key: cord-1052336-5v5zjbnj authors: Amini-Farsani, Zeinab; Yadollahi Farsani, Mahtab; Arab, Samaneh; Forouzanfar, Fatemeh; Yadollahi, Mitra; Asgharzade, Samira title: Prediction and analysis of microRNAs involved in COVID-19 inflammatory processes associated with the NF-kB and JAK/STAT signaling pathways date: 2021-08-18 journal: Int Immunopharmacol DOI: 10.1016/j.intimp.2021.108071 sha: 65c446a87ca7d124dcdd6c6c17e33967f8682243 doc_id: 1052336 cord_uid: 5v5zjbnj COVID-19 is the cause of a pandemic associated with substantial morbidity and mortality. As yet, there is no available approved drug to eradicate the virus. In this review article, we present an alternative study area that may contribute to the development of therapeutic targets for COVID-19. Growing evidence is revealing further pathophysiological mechanisms of COVID-19 related to the disregulation of inflammation pathways that seem to play a critical role toward COVID-19 complications. The NF-kB and JAK/STAT signaling pathways are highly activated in acute inflammation, and the excessive activity of these pathways in COVID-19 patients likely exacerbates the inflammatory responses of the host. A group of non-coding RNAs (miRNAs) manage certain features of the inflammatory process. In this study, we discuss recent advances in our understanding of miRNAs and their connection to inflammatory responses. Additionally, we consider the link between perturbations in miRNA levels and the onset of COVID-19 disease. Furthermore, previous studies published in the online databases, namely web of science, MEDLINE (PubMed), and Scopus, were reviewed for the potential role of miRNAs in the inflammatory manifestations of COVID-19. Moreover, we disclosed the interactions of inflammatory genes using STRING DB and designed interactions between miRNAs and target genes using Cityscape software. Several miRNAs, particularly miR-9, miR-98, miR-223, and miR-214, play crucial roles in the regulation of NF-kB and JAK-STAT signaling pathways as inflammatory regulators. Therefore, this group of miRNAs that mitigate inflammatory pathways can be further regarded as potential targets for far-reaching-therapeutic strategies in COVID-19 diseases. The coronavirus disease 2019 is the cause of morbidity and mortality, which is associated with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection that is characterized by acute respiratory failure. Usually, COVID-19 is manifested as a mild respiratory disease, although it can occasionally lead to severe alveolar inflammation. Lung histopathological findings of patients with COVID-19 showed extensive alveolar damaging via widespread infiltration of immune cells, thrombosis of pulmonary vessels and fibrin deposits (Asgharzad et al., 2021; . In patients with COVID-19, an excessive or uncontrolled innate immune response leads to a cytokine storm and acute respiratory distress syndrome (ARDS). Infected macrophages with COVID-19 present COVID-19 antigens to T cells. Activated T cells produce cytokines that are associated with different T cell subtypes, and following the release of large quantities of cytokines, there is immune response amplification. In response to COVID-19 infection, the secretion of massive chemokines and cytokines like Interleukin-1 (IL-1), Interleukin-6 (IL-6), Interleukin-8 (IL-8), Interleukin-21 (IL-21), tumor necrosis factor-beta (TNF-β), and monocyte chemoattractant protein-1 (MCP-1) is promoted in infected cells. Afterward, these chemokines and cytokines, in turn, recruit lymphocytes and leukocytes to the infection site. CD8 T cells produce very effective mediators to destroy COVID-19, but the continued release of these mediators led to viral persistence and a negative effect on CD8 T cell activation (A. J. Chauhan et al., 2020) . S protein of COVID-19 attaches to dipeptidyl peptidase-4 receptor (DPP4R) on the host cell and leads to the appearance of genomic RNA (dsRNA) in the cytoplasm of the host cell. Toll-like receptor 3 (TLR-3) sensitized by dsRNA and signaling cascades of interferon-regulatory factors (IRFs) and nuclear factor-kB (NF-kB) are activated and then produced interferons (IFNs) (IFN-I, IFN-II, and IFN-III) and pro-inflammatory cytokines. The engagement of IFN (I and III) receptors induced different members of the Janus kinase (JAK) and signal transducer and activator of transcription (STAT) families and also helped form specific transcription factor complexes. In contrast, IFN-II activated JAK1 and JAK2, producing a phosphorylated STAT1 homodimer (Widiasta et al., 2020) . Small molecules (17-24nt) of non-coding RNAs known as miRNAs regulate the posttranscriptional expression thought to block the target genes by binding to the 3′UTR during transcription, repressing the messenger RNA, and promoting their degradation by cleavage in various cellular processes, such as cell proliferation, apoptosis, and differentiation. More than two thousand miRNAs have been identified in humans. One miRNA targets several genes, while one gene is targeted by multiple miRNAs. Some miRNAs are produced during viral infection, modulating the function of the immune response against respiratory viruses (Abu-Izneid et al., 2020; Widiasta et al., 2020) . This review will analyze the role of miRNAs in the regulation of immune response against COVID-19 especially inflammatory pathway based on bioinformatics prediction. The renin-angiotensin-system (RAS) is a cascade, which results in the conversion of the inactive decapeptide angiotensin I (Ang I) into the active peptide angiotensin II (Ang II). The initial enzyme in the cascade is renin, an aspartic proteinase, which cleaves the amino terminus of the angiotensinogen, thereby releasing the decapeptide pro-hormone Ang I. Angiotensin-converting enzyme (ACE) removes two additional amino acids to yield the active octapeptide hormone Ang II (Zisman, 2005) . Ang II exerts its actions via the (JAK-STAT) signaling pathway, which leads to vasoconstriction, hypertension, and chronic tissue injury (Adrian Recinos III et al., 2007; Seif et al., 2017) . Renin-Angiotensin system mediates pro-inflammatory cytokine production. Indeed, Ang II increases the infiltration of immune cells, which, in turn, augments the local production of pro-inflammatory cytokines such as interferon (IFN)-γ, IL-1, IL-6, and tumor necrosis factor alpha (TNF-α) in target tissues (Seif et al., 2017) . Activation of the Ang II signaling by JAK STAT pathway can occur in the mesangial cells, hepatocytes, cardiovascular system, renal proximal tubular cells, and brainstem astrocytes (Seif et al., 2020) . In the pathogenesis of ARDS, Ang II increases through ACE and drives severe lung failure through the Angiotensin II type I receptor (AT1R) (Gu et al., 2016) . AT1R recruits and phosphorylates JAK2, and, based on the target cell, one of the STAT is phosphorylated by activated JAK2 (F. et al., 2020) . In COVID-19, macrophage infiltration into the lung tissue occurs, which is accompanied by the apoptosis of epithelial cells and pneumocytes. Infiltration of macrophages into alveolar cavities may be mediated by monocyte chemoattractant protein-1 (MCP-1) through transforming growth factor beta 1 (TGF-β1) and TNF-α, which leads to cytokine production (He et al., 2008) . Markers of infiltration include the neutrophil chemokine receptor 2 (CXCR2), along with monocyte chemotactic protein (MCP-1/CCL2) and its receptor chemokine receptor type 2 (CCR2). NF-kB signaling pathway genes (NFKBIA, NFKB1, RELA, NFKB2) were upregulated in COVID-19 patients (Hadjadj et al., 2020; Sohn et al., 2020) . Cytokine storm is emerging as one of the mechanisms leading to ARDS, defined as the release of high concentrations of pro-inflammatory cytokines, including IL-1, IL-2, IL-6, IL-7, IL-10, TNF-α, granulocytecolony stimulating factor, interferon (IFN)-γ inducible protein-10 (IP-10), MCP-1, and macrophage inflammatory protein 1-α (MIP1-α) by immune cells. The cytokine storm initiates a fulminant response by the immune system, leading to multisystemic organ dysfunction and even death in severe COVID-19 (Huang et al., 2020) . Elevated concentrations of inflammatory markers including IL-6, c-reactive protein (CRP), procalcitonin (PCT), and erythrocyte sedimentation rate (ESR) are detected in fatal cases (Zeng et al., 2020) . Cytokine storm on the IL-6/TNF-α axis appears to be mediated likely through the phosphorylation of the NF-kB subunit p65. In SARS-CoV infection, the viral spike protein induces the activation of NF-kB via the nuclear factor of kappa light polypeptide gene enhancer in B-cells inhibitor, alpha (IkB-α) degradation, resulting in the production of IL-6 and TNF-α . The viral nucleocapsid protein of SARS-CoV can also bind to the NF-kB regulatory element on the IL-6 promoter, with the highest activity in the presence of the p65 subunit (Zhang et al., 2007) . Regulatory elements in the ACE2 gene control the transcription of PIR (pirin), a negative regulator of NF-kB subunit RELA (p65). SARS-CoV-2 disruption of ACE2 has been proposed to decrease PIR expression (Fadason et al., 2020) . PIR has been proposed to act as a reversible switch that provides the NF-kB response with changes in the level of redox (oxidative stress) in the cell nucleus (Liu et al., 2013) . Repression of PIR ablates the inhibition of IL-6 expression . Studies in addressing bronchoalveolar lavage fluid (BALF) show promise since findings in the blood do not essentially describe the events occurring in tissues directly affected by the infection (García, 2020) . In a study conducted by Xiong et al (Xiong et al., 2020) , a comparison of the cytokine genes demonstrated that in BALFs of three COVID-19 patients, the genes for IL-10, CCL2/MCP-1 (together with its CCR2 receptor), macrophage inflammatory protein-1α (CCL3/MIP-1A) together with its CCR5 receptor, interferon gamma-induced protein 10 (CXCL10/IP-10), and macrophage inflammatory protein-1 beta (CCL4/MIP1β) increased. Interestingly, in PBMC, genes related to apoptosis, autophagy, and p53 pathways increased; a finding that could be related to the lymphopenia found in the COVID-19 patients. Moreover, IL-6 transcripts were not enhanced in PBMCs, although the patients had high plasma contents of such cytokine, suggesting that circulating IL-6 could have been produced in the lungs, either by alveolar epithelial cells or by recruited inflammatory cells. One study showed that in BALFs of patients with severe COVID-19, macrophages were predominant, with a minor proportion of T and natural killer (NK) cells, compared with moderate COVID-19 patients. Fatty acid binding protein 4 (FABP4) produced by alveolar macrophages was preferentially expressed in healthy controls and in patients who had a moderate COVID-19 infection, while FCN1 (monocyte-derived) and SPP1 (pro-fibrotic) were expressed in patients with severe COVID-19. Besides, these results suggest that during COVID-19 infection, inflammatory monocytes (FCN1 + ) are recruited from the circulation into the lungs, where they differentiate into SPP1 + macrophages, constituents of the severe inflammatory reaction. Analysis of the BALF transcriptome displayed that T and NK cells increased in COVID-19 patients in comparison with healthy controls, which according to their gene expression can be classified in NK, cluster of differentiation 8 (CD8), (cluster of differentiation 4 (CD4), Tregs, and proliferating cells. Interestingly, genes related to activating molecules, migration, calcium signaling, and effector molecules were highly expressed by CD8 cells in patients who had a moderate COVID-19 infection compared with those with severe COVID-19 infection; this further supports the role of CD8 cells in the elimination of the virus and their subsequent protective immunity. In contrast, higher expression of genes related to proliferation, energy generation, and initiation of translation was seen in patients with severe disease, suggesting that in patients with moderate COVID-19 infection, CD8 cells are more differentiated and efficient, while in severe infection, T cells are in a proliferative stage. Furthermore, the finding that the TCR repertoire is higher in CD8 than CD4 cells suggests a larger clonal expansion of the CD8 cells taking part in the resolution of the infection (Liao et al., 2020) . The results of Zhou et al. demonstrated that in the BALF of patients with COVID-19, the differentially expressed genes (DEGs) included more proinflammatory chemokines, such as CXCL2, CXCL8, and, CXCL17, as well as the CXCR2 receptor, essential to neutrophil recruitment, and CCL2 and CCL7, needed for monocyte recruitment. Besides, in patients with COVID-19, IL-1b, antiviral interferon stimulated genes (ISGs), and genes related to the TNF, IL-17, and NF-kB signaling pathways increased. Furthermore, the cellular analysis demonstrated an enhanced neutrophil to lymphocyte ratio (NLR) in COVID-19 patients compared with other pneumonias patients. The miRNA is referred to a short RNA originated from a precursor with a stem-loop structure that interferes mainly with the regulation of gene expression by targeting mRNAs and dissecting or inhibiting their expression (Watanabe et al., 2016) (Fig. 1 ). Seven nucleotides situated at the 5'-end of mature miRNA molecules comprise what is known as the seed region, which binds to the non-coding end [3'-untranslated Injinari et al., 2021; Lim et al., 2003) . MiRNAs comprise about 1-5% of the animal genomes, and it is estimated that there are approximately 800-1000 miRNAs in the human genome and that over one-third of the protein-coding genome is regulated by miRNAs. Each miRNA can target and regulate the mRNA transcription of hundreds of downstream genes. A miRNA can have several target sites in the mRNA of a downstream gene, while a mRNA can be targeted by multiple miRNAs. Therefore, miRNAs contribute to a recently known level of gene expression regulation and play an important role in biological processes, function of almost all tissues and organ systems in the body, and development of various diseases (Watanabe et al., 2016) . Coronaviruses are a large family of viruses that are known to cause illness ranging from the common cold to SARS (Al-Hazmi, 2016). MiR-155 augmented antiviral T cell responses including cytokine production, cytotoxic activity, and homing to the central nervous system (CNS) in coronavirus-induced neurologic disease model (Dickey et al., 2016) . The human coronaviruse OC43 nucleocapsid protein potentiates NF-kB pathway activation. It has been demonstrated that miR-9 is capable of diminishing NF-kB translation by targeting its 3' UTR region. SARS, a relatively rare disease, is caused by severe acute respiratory syndrome coronavirus (SARS-CoV or SARS-CoV-1). This is an acute infectious disease with a significant mortality rate and common clinical features such as pulmonary fibrosis and pulmonary insufficiency (Stark & Atreya, 2005) . Induction of miR-574 and miR-214 expression was reported in bronchoalveolar stem cells infected by SARS-CoV (Qian et al., 2008) . The expression of miR-223 and miR-98 was downregulated in BASCs through some viral nucleocapsid proteins controling pro-inflammatory cytokine secretion (Mallick et al., 2009) . A recent study showed that the response to tocilizumab which was tested on COVID-19-induced cytokine storm is associated with a significant reduction of plasma IL-6 and a significant elevation of miR-146a levels. However, COVID-19 patients who did not respond to this antibody had lower serum levels of miR-146a-5p after the treatment (Sabbatinelli et al., 2021) . The other established effect of miRNAs on coronavirus is the regulation of ACE2 expression. This enzyme catalyzes the conversion of Ang I into Ang 1-9 and II into 1-7. Several studies have confirmed the potential role of ACE2 in COVID-19-related diseases (Abedi et al., 2020; Hoffmann et al., 2020; . MiR-125b acts as a negative regulator targeting 3'UTR of ACE2 mRNA. Similarly, miR-18 was disregulated in ACE-associated nephropathy, which offers antimiR-18 as a novel therapeutic candidate in COVID-19 related diseases (Widiasta et al., 2020) . Computational prediction analysis detected that 29 miRNA precursors in the SARS-CoV-2 genome can target host genes involved in vital signaling pathways such as Wnt and epidermal growth factor (EGF)-receptor signaling pathways. On the other hand, some SARS-Cov-2-encoded genes could be targeted by host miRNAs. The products of these genes are essential in the entrance and replication of the virus. For example, has-miR-203b-3p and has-miR-148-3p targeted ORF genes that play key roles in viral replication. Liu et al. recruited bioinformatics approaches and showed that miR-147-3p derived virus through serine 2 as a transmembrane protease, facilitating entrance into gastrointestinal cells. STAT1 is an essential element in IFN signaling and anti-virus defense. Panther analysis reported that some miRNAs derived from SARS-CoV-2 might suppress STAT1 protein (Saçar Demirci & Adan, 2020). Analyszing the transcriptomic data of human alveolar and bronchial epithelial cells demonstrated that the CSF1 gene, a known target of miR-1207-5p, is over-expressed during SARS-CoV-2 infection. CSF1 promotes macrophage migration and activation and may be associated with the acute inflammatory response in severe COVID-19 (Bertolazzi et al., 2020) . Yousefi et al. employed Viruses-STRING, a protein-protein interaction database, and illustrated that the signaling pathway through TGF-beta with the largest numbers of involved genes plays a crucial role during SARS-CoV-2 infection (Yousefi et al., 2020) . infection (A. Kumar et al., 2020; Paul Zimmet et al., 2020) . It was indicated that the upregulation of the cardiac miR-133a levels in the diabetic heart diminished cardiac lipid accumulation. Thus, miR-133a is a promising candidate for investigating its role in heart failure in patients with diabetes and COVID-19 (Kim, 2013) . Upregulation of miRNA 197-5p was described in cardiovascular patients, and miRNA 197-5p was proposed as a prognostic and risk factor of such cardiac events. MiRNA 197-5p also has a role in defense against SARS-CoV-2 infection. Therefore, a higher COVID-19 mortality rate and susceptibility in patients with the cardiovascular disease may be connected to the loss of the miRNA 197-5p binding site across the SARS-CoV-2 genome (Ali & McLellan, 2020) . On the other hand, Balme et al.'s prediction study found that an increased expression of hsa-miR-1307-3p might lead to a reduction in SARS-CoV-2 replication and had a high affinity to SARS-CoV-2 genome 3′UTR and high expression in lung tissue. The hsa-miR-1307-3p could promote glycogenesis and glucose uptake by inhibiting protein kinase c (PKC) and extracellular signal-regulated kinase (ERK), which are inhibitors of RS1 and indirectly reduce blood sugar (Balmeh et al., 2020) . In the field of miRNA-based antiviral therapies, Hum et al. introduced three microRNAs (miR-16, miR-200, and miR-24) that can serve as modulating approaches in SARS-CoV-2 infection (Hum et al., 2021) . It was demonstrated that miR-24 had a crucial role in furin regulation. It is a serine protease that activates proteolytic function and the host-cell entrance of some respiratory viruses and also increases TGF-β1secration that leads to lung damage (Mille & Whittaker, 2014) . MiR-200 binds to a highly conserved region of the ACE2 3′-UTR gene as a major regulator of heart function (Hum et al., 2021) , and miR-16 induces apoptosis by downregulating BCL2 (Cimmino et al., 2005) . Suppression of miR-16 was also found to increase mitochondrial reactive oxygen species (ROS) generation and toll-like receptor 4 (TLR4) expression (Moon et al., 2014) . All in all, it seems that the identification of host and virus miRNAs may bring about perspective therapeutic strategies for the control of viral pathogenicity and its side effects in COVID-19 related diseases (El-Nabi et al., 2020) . Potential targets of a number of most important miRNAs (miR-9, miR-98, miR-223, and miR-214), whose expression is predicted to change in host cells infected with COVID-19 (Soheilifar et al., 2020) , were specified using bioinformatics analysis and shown in Table 1 The 3'untranslated region (3' UTR) of TLR7 is directly targeted by miR-9-5p and miR-223-5p. TLR8 is also directly targeted by miR-98-5p and miR-223-3p. Interestingly, the activation of TLR2 and TLR7/8 in the MyD88-dependent pathway increased the expression of miR-9 in polymorphonuclear neutrophils (PMN) and monocyte cells (Bazzoni et al., 2009 ). Various cytokines including interleukins IL-10, IL-7, IL-6, IL-4, and IL-2 and interferons IFN-α, IFN-β, IFN-γ, and granulocyte colony-stimulating factor (GCS-F) induce different biologic effects through activating the JAK/STAT pathway (Catanzaro et al., 2020) . IL-6 that severely increases in COVID-19 patients and plays a significant role in acute inflammatory response is a major potential target of miR-98-5p, miR-9-5p, and miR-223-3p. Moreover, it is predicted that the expression of IL6R, the membrane receptor of IL-6, is also partially regulated by miR-98-5p. Via activating various cells expressing glycoprotein 130 (GP130) and IL-6 receptors, IL-6 activated JAK/STAT pathway (Chi et al., 2020) . The IL-6/JAK/STAT pathway is highly activated in acute inflammation, and the excessive activity of this pathway in COVID-19 patients likely exacerbates the inflammatory responses of the host. Interestingly, the activation of this pathway in a positive feedback loop produced IL-6 (Catanzaro et al., 2020) . Investigating samples of osteoarthritis (OA) patients indicated that miR-9-5p elevated the expression of IL-6 in human chondrocytes through inhibitng monocyte chemotactic protein-1-induced protein-1 (MCPIP1) (Makki et al., 2015) . The results of a study on the peripheral blood mononuclear cells (PBMCs) samples of systemic lupus erythematosus (SLE) patients showed that miR-98-5p directly targeted IL-6 and inhibited the production of inflammatory cytokines and cell proliferation through IL-6/STAT3 pathway (Yuan et al., 2019) . Moreover, the results of a dual luciferase reporter assay in human adipose stem cells indicated that miR-223-3p regulated the expression of inflammatory cytokines IL-6 and STAT3 (J. Wu et al., 2019) . Furthermore, the results of another study showed that miR-223-3p might be able to increase the expression of IL-6 in synoviocytes through the downregulation of IL-17 D receptor (Moriya et al., 2017) . Note that miR-223-3p affects the secretion of IL-6 in mast cells via regulating insulin growth factor I receptor/phosphatidylinositol-3-kinase (IGF1R/PI3K) signaling pathway (Jianjun . The GP130 (IL6ST) receptor is also targeted by miR-223-5p, miR-223-3p, and miR-9-5p. The results of an in vitro study on mouse NPCs demonstrated that miR-9 directly targeted LIFR beta, IL6ST (GP130), and JAK1 genes, lowered their expression, and thus inhibited astrogliogenesis in these cells (Zhao et al., 2015) . A research on Kawasaki disease (KD) mouse found that the increased expression of miR-223-3p, which is an important regulatory factor in endothelial damage to the KD, lowered the expression of IL6ST ( Wang et al., 2019) . On the other hand, angiotensin II that is produced locally in the inflamed vessels leads to the production of IL-6 after attachment to angiotensin II receptor through activating JAK/STAT pathway. AT1R is directly targeted by miR-98-3p and miR-223-3p. IL-10, acting as a key mediator in immune response, and interleukin-4 receptor are also directly targeted by miR-98-5p. The results of a study on HEK-293 and RAW 264.7 cell lines demonstrated that miR-98-5p directly targeted the 3' UTR of IL-10 and inhibited its expression at the posttranscriptional level (Liu et al., 2011) . IFNA21, a member of type 1 IFN family, is directly targeted by miR-223-3p. An in vitro functional study in macrophages demonstrated that miR-223-3p increased the production of type 1 IFN via directly targeting FOXO3. Moreover, a type 1 IFN raised the expression of miR-223-3p in a positive feedback loop . Furthermore, miRNAs could also affect the expression of IFN receptors at the post-transcriptional level. IFNAR1 (interferon alpha receptor 1) is a direct target of miR-98-5p, miR-98-3p, miR-9-3p, and miR-214-3p. Interferon gamma receptor 1(IFNGR1) is also targeted by miR-9-3p and miR-98-5p. The expression of genes in the downstream signaling pathway of IFN receptors, JAK/STAT, is affected by many miRNAs (Kohanbash & Okada, 2012; Witte & Muljo, 2014) . JAK1, a member of the Janus kinase (JAK), a family of non-receptor tyrosine kinases, is directly targeted by miR-9-5p. Moreover, JAK3, as another member of this family, is targeted by miR-9-5p, miR-9-3p, and miR-214-5p. The findings of an in vitro functional study indicated that the 3' UTR region of JAK1 and MMP-13 genes was a direct target of miR-9 in human atherosclerosis models and miR-9 was able to inhibit downstream NLR family pyrin domain containing 3 (NLRP3) inflammasome through targeting JAK1 (Y. Wang et al., 2017) . STAT transcription factors, which are phosphorylated and activated after the activation of JAK proteins, are the regulatory targets of miRNAs. The expression of STAT1 is regulated by miR-9-3p and miR-223-3p, and the expression of STAT2 is regulated by miR-98-5p, miR-223-3p, and miR-214-3p. In addition, STAT3 and STAT5A are also directly targeted by miR-98-5p and miR-223-3p, respectively. A luciferase reporter assay in human T cells revealed that miR-150 and miR-223 specifically targeted the 3'UTR of STAT1 and consequently reduced the expression of this gene and inhibited the downstream signaling pathway independent from or dependent on IFN (Moles et al., 2015) . Microarray analysis results in the nucleus pulposus tissues of patients with intervertebral disc degeneration showed that the miR-98 expression significantly declined. Furthermore, the results of an in vitro gain-of-function study indicated that miR-98 was able to target STAT3 and inhibit IL-6/STAT3 signaling pathway in IDD patients (Ji et al., 2016) ). In addition, the knockdown of miR-98 in nasopharyngeal carcinoma cells reduced the expression of STAT3 protein and consequently inhibited the proliferation, migration, and invasion of these cells through miR-98/STAT3 signaling pathway (J. Liu et al., 2017) . In another study, it was shown that the induced underexpression of miR-223 in murine macrophages activated its direct target, STAT3, and thus significantly increased the production of pro-inflammatory cytokines IL-6 and IL-1 β (Q. Chen et al., 2012) . The increased expression of miR-223 in mouse embryonic fibroblasts and HEK293 cells lowered the expression of different genes in the cell survival or death pathway including STAT5A, and more comprehensive analyses verified STAT5A as a direct target of miR-223 in this study (Pinatel et al., 2014) . MiRNAs are able to indirectly affect signaling pathways activated by cytokines and thus innate and adaptive immune responses through targeting the JAK/STAT pathway regulators (Piganis et al., 2011) . PIAS3, an inhibitor of STAT3 transcription factor in cell nucleus, was identified as a direct target of miR-214 and miR-9-5p. Increasing the expression of miR-9-5p by reducing the expression of protein inhibitor of activated STAT 3 (PIAS3), an inhibitor of JAK/STAT pathway, increased the expression of STAT3, resulting in an increase in the population of Th17 cells in MS patients (Majd et al., 2018) . Furthermore, miR-9 and miR-181a led to the development of earlystage myeloid-derived suppressor cells (eMDSCs) in breast cancer through inhibiting the function of negative regulators of JAK/STAT pathway, SOCS3, and PIAS3 . Histone acetyltransferase EP300 that functions in chromatin remodeling and transcription regulation of the target genes of JAK/STAT pathway is also a direct target of miR-9-5p. The overexpression of miR-9 increased the expression of miR-132 and indirectly affected the expression of the direct target genes of miR-132, namely phosphatase and tensin homolog (PTEN), FOXO3A, and EP300, and apoptosis in rheumatoid arthritis . STAM, an adaptor molecule playing a role in facilitating the transcription of specific target genes of JAK/STAT signaling pathway (Rawlings et al., 2004) , and SLIM, a nuclear E3 ubiquitin ligase that is a negative regulator of STAT pathway (Tanaka et al., 2005) , are targets of miR-9-5p. Finally, JAK/STAT pathway activates the transcription of numerous genes in the inflammatory pathway, including ACE2, IL-8, IL-6, IL-2, IL-17, IL-1β, CSF3, CSF2, CCL3, TNF-α, and IFN-γ, which are regulated by different miRNAs (Catanzaro et al., 2020; Luo et al., 2020) . IL-7 was identified as a direct target of miR-9-5p and miR-9-3p, and also, CCL3 was identified as a direct target of miR-98-5p and miR-223-3p. The results of high-throughput sequencing demonstrated that the expression of miR-98-5p severely increased in immunoglobulin a nephropathy (IgAN) patients and this miRNA was able to directly target CCL3, and when its expression was inhibited, increase the expression of IL6 (D. Liu et al., 2020) . Through directly targeting CCL3 and IL-6, miR-223 controls the migration of inflammatory cells to the lung tissue and the formation of stable local inflammation in tuberculosis disease (Dorhoi et al., 2013) . NF-kB transcription factor plays an important role in innate and adaptive immunity. In normal conditions, NF-kB is inhibited by IKB proteins in cytoplasm. Various cellular stimuli including pathogens lead to the induction of NF-kB and the displacement of its nucleus through the phosphorylation and ubiquitin-dependent proteasomal degradation of the inhibitor of IKB. NF-kB leads to the transcription of a wide variety of pro-inflammatory cytokines, chemokines, antiapoptotic proteins, and stress-response proteins (Catanzaro et al., 2020) . NF-kB pathway in lung inflammatory immunopathology, which is induced after infection with respiratory viruses such as SARS-CoV, is very active (DeDiego et al., 2014; Smits et al., 2010) . Genes regulating NF-kB signaling pathway are the regulatory targets of different miRNAs. EDA, a cytokine activating NF-kB pathway after attachment to ectodysplasin A receptor (EDAR) is a direct target of miR-98-5p. Lyn, a member of Src family of tyrosine kinases that activates NF-kB via activating PI3K signaling pathway, is a target of miR-98-5p (Toubiana et al., 2015) . PLCG2 is also directly targeted by miR-98-5p. Cytokines such as TNF and TGF-β1 can activate PLCG2 and lead to the activation of ERK and NF-kB signaling pathways (Ma et al., 2019) . On the other hand, miR-223 and miR-214 can directly target 3' UTR of ERC1 gene. The miRNA microarray profiling results demonstrated that the expression of miR-223-3p increased by 13.01 times in patients with ulcerative colitis, and the results of in silico prediction tools determined ERC1 as one of the most important targets of this miRNA in NF-kB pathway (Valmiki et al., 2017) . MyD88, an adaptor protein in NF-κB signaling pathway, is also targeted by miR-214-5p and miR-223-5p. The increased expression of miR-214 by targeting MyD88 reduced the production of inflammatory cytokines in V. harveyi bacterial infection and prevented the exacerbation of inflammation (Chu et al., 2017) . Through the utilization and phosphorylation of IRAK and TRAF6 complexes, MyD88 activates MAPK and NF-kB pathways. IRAK4 enzyme, which is an important threonine/serine protein kinase in MAPK pathway and TLR-mediated immune responses (Kawagoe et al., 2007) , is directly targeted by miR-214. Receptor-interacting protein kinase 1 (RIPK1) that determines the cell fate, death or survival, through NF-kB signaling pathway after receiving a wide variety of pro-inflammatory or pro-death cytokines is a target of miR-223-5p and miR-9-5p (Mifflin et al., 2020) . The results of gene expression analysis and luciferase reporter assay showed that the expression of RIPK1 and RIPK3 in acute kidney injury induced in C57BL/6 mice increased, and miR-223 could directly target 3' UTR of RIPK3 gene (Huang et al., 2018) . TNFR1 associated death domain protein (TRADD), an adaptor protein mediating cell death and activating NF-kB pathway through interacting with TNFRSF1A/TNFR1, is a direct target of miR-214-3p. TRAF2 and TRAF5 were identified as targets of miR-214-5p and miR-223-5p, respectively. Tumor necrosis factor receptor associated factor-2 (TRAF2) and tumor necrosis factor receptor associated factor-5 (TRAF5) lead to the activation of TAK1 via the ubiquitination of IKKγ/NEMO. TAK1 finally activates IKK enzyme and leads to the phosphorylation and deactivation of IκBα and the activation NF-kB pathway (Matsui et al., 2018) . MiR-98-5p is capable of directly targeting the epsilon subunit of this enzyme (IKBKE). BIRC3 (cIAP2), as a member of the family of proteins inhibiting apoptosis by NF-kB pathway, was determined as one target of miR-98-5p. TRAF3 was identified as a potential target of miR-214 and miR-9-5p. The expression of miR-214 increased in the tissues of osteosarcoma patients as well as osteosarcoma cell lines, and this miRNA was capable of directly targeting TRAF3 gene (Rehei et al., 2018) . TAB2, an adaptor protein that forms a complex with TAK1 and is necessary for the activation of NF-kB pathway, is a target of many miRNAs including miR-9-5p, miR-9-3p, miR-98-5p, miR-98-3p, and miR-214-5p. The findings of qRT-PCR in ovariectomized rats (OVX-ASCs) suggested the increased expression of miR-214. In addition, two new targets of miR-214, CTNNB1 (βcatenin) and TAB2, were identified in this study via luciferase reporter assay (K. C. Li et al., 2017) . MiRNAs can directly target the negative regulators of NF-kB pathway. In this regard, miR-223-3p is able to directly target NFKB inhibitor alpha (NFKBIA). It was shown that notch and NF-kB signaling pathways could raise the expression of miR-223, which in turn led to the reduced expression of onco-suppressor FBXW7 in T-cell acute lymphoblastic leukemia (V. Kumar et al., 2014) . Tripartite motif containing 25 (TRIM25) that is a positive regulator of NF-kB signaling pathway is targeted by miR-98-3p, miR-9-5p, miR-223-3p, and miR-214-5p. PARP1, a DNA polymerase playing an important role in the identification of DNA damages and the utilization of DNA repair proteins, is a direct target of miR-223-3p. PARP1 leads to the activation of NF-kB pathway via a TNF-α-induced pathway. Microarray analysis results in JHesoAD1 and esophageal OE33 (adenocarcinoma) cell lines transfected with miR-223 mimics indicated the reduced expression of several target genes including PARP1, CYB5A, and SMARCD1. Afterwards, the luciferase assay established that the 3' UTR of PARP1 gene is directly identified by miR-223 (Streppel et al., 2013) . Note that transcription factors NF-kB1 and NF-kB2 are directly regulated by miR-9-5p and miR-98-5p. MiR-9 in human monocytes was induced by inflammatory cytokines TNF-α and IL-1β, and NF-kB1 was identified as an important target of miR-9 in both in silico and experimental investigations (Bazzoni et al., 2009) . C-FLIP, Bcl-XL, and BCL2, as target genes of NF-kB pathway, are directly targeted by miR-9-5p, miR-98-5p, and both of these miRNAs, respectively. The elevated expression of miR-98 through targeting Bcl2 gene inhibits the growth and resistance tolerance in esophageal squamous cell carcinoma (ESCC) . Moreover, miR-98 plays a critical role in the pathogenesis of osteoarthritis disease through targeting Bcl2 (Jing Wang et al., 2016) . Furthermore, the expression of miR-98 in rat uterus declined in the receptive phase, and miR-98 inhibited apoptosis and increased proliferation in this phase by directly targeting Bclxl (Xia et al., 2014) . As other target genes of NF-kB pathway, TRAF1 and TRAF2 are directly targeted by miR-98-5p and miR-214-5p. EDA2R, which is a member of TNFR family and activates NF-kB signaling pathway, and intercellular adhesion molecule 1 (ICAM1), which is a target of this pathway, are also directly targeted by miR-223-5p and miR-223-3p, respectively. The targeting of other members of TNFR family by miR-223 has been established in the literature. The findings of luciferase reporter assay in HEK293 cells or rat cardiac H9c2 cells demonstrated that miR-223-5p was able to target 3' UTR of TNFR1 and DR6 genes (Qin et al., 2016) . LTA and TNFSF14 ligands, activating NF-kB signaling pathway after attachment to LT-βR receptor, are targeted by miR-214-5p and miR-223-5p, respectively. LT-βR, a member of TNFR family, was also identified as a direct target of miR-98-5p. Finally, TRAM1, a member of steroid receptor coactivator (SRC) family inducing gene overexpression through NF-kB transcription factor, is a target of miR-9-5p. Therefore, it can be concluded that the expression of genes in JAK/STAT and NF-kB signaling pathways is significantly affected by miR-9, miR-98, miR-223, and miR-214, and these miRNAs may play an important role in the pathogenesis of COVID-19. Multiple destructive inflammatory signaling pathways have been implicated in COVID-19 pathogenesis, which leads to multiple-organ damages, physiological deterioration, and eventually death. In the current study, the results obtained from targeting the aforementioned inflammatory pathways in COVID-19 patients were reviewed. It was found that NF-kB and JAK-STAT signaling pathways have key roles in inflammation involved in COVID-19 pathogenesis. NF-kB signaling pathway genes (NFKBIA, NFKB1, RELA, NFKB2) were upregulated in patients with COVID-19 infection. JAK/STAT pathway induced various cytokines including interleukins IL-10, IL-7, IL-6, IL-4, and IL-2 and interferons IFN-α, IFN-β, IFN-γ, and GCS-F. Additionally, we reviewed research works in which miRNAs targeted inflammatory signaling pathways involved in the inflammatory diseases. Our study focused on miR-9, miR-98, miR-223, and miR-214, which make a unique contribution to the regulation of inflammatory pathways related to COVID-19 pathogenesis, and whose target genes are related to NF-kB and JAK-STAT pathways. Based on the results, TRIM25, which is a positive regulator of NF-kB signaling pathway, is targeted by miR-98-3p, miR-9-5p, miR-223-3p, and miR-214-5p. PARP1, which leads to the activation of NF-kB pathway via a TNFα-induced pathway, is targeted by miR-223-3p. Transcription factors NFKB1 and NFKB2 are directly regulated by miR-9-5p and miR-98-5p. In addition, TNF-α, IL-1β, and NF-kB1 were targeted by miR-9. C-FLIP, Bcl-XL, and BCL2, as target genes of NF-kB pathway, are directly targeted by miR-9-5p, miR-98-5p, and both of these miRNAs, respectively. TRAF1 and TRAF2 are directly targeted by miR-98-5p and miR-214-5p. TNFR family and ICAM1 are also directly targeted by miR-223-5p and miR-223-3p, respectively. Recently, several miRNA-based treatments have illustrated therapeutic promise in clinical trials. These treatments include the use of miRNA antagonists to increase the expression of specific proteins that are downregulated or miRNA mimics to decrease the expression of proteins that induce abnormally (Hanna et al., 2019) . For example, miravirsen was the first miRNA-based drug that was successfully applied for hepatitis C virus (HCV) infection. This drug suprresses viral RNA levels by sequestering miR-122 away from the viral genome (Janssen et al., 2013) . In regard to the delivery of miRNA targets, the poor half-life of miRNAs and the toxicity of delivery vehicles are obstacles to treatment. Nanoparticles, lipid-based carriers, and exosomes are highly recommended for direct delivery into the lungs in the clinical setting (N. Chauhan et al., 2021; Fu et al., 2019) . In conclusion, miRNAs are associated with inflammation processes, and their target networks provide new knowledge that can pave the way for new therapeutic strategies for COVID-19 pathogenesis. In this regard, miR-9, miR-98, miR-223, and miR-214 can be considered as potential targets for new therapeutic strategies in COVID-19 diseases by targeting inflammatory pathways. Not applicable. None. The authors declare no conflict of interest, financial or otherwise. The JAK/STAT signaling pathway is a transducer of a wide variety of signals for development and homeostasis in animals from humans to flies. In mammals, this pathway is the main mechanism of signal transduction from growth factors and cytokines. Following the binding of a cytokine to a receptor, the JAK tyrosine kinase activates STAT. Upon the activation of STAT, it is dimerized and transferred to the nucleus, leading to the regulation of the transcription of target genes. The targets of the miR-223, miR-98, miR-9, and miR-214 in these pathway are shown using red arrows (KEGG PATHWAY database). Nuclear factor-kappa B (NF-kappa B) regulates genes played important roles in immunity, inflammation and cell survival. There are several pathways resulting in NF-kappa B-activation. The canonical pathway is induced by TNF-α, IL-1 or byproducts of bacterial and viral infections. This pathway depends on IKKmediated IkappaB-alpha phosphorylation on Ser32 and 36, leading to its degradation, which allows the p50/p65 NF-kappa B dimer to enter the nucleus and activate gene transcription. The targets of the miR-223, miR-98, miR-9, and miR-214 in these pathway are shown using red arrows (KEGG PATHWAY database). MiRTarBase, and Target scan). Potential target genes of miR-9-5p (pink), miR-9-3p (light pink), miR-98-5p (light blue), miR-98-3p (medium blue), miR-214-5p (green), miR-214-3p (dark purple), and miR-223-3p (medium purple), and miR-223-5p (light purple) are shown in the picture. MiRNAs with more interactions are illustrates with larger sizes. Potential interactions between genes with each other and miRNAs of interest were depicted via Cytoscape software. TRAF3 TRAM1 NFKB1 CFLAR TAB3 IL17A IRAK4 TRAF2 LTA CARD14 TRADD miR-9-5p miR-9-5p miR-9-5p, miR-214-5p, miR-9-3p miR-9-5p miR-9-5p, miR-214-5p miR-9-5p miR-9-5p miR-9-5p miR-9-5p miR-9-5p miR-9-5p miR-9-5p MicroRNAs and SARS-CoV-2 life cycle, pathogenesis, and mutations: biomarkers or therapeutic agents? Micro-RNAs in the regulation of immune response against SARS CoV-2 and other viral infections Angiotensin II induces IL-6 expression and the Jak-STAT3 pathway in aortic adventitia of LDL receptordeficient mice Challenges presented by MERS corona virus, and SARS corona virus to global health Implications of SARS-CoV-2 mutations for genomic RNA structure and host microRNA targeting The impact of miR-183/182/96 gene regulation on the maturation, survival, and function of photoreceptor cells in the retina MiR-221/222 promote chemoresistance to cisplatin in ovarian cancer cells by targeting PTEN/PI3K/AKT signaling pathway Regenerative Medicine Approaches in COVID -19 Pneumonia The effect of oleuropein on apoptotic pathway regulators in breast cancer cells Predicted therapeutic targets for COVID-19 disease by inhibiting SARS-CoV-2 and its related receptors. Informatics in Medicine Unlocked Induction and regulatory function of miR-9 in human monocytes and neutrophils exposed to proinflammatory signals miR-1207-5p Can Contribute to Dysregulation of Inflammatory Response in COVID-19 via Targeting SARS-CoV-2 RNA Clinical features of patients infected with 2019 novel coronavirus in Wuhan Immune response in COVID-19: addressing a pharmacological challenge by targeting pathways triggered by SARS-CoV-2 COVID-19: A collision of complement, coagulation and inflammatory pathways COVID-19: fighting the invisible enemy with microRNAs MicroRNA-223 promotes type i interferon production in antiviral innate immunity by targeting forkhead box protein O3 (FOXO3) Inducible microRNA-223 down-regulation promotes TLR-triggered IL-6 and IL-1β production in macrophages by targeting STAT3 Cytokine Release Syndrome in Severe COVID-19: Interleukin-6 Receptor Antagonist Tocilizumab May Be the Key to Reduce Mortality Inducible microRNA-214 contributes to the suppression of NF-κB-mediated inflammatory response via targeting myd88 gene in fish miR-15 and miR-16 induce apoptosis by targeting BCL2 MicroRNA-155 enhances T cell trafficking and antiviral effector function in a model of coronavirusinduced neurologic disease MicroRNA-223 controls susceptibility to tuberculosis by regulating lung neutrophil recruitment A new approach for COVID-19 treatment by micro-RNA. Medical Hypotheses JAK Inhibition as a New Treatment Strategy for Patients with COVID-19 A transcription regulatory network within the ACE2 locus may promote a pro-viral environment for SARS-CoV-2 by modulating expression of host factors Recent progress in microRNA-based delivery systems for the treatment of human disease Immune Response, Inflammation, and the Clinical Spectrum of COVID-19 Angiotensin-converting enzyme 2 inhibits lung injury induced by respiratory syncytial virus Impaired type I interferon activity and exacerbated inflammatory responses in severe Covid-19 patients The potential for microRNA therapeutics and clinical research SARS-CoV-2 Cell Entry Depends on ACE2 and TMPRSS2 and Is Blocked by a Clinically Proven Protease Inhibitor MicroRNA Mimics or Inhibitors as Antiviral Therapeutic Approaches Against COVID-19 Apoptotic effects of valproic acid on miR-34a, miR-520h and HDAC1 gene in breast cancer Treatment of HCV Infection by Targeting MicroRNA Dysregulated miR-98 Contributes to Extracellular Matrix Degradation by Targeting IL /STAT3 Signaling Pathway in Human Intervertebral Disc Degeneration Cancer exosome-derived miR-9 and miR-181a promote the development of early-stage MDSCs via interfering with SOCS3 and PIAS3 respectively in breast cancer Upregulation of microRNA-98 increases radiosensitivity in esophageal squamous cell carcinoma Essential role of IRAK-4 protein and its kinase activity in Toll-like receptor-mediated immune responses but not in TCR signaling MicroRNA regulation of cardiac conduction and arrhythmias MicroRNAs and STAT interplay Is diabetes mellitus associated with mortality and severity of COVID-19? A meta-analysis. Diabetes and Metabolic Syndrome Notch and NF-kB signaling pathways regulate miR-223/FBXW7 axis in T-cell acute lymphoblastic leukemia Expression of elevated levels of proinflammatory cytokines in SARS-CoV-infected ACE2+ cells in SARS patients: relation to the acute lung injury and pathogenesis of SARS Coronavirus infections and immune responses Baculovirus-Mediated miR-214 Knockdown Shifts Osteoporotic ASCs Differentiation and Improves Osteoporotic Bone Defects Repair Role of miR-9-5p in preventing peripheral neuropathy in patients with rheumatoid arthritis by targeting REST/miR-132 pathway The landscape of lung bronchoalveolar immune cells in COVID-19 revealed by single-cell RNA sequencing Vertebrate microRNA genes The upregulation of miR-98-5p affects the glycosylation of IgA1 through cytokines in IgA nephropathy Pirin is an iron-dependent redox regulator of NF-κB The effects of microRNA-98 inhibits cell proliferation and invasion by targeting STAT3 in nasopharyngeal carcinoma MicroRNA-98 negatively regulates IL-10 production and endotoxin tolerance in macrophages after LPS stimulation Targeting JAK-STAT Signaling to Control Cytokine Release Syndrome in COVID-19 PLCG2 promotes hepatocyte proliferation in vitro via NF-κB and ERK pathway by targeting bcl2, myc and ccnd1. Artificial Cells MiR-9-5p and miR-106a-5p dysregulated in CD4+ T-cells of multiple sclerosis patients and targeted essential factors of T helper17/regulatory T-cells differentiation MicroRNA-9 promotes IL-6 expression by inhibiting MCPIP1 expression in IL-1β-stimulated human chondrocytes HHS Public Access MicroRNome analysis unravels the molecular basis of SARS infection in bronchoalveolar stem cells TLR7 and TLR8 activate distinct pathways in monocytes during RNA virus infection MiR-200b attenuates IL-6 production through IKKβ and ZEB1 in human gingival fibroblasts Receptor-interacting protein kinase 1 (RIPK1) as a therapeutic target Host cell entry of Middle East respiratory syndrome coronavirus after two-step, furin-mediated activation of the spike protein STAT1: A novel target of miR-150 and miR-223 is involved in the proliferation of HTLV-I-transformed and ATL cells miR-15a/16 Regulates Macrophage Phagocytosis after Bacterial Infection The impact of microRNA-223-3p on IL-17 receptor d expression in synovial cells Overview of MicroRNA Biogenesis, Mechanisms of Actions, and Circulation New-Onset Diabetes in Covid-19 Suppressor of Cytokine Signaling (SOCS) 1 inhibits type I interferon (IFN) signaling via the interferon α receptor (IFNAR1)-associated tyrosine kinase tyk2 miR-223 is a coordinator of breast cancer progression as revealed by bioinformatics predictions MicroRNA expression profile of bronchioalveolar stem cells from mouse lung MicroRNA-223-5p and -3p Cooperatively suppress necroptosis in ischemic/reperfused hearts The JAK/STAT signaling pathway MicroRNA-214 functions as an oncogene in human osteosarcoma by targeting TRAF3 Decreased serum levels of the inflammaging marker miR-146a are associated with non-clinical response to tocilizumab in COVID-19 patients Computational analysis of microRNA-mediated interactions in SARS-CoV-2 infection The role of JAK-STAT signaling pathway and its regulators in the fate of T helper cells Biological Impacts of MicroRNAs in Covid-19: Implications for Anti-Viral miRNA-Based Therapies. Archives of Clinical Infectious Diseases COVID-19 patients upregulate toll-like receptor 4-mediated inflammatory signaling that mimics bacterial sepsis Molecular advances in the cell biology of SARS-CoV and current disease prevention strategies MicroRNA 223 is upregulated in the multistep progression of barrett's esophagus and modulates sensitivity to chemotherapy by targeting PARP1 SLIM is a nuclear ubiquitin E3 ligase that negatively regulates STAT signaling Src-familytyrosine kinase Lyn is critical for TLR2-mediated NF-κ activation through the PI 3-kinase signaling pathway MicroRNA exhibit altered expression in the inflamed colonic mucosa of ulcerative colitis patients Over-expression of microRNA-223 inhibited the proinflammatory responses in Helicobacter pylori-infection macrophages by down-regulating IRAK-1 MiR-98 promotes chondrocyte apoptosis by decreasing Bcl-2 expression in a rat model of osteoarthritis Up-regulation of IL-6 and TNF-α induced by SARS-coronavirus spike protein in murine macrophages via NF-κB pathway MiR-223-3p alleviates vascular endothelial injury by targeting IL6ST in Kawasaki disease MicroRNA-9 inhibits NLRP3 inflammasome activation in human atherosclerosis inflammation cell models through the JAK1/STAT signaling pathway MicroRNAs in retina during development of experimental autoimmune uveoretinitis in rats Potential role of ACE2-related microRNAs in COVID-19-associated nephropathy Integrating non-coding RNAs in JAK-STAT regulatory networks Pir-B inhibits the DC function and disturbs the Th17/Treg balance in lung cancer murine model Impact of miR-223-3p and miR-2909 on inflammatory factors IL-6, IL-1ß, and TNF-α, and the TLR4/TLR2/NF-κB/STAT3 signaling pathway induced by lipopolysaccharide in human adipose stem cells MiR-98 is involved in rat embryo implantation by targeting Bcl-xl Transcriptomic Characteristics of Bronchoalveolar Lavage Fluid and Peripheral Blood Mononuclear Cells SARS-CoV infection crosstalk with human host cell noncoding-RNA machinery: An in-silico approach MiR-98 modulates cytokine production from human PBMCs in systemic lupus erythematosus by targeting IL-6 mRNA Association of inflammatory markers with the severity of COVID-19: A meta-analysis Angiotensin-converting enzyme 2 (ACE2) as a SARS-CoV-2 receptor: molecular mechanisms and potential therapeutic target Downregulated expression of miR-223 promotes Toll-like receptor-activated inflammatory responses in macrophages by targeting RhoB Nucleocapsid protein of SARS-CoV activates interleukin-6 expression through cellular transcription factor NF-κB Ngn1 inhibits astrogliogenesis through induction of miR-9 during neuronal fate specification Heightened Innate Immune Responses in the Respiratory Tract of COVID-19 Patients ACE and ACE2: A tale of two enzymes signaling pathways are highly activated in acute inflammation, and the excessive activity of these pathways in COVID-19 patients likely exacerbates the inflammatory responses of the host A groups of non-coding RNAs (miRNAs) such as miR-9 This review was supported by the Shahrekord University of Medical Sciences, Shahrekord, Iran, Semnan University of Medical Sciences, Semnan, Iran, and Mashhad University of Medical Sciences, Mashhad, Iran.