearance. Since viruses are highly dependent on the host small RNAs (microRNAs) for their replication and propagation, in this study, top miRNAs targeting SARS-CoV-2 genome and top miRNAs targeting differentially expressed genes (DEGs) in lungs of patients infected with SARS-CoV-2, were predicted. METHODS: All human mature miRNA sequences were acquired from miRBase database. MiRanda tool was used to predict the potential human miRNA binding sites on the SARS-CoV-2 genome. EdgeR identified differentially expressed genes (DEGs) in response to SARS-CoV-2 infection from GEO147507 data. Gene Set Enrichment Analysis (GSEA) and DEGs annotation analysis were performed using ToppGene and Metascape tools. RESULTS: 160 miRNAs with a perfect matching in the seed region were identified. Among them, there was 15 miRNAs with more than three binding sites and 12 miRNAs with a free energy binding of −29 kCal/Mol. MiR-29 family had the most binding sites (11 sites) on the SARS-CoV-2 genome. MiR-21 occupied four binding sites and was among the top miRNAs that targeted up-regulated DEGs. In addition to miR-21, miR-16, let-7b, let-7e, and miR-146a were the top miRNAs targeting DEGs. CONCLUSION: Collectively, more experimental studies especially miRNA-based studies are needed to explore detailed molecular mechanisms of SARS-CoV-2 infection. Moreover, the role of DEGs including STAT1, CCND1, CXCL-10, and MAPKAPK2 in SARS-CoV-2 should be investigated to identify the similarities and differences between SARS-CoV-2 and other respiratory viruses. The COVID-19 disease, resulting from a novel coronavirus, is currently a global threat leading to considerable disease and mortality worldwide. Since November 1, 2020, a total of 46,493,580 confirmed cases, as well as 1,203,902 deaths from COVID-19 in 215 countries and territories have been reported [1] . The Severe Acute Respiratory Syndrome Coronavirus-2 (SARS-CoV-2) is a close relative of SARS-CoV with 45-90% sequence similarity, which has resulted in the severe acute respiratory syndrome in over 8000 confirmed cases and about 800 deaths in 2003 [2, 3] . Coronaviruses are a diverse family of viruses associated with multiple respiratory diseases with different severity, like common cold, pneumonia, and now COVID-19 [4] .With a singlestranded positive-sense RNA genome with genome sizes of 26-32 kilobases (kb) in length, they have the largest known genomes among all known RNA viruses [5] . The virus genome possesses a 5' cap structure together with a 3' poly(A) tail, like an mRNA to translate its proteins. About two-thirds of the genome at 5' end is occupied by the replicase gene, which encoded two polyproteins, ORF1a and ORF1b. These polyproteins are further processed to generate the non-structural proteins (nsp). ORF1a is contributed to produce the nsp1-nsp11, while the rest of nsps (nsp12-nsp16) are originated from ORF1b [6] . Additionally, the viral structural proteins comprise surface (S), envelope (E), membrane (M), and nucleocapsid (N) proteins encoded by the one-third of genome at 3' end [6, 7] . A group of small non-coding RNAs, almost 19-24 base pairs in length, named microRNAs (miRNAs) plays a key role in the modulation of a wide range of biological processes, including J o u r n a l P r e -p r o o f development, immune system response, and cell death through gene expression regulation [8] . In addition, various aspects of the viral replication and proliferation, including host antiviral responses and viral pathogenesis can be influenced by miRNAs. MiRNAs mediate their regulatory function through direct binding to the target transcript. Perfect pairing in the seed region (position 2 to 8 from 5' end) has an important impact on the regulatory function of a miRNA. MiRNAs play a negative or positive role in virus-related processes in three ways: direct binding to the viral genome, binding to the viral transcripts, or binding to the host transcripts [9] .Host miRNAs may promote viral RNA stability, replication, and infection or conversely, reinforce host antiviral responses against viruses. However, the position, number, and distance between binding sites and point mutations in the seed region of a miRNA, can alter its target specificity and its subsequent impact [9] [10] [11] . It has been reported that in the samples infected with H5N1 influenza, miR-485 directly targets the viral PB1 gene coding an RNA-dependent RNA polymerase that is essential for virus replication [12] . Moreover, the induction of host immunity pathway like the interferon pathway upon viral infection can result in the enhanced expression of certain miRNA, including miR-155 to regulate the corresponding pathway [13] . For MERS-CoV genome, a total of 13 host miRNAs affecting the virus genome has been recognized, hence, their application as the appropriate therapeutics against viral infection appear promising as microRNAs are very specific in selecting the target regions [14] . Considering the current COVID-19 pandemic, it would be of great importance to investigate miRNAs involved in the host-SARS-CoV-2 interface. In this study, top miRNAs miRanda tool (version 3.3a) was used to predict the potential human miRNA binding sites on the SARS-CoV-2 genome sequence [15] . For this purpose, the thermodynamic folding energy and alignment score threshold values of -20 and 150 kcal/mol were set for miRanda tool; the strict alignment in the seed region was also considered with including the strict parameter. The gene count data derived from RNA sequencing in the lung tissue of COVID-19 patients p-values [17] . Finally, Cytoscape software version 3.7.2 was used to visualize miRNA-mRNA network and important GO term. Among the 2654 human mature miRNAs, 444 miRNAs were identified with direct binding site on different positions along with the coronavirus 2 reference genome (Table S1 ). It was focused on the interactions with perfect matching in the seed region and 160 miRNAs were sorted out. Among them, there was 15 miRNAs with more than three binging sites (Tables 1 and 2 ) and 12 miRNAs bound to the coronavirus 2 reference genome with a free energy (ΔG) less than -29 kCal/Mol (Tables 2 and 3 ). According to the results, miR-29 family (miR-29a, miR-29b, and miR-29c) had the most binding sites (11 sites) and miR-3175 had the least ΔG (-35 kCal/Mol). The position of binding sites on the SARS-CoV-2 for miRNAs with more than three binding sites or ΔG less than -29 kCal/Mol was also explored and ORF1ab, nucleocapsid, spike, ORF3a, membrane, and ORF7a coding regions with high capability for binding to host human miRNAs were found (Fig. 1) . ORF1ab, nucleocapsid, and spike sequences had the most binding sites. Among the miRNAs, miR-29 exhibited various binding sites on ORF1ab, nucleocapsid, and spike sequences. MiR-21 had binding sites on ORF1ab, spike, and ORF3a. The spike region, which encodes the spike protein, is necessary for viral entry and is a promising target for antiviral therapy. Eight binding sites for miR-29a-3p, miR-29c-3p, miR-21-3p, miR-761, miR-3130-3p (2 sites), miR-3167, and miR-3175 were recognized on the spike coding region (Fig. 1) . In particular, the binding pattern of miRNAs among genome sequences released from 10 different geographical locations was explored and no mutation and 100% similarity were found. RNA 3p, miR-186-5p, miR-93-5p, and miR-20a-5p were predicted to target only down-regulated DEGs (Fig. 3B) , and miR-155-5p, miR-146a-5p, miR-24-3p, and miR-21-5p were predicted to target only up-regulated DEGs (Fig. 3B ). MiR-16-5p, miR-484, let-7b-5p, miR-17-5p, miR-106b-5p, let-7e-5p, and miR-320a targets were from both up-and down-regulated DEGs. Analysis of DEGs targeted by miRNAs demonstrated that GO terms and biological pathways related to response to virus, influenza A, antiviral INF-stimulated genes, and positive regulator of NF-κB signaling were significantly enriched (Fig. 3E) . However, the pathways and GO terms related to eukaryotic translation initiation and signaling by interleukins were significantly depleted (Fig. 3F ). miRNA-mRNA network for up-and down-regulated DEGs in response to Figures 4A and 4B , respectively. SARS-CoV-2-induced DEGs related to viral processes were also sorted out and top miRNAs, which target them were explored ( Fig. 3C and D) . According to the results, it was identified that 38 up-regulated (11%) and 77 (17%) down-regulated DEGs were enriched in viral processes. MiR-146a-5p, miR-203a-3p, and miR-24-3p, which were predicted to target 26%, 10%, and 10% of the up-regulated DEGs, were respectively involved in viral processes (Fig. 3C) . Otherwise, 32% and 31% of the J o u r n a l P r e -p r o o f down-regulated DEGs involved in viral processes, were targeted by miR-615-3p and miR-16, respectively (Fig. 3D) . miRNA-mRNA network for miRNAs that target SARS-CoV-2-induced DEGs involved in viral processes are depicted in Figure 5 . Viral proteins have been broadly considered as targets for antiviral therapies, but the problem arises when the selective pressure results in the emergence of a new antiviral drug resistance lineage. Therefore, host-coded factors and particularly, microRNAs seem to be a better strategy [18] . miRNAs as antiviral targets is so important that the first clinical trial of miRNAs relates to their involvement in viral processes [19] . MiR-122, which is highly expressed in liver, binds (Table S1 ) reported in the present study with one binding site on the SARS-CoV-2 genome. However, in this study, it was focused on miRNAs with more than three binding sites on the SARS-COV-2 reference genome. Top miRNAs, which target host-related DEGs involved in viral processes in response to SARS-CoV-2 infection, were also predicted. In the present study, the number of binding sites, ΔG > -20 Kcal, and perfect complementarity in the seed region were considered. Among covid-19-binded miRNAs, miR-29 family had the greatest number of interactions (11 sites). miR-29 family consists of three members, namely, miR-29a, miR-29b, and miR-29c. In previous studies, the impact of host miR-29s on the regulation of viral processes depended on whether they directly bind to the viral genome or to the host transcripts [27] [28] [29] . Direct binding of miR-29a to the 3' UTR region of the HIV genome, increased the transport of virus to p-bodies and reduction of HIV replication. Ahluwalia et al. (2008) also reported that the inhibitory impact of miR-29a on HIV infection is mediated through binding to the accessory viral protein negative factor (Nef), which is critical for viral persistence and release [30] . Therefore, miR-29a has been considered as a potential therapeutic target for HIV eradication [29] . According to the results of the present study, five miR-29s binding sites were predicted in the spike and nucleocapsid coding regions of SARS-CoV-2. Spike proteins protruded from the viral envelope are responsible and critical for host-receptor binding and viral entry. Nucleocapsid proteins specifically bind to the viral genome and facilitate viral entry, replication, and release. Both spike and nucleocapsid proteins were considered as targets for SARS-CoV-2 antiviral drug development. miR-29s also targeted sequences in the ORF1ab region, which is the largest part of the genome and encoded for 16 nsps [31] . Despite having a large number of direct binding sites on the SARS-CoV-2, no miR-29s was found among the top miRNAs targeting host DEGs. However, in previous studies, direct binding of miR-29s to the host A20/TNFAIP3 transcript in response to influenza A and JEV infections, and subsequently, modulation of antiviral and proinflammatory responses have been reported [27] . Considering the high levels of miR-29s in the lungs of healthy adults and better response of these people to SARS CoV-2 compared to those with respiratory diseases with low levels of miR-29s, the probable role of miR-29s in modulating SARS-CoV-2 infection was suggested. MiR-21, another SARs-CoV-2 binding microRNA, had four binding sites on the SARS-CoV-2 genome. miR-21 is one of the best known miRNAs whose expression increases in many pathological conditions including asthma, pulmonary fibrosis, and viral infection [32, 33] . There is no report about direct binding of miR-21 on human viral genomes, and current reports about the involvement of miR-21 in viral infections are limited to modulating host transcripts. For example, the positive role of miR-21 in influenza A replication has been attributed to miR-21-host HDAC8 interaction [34] . In addition, it has been shown that miR-21 reduced the antiviral NF-KB pathway through binding to IRAK1 and TRAF6 transcripts in HIV and HCV infections [35, 36] . According to the results of the present study, miR-21 had two binding sites on the spike coding regions. In addition, miR-21 was one of the top miRNAs which targeted upregulated host-DEGs in response to SARS-CoV-2. One of the miR-21 targets is CXCL-10 that is a biomarker for viral, bacterial, fungal, and parasitic contamination [37] . In the present study, suppressed cell cycle and prevented EV71 replication through decreasing CCND1 level, which is an important protein in G1 to S phase transition during cell cycle processes [39] . In the present study, decreased level of CCND1 in the lungs of patients infected with SARS-CoV-2 was observed, which seems to be not as effective as its reduction in EV1 infection, because the studied patients died from COVID-19. Let-7e and let-7b, two let-7 family members, were also among the top miRNAs targeting host DEGs in SARS-CoV-19. The association between Let-7 family and several viral infections including RSV, influenza A, and hMPV has been demonstrated [40, 41] . Similar to miR-16, Let-7 family also targets CCND1 [42] . In contrast to EV71 infection, the decreased level of replication has been previously reported [43] . In order to retaliate, SARS-CoV encodes a STAT1 antagonist (ORF6) to escape from eradication [44] . Although in this study, increased levels of STAT1 were observed in the lungs of patients infected with SARS-CoV-2 but it did not lead to an effective antiviral response. It may be, at least in part, due to an antagonizing strategy by SARS-CoV-2 similar to SARS-CoV. In addition to STAT1, mitogen-activated protein kinase-activated protein kinase 2 J o u r n a l P r e -p r o o f J o u r n a l P r e -p r o o f A major outbreak of severe acute respiratory syndrome in Hong Kong The proximal origin of SARS-CoV-2 Human coronaviruses: a review of virus-host interactions Genotype and phenotype of COVID-19: Their roles in pathogenesis Genomic characterization of a novel SARS-CoV-2 Coronavirus nonstructural protein 1: Common and distinct functions in the regulation of host and viral gene expression Overview of microRNA biogenesis, mechanisms of actions, and circulation MicroRNA Regulation of RNA Virus Replication and Pathogenesis Viruses, microRNAs, and host interactions Distance constraints between microRNA target sites dictate efficacy and cooperativity The microRNA miR-485 targets host and influenza virus transcripts to regulate antiviral immunity and restrict viral replication microRNAs in viral acute respiratory infections: immune regulation, biomarkers, therapy, and vaccines The impact of MicroRNAs (miRNAs) on the genotype of coronaviruses ToppGene Suite for gene list enrichment analysis and candidate gene prioritization Metascape provides a biologist-oriented resource for the analysis of systems-level datasets Broad-Spectrum Inhibition of Respiratory Virus Infection by MicroRNA Mimics Targeting p38 MAPK Signaling Safety, tolerability, and antiviral J o u r n a l P r e -p r o o f effect of RG-101 in patients with chronic hepatitis C: a phase 1B, double-blind Modulation of hepatitis C virus RNA abundance by a liver-specific MicroRNA The diverse roles of microRNAs at the Host-Virus Interface Computational analysis of microRNA-mediated interactions in SARS-CoV-2 infection What is the potential function of microRNAs as biomarkers and therapeutic targets in COVID-19? How miRNAs can protect humans from coronaviruses COVID-19 The Prediction of miRNAs in SARS-CoV-2 Genomes: hsa-miR Databases Identify 7 Key miRs Linked to Host Responses and Virus Pathogenicity-Related KEGG Pathways Significant for Comorbidities Implications of SARS-CoV-2 Mutations for Genomic RNA Structure and Host microRNA Targeting Induction of the cellular miR-29c by influenza virus inhibits the innate immune response through protection of A20 mRNA Micro RNA-29b modulates Japanese encephalitis virus-induced microglia activation by targeting tumor necrosis factor alphainduced protein 3 Cellular microRNA and P bodies modulate host-HIV-1 interactions Human cellular microRNA hsa-miR-29a interferes with viral nef protein expression and HIV-1 replication The Proteins of Severe Acute Respiratory Syndrome Coronavirus-2 (SARS CoV-2 or n-COV19), the Cause of COVID-19 MicroRNA-21 is up-regulated in allergic airway inflammation and regulates IL-12p35 expression mediates fibrogenic activation of pulmonary fibroblasts and lung fibrosis miR-21-3p regulates influenza A virus replication by targeting histone deacetylase-8 HCV-induced miR-21 contributes to evasion of host immune system by targeting MyD88 and IRAK1 MicroRNA profile changes in human immunodeficiency virus type 1 (HIV-1) seropositive individuals CXCL10/IP-10 in infectious diseases pathogenesis and potential therapeutic implications restrain memory T cell differentiation, cell cycle, and survival MiR-16-5p mediates a positive feedback loop in EV71-induced apoptosis and suppresses virus replication Non-Coding RNAs and Their Role in Respiratory Syncytial Virus (RSV) and Human Metapneumovirus (hMPV) Infections Cellular micro RNA let-7c inhibits M1 protein expression of the H1N1 influenza A virus in infected human lung epithelial cells Respiratory syncytial virus modifies microRNAs regulating host genes that affect virus replication The role of miR-146a in viral infection The authors would like to gratitude Kerman University of Medical Sciences, Kerman, Iran, for its financial support (Grant No: 99000343). This Study was approved by the Ethics Committee of Kerman University of Medical Sciences (IR.KMU.REC.1399.435). We are also grateful to Dorsay Hasani for her kind advice in reviewing the English text. The authors declare that they have no conflict of interest.