Open Access

Host cell entry mediators implicated in the cellular tropism of SARS‑CoV‑2, the pathophysiology of COVID‑19 and the identification of microRNAs that can modulate the expression of these mediators (Review)

  • Authors:
    • Periklis Katopodis
    • Harpal S. Randeva
    • Demetrios A. Spandidos
    • Sayeh Saravi
    • Ioannis Kyrou
    • Emmanouil Karteris
  • View Affiliations

  • Published online on: December 20, 2021     https://doi.org/10.3892/ijmm.2021.5075
  • Article Number: 20
  • Copyright: © Katopodis et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The pathophysiology of coronavirus disease 2019 (COVID‑19) is mainly dependent on the underlying mechanisms that mediate the entry of severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) into the host cells of the various human tissues/organs. Recent studies have indicated a higher order of complexity of the mechanisms of infectivity, given that there is a wide‑repertoire of possible cell entry mediators that appear to co‑localise in a cell‑ and tissue‑specific manner. The present study provides an overview of the ‘canonical’ SARS‑CoV‑2 mediators, namely angiotensin converting enzyme 2, transmembrane protease serine 2 and 4, and neuropilin‑1, expanding on the involvement of novel candidates, including glucose‑regulated protein 78, basigin, kidney injury molecule‑1, metabotropic glutamate receptor subtype 2, ADAM metallopeptidase domain 17 (also termed tumour necrosis factor‑α convertase) and Toll‑like receptor 4. Furthermore, emerging data indicate that changes in microRNA (miRNA/miR) expression levels in patients with COVID‑19 are suggestive of further complexity in the regulation of these viral mediators. An in silico analysis revealed 160 candidate miRNAs with potential strong binding capacity in the aforementioned genes. Future studies should concentrate on elucidating the association between the cellular tropism of the SARS‑CoV‑2 cell entry mediators and the mechanisms through which they might affect the clinical outcome. Finally, the clinical utility as a biomarker or therapeutic target of miRNAs in the context of COVID‑19 warrants further investigation.

1. Introduction

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is an enveloped RNA virus that is transmitted mainly via air droplets, and can cause a viral infectious disease in humans, namely coronavirus disease 2019 (COVID-19), which was first identified towards the end of 2019 (1). COVID-19 manifestations range from mild (asymptomatic or mild respiratory tract infection in the majority of reported cases) to severe and the disease can be fatal in high-risk individuals, with a spectrum of respiratory and/or extra-pulmonary manifestations that may require hospitalization and potentially mechanical ventilation and intensive care unit support (1). Of note, SARS-CoV-2 is highly virulent, has a tendency to mutate and exhibits a cellular tropism within the human body that appears to be associated with the expression of various host cell entry mediators, such as the angiotensin converting enzyme 2 (ACE2) (2-4). The present review article presents a concise overview of the key host cell entry mediators which have been identified to play a role in the infection of human cells by SARS-CoV-2, summarizing the evidence on the mechanisms through which ACE2 and additional host receptors/proteins appear to be implicated in the cellular tropism of SARS-CoV-2 and the pathophysiology of COVID-19. Moreover, the present review article aims to provide insight into the mechanisms through which these cell entry mediators may be regulated by certain microRNAs (miRNAs/miRs).

2. Angiotensin converting enzyme 2, and transmembrane protease serine 2 and 4

Both SARS-CoV and SARS-CoV-2 have been reported to use ACE2 as the key receptor for their host cell entry (4-6). Indeed, an initial step in these viral infections is the binding of this cellular receptor with the viral spike S glycoprotein (S protein), which consists of the S1 and S2 subunits (Fig. 1). The S1 subunit contains a C-terminal receptor-binding domain that recognizes and binds to the specific host receptor (e.g., ACE2), whilst the S2 subdomain is responsible for the fusion of the viral membrane with the host cell membrane via the fusion peptide. Following this binding to ACE2, a protease cleavage of the S protein follows. This process is catalysed by transmembrane protease serine (TMPRSS)2, and other host proteases, such as cathepsin L and furin at the S1-S2 site, which activate the entry of SARS-CoV-2 (5,7-11).

Based on consistent data, ACE2 is now well-established as the key mediator that facilitates the entry of SARS-CoV-2 into human host cells (6,12,13). Notably, ACE2 is highly expressed in the kidneys, small intestine, prostate and lung alveolar epithelial type II cells, which explains the documented tropism of this novel coronavirus and the related pulmonary and extrapulmonary symptoms that a SARS-CoV-2 infection may cause (14). The analysis of autopsy tissue from fatal COVID-19 cases has most frequently detected SARS-CoV-2 RNA in the airway epithelium, whilst SARS-CoV-2 RNA has also been found to be highly co-localised in cells expressing TMPRSS2 (15-18). Of note, it has been suggested that DNA polymorphisms for ACE2 and/or TMPRSS2 may be associated with the genetic susceptibility of different populations to severe COVID-19 infection (9). Along with TMPRSS2, TMPRSS4 has also been shown to be involved in the infectivity of SARS-CoV-2. In human small intestinal enterocytes, the presence of both enzymes drives the cleavage of the spike S glycoprotein of SARS-CoV-2 and enhances subsequent membrane fusion (19). Moreover, TMPRSS2 and TMPRSS4, along with ACE2, are abundantly co-expressed in the small intestine and colon (20). Subsequently, the authors have previously demonstrated that TMPRSS4 is overexpressed in 11 types of cancer in the colon mucosa and in all the brain regions that are associated with the sense of taste and smell (21).

3. Neuropilin-1

It is noteworthy that the relatively low/moderate expression of ACE2 in the human respiratory system has triggered further research focusing on the discovery of potential additional cell proteins/receptors that may mediate the SARS-CoV-2 infectivity and contribute to the selective tissue/organ tropism of this new coronavirus (22,23). In this context, two elegant studies by Daly et al (24) and Cantuti-Castelvetri (25) et al have demonstrated that neuropilin (NRP)1 constitutes an additional cellular mediator implicated in the entry of SARS-CoV-2 into human cells. Indeed, based on the experimental data of both these studies, it is evident that, not only ACE2, but also NRP1 can act as a host cell mediator that enhances the infectivity of SARS-CoV-2 and contributes to its documented tissue/organ tropism.

Overall, NRPs are classed as non-tyrosine kinase, single-transmembrane glycoproteins which are greatly conserved in vertebrates and constitute co-receptors for a number of molecules, such as semaphorins and vascular endothelial growth factors (VEGFs), playing a key role in numerous physiological processes (e.g., neuronal development and axon control, immune function angiogenesis, cell proliferation and vascular permeability) (23-26).

Of note, the aforementioned studies by Daly et al (24) and Cantuti-Castelvetri et al (25) suggest that, unlike SARS-CoV S proteins, SARS-CoV-2 S proteins have a polybasic sequence motif (Arg-Arg-Ala-Arg) at the S1-S2 junction/boundary that conforms to the 'C-end rule' (CendR motif) [R-XX-R; where R denotes arginine (Arg), can be replaced by lysine (Lys)] and serves as a cleavage site for the convertase furin (Fig. 1) (24-26). Thus, SARS-CoV-2 can enter host cells more easily with the aid of NRP1, boosting its infectivity and promoting its tropism (24,25). Indeed, with the use of human cell lines, it has been shown that NRP1 enhances infection by a clinical SARS-CoV-2 isolate and lentiviral particles pseudotyped with the S protein of SARS-CoV-2 by binding to this CendR motif of the furin-cleaved SARS-CoV-2 S1 protein. This NRP1-mediated increase in SARS-CoV-2 infectivity appears to be due to increased viral entry into host cells, rather than increased viral binding to the cell membrane, whilst it is amplified when ACE2 and TMPRSS2 are present. On the other hand, the rate of NRP1-enhanced SARS-CoV-2 infection in human cell cultures has been shown to be suppressed by using a monoclonal blocking antibody against the extracellular NRP1 b1b2 space, or a specific NRP1 mutant which ties the CendR-binding b1 domain (24,26,27). In addition, as previously demonstrated, mutated variants of SARS-CoV-2 with a modified furin cleavage site of the S protein (erased poly-basic cleavage site or impervious to furin-intervened cleavage) were not subject to NRP1-mediated infectivity, and similarly transformations in the NRP1 b1 domain likewise restrained the NRP1-S1 interaction (24,26,27).

Notably, the upregulation of NRP1 gene expression has also been demonstrated in lung tissue samples and infected olfactory epithelial cells from patients with COVID-19; further co-staining revealed the infection of cells that were positive for oligodendrocyte transcription factor 2, which is primarily expressed by olfactory neuronal progenitors (27).

Given these data on NRP1 and since extra-pulmonary symptoms (e.g., gastrointestinal and neurological symptoms) are frequently identified in relation to COVID-19, extensive additional research is still required to identify the association between SARS-CoV-2 tropism and the mechanisms through which host cell infection mediators, such as NRP1, are dispersed across the various tissues/organs of the human body. This may further aid in the development of novel antiviral drugs against COVID-19, which for example could be targeting NRP1 and its role in promoting the entry of SARS-CoV-2 into host cells.

4. Emerging SARS-CoV-2 cell entry mediators

Following the initial findings regarding the role of host cell receptors/mediators, such as ACE2 and NRP1, data are also emerging for a number of additional cellular factors which may also facilitate or inhibit the infectivity of SARS-CoV-2, including glucose-regulated protein 78 (GRP78), basigin (BSG; CD147), kidney injury molecule-1 (KIM1), heparan sulfate (HS), ADAM metallopeptidase domain 17 [also termed tumour necrosis factor-α convertase (ADAM17)], surfactant protein D (SP-D), metabotropic glutamate receptor subtype 2 (mGluR2) and Toll-like receptor 4 (TLR4).

In this context, heat shock protein A5 (HSPA5), also termed GRP78, has been reported to be implicated in a possible route for SARS-CoV-2 cell attachment and entry. Indeed, it has been proposed that the recognition site for GRP78 is found in SARS-CoV-2 (28), and that the binding is more favourable between regions III (C391-C525) and IV (C480-C488) of the SARS-CoV-2 protein spike model and GRP78 (29). Moreover, CD147 (also known as EMMPRIN or BSG; a transmembrane glycoprotein that belongs to the large immunoglobulin superfamily) has also been proposed as another molecule that can facilitate the entry of SARS-CoV-2 into host cells by endocytosis. This is supported by the findings of an in vitro study in which SARS-CoV-2 amplification was inhibited when CD147 was blocked in Vero E6 and BEAS-2B cell lines (30). However, based on the findings of a later study, another group argued that there was no evidence for a direct interaction between the viral spike protein and this particular molecule (31). Such contradictory findings highlight the current need for further research into the entire spectrum of potential host-pathogen interactions for SARS-CoV-2.

Furthermore, given that kidney-related complications are common in patients with COVID-19 (32), the involvement of KIM1 has been recently investigated. Accordingly, through a series of experiments, it was previously demonstrated that KIM1 is capable of interacting with the receptor-binding domain of this new coronavirus, subsequently enabling its attachment to the plasma membrane (33). These findings suggest that this molecule may mediate and exacerbate SARS-CoV-2 infection of the kidneys (33). Moreover, KIM1 may function as a biomarker of acute kidney injury, which has been associated with a poor prognosis of patients with COVID-19 (34).

mGluR2 (GRM2), a G protein-coupled receptor which inhibits adenylyl cyclase and regulates the mechanisms through which glutamate can control cell excitability, is another potential cell entry receptor for SARS-CoV-2. Indeed, recent experiments have demonstrated that mGluR2 interacts directly with the S protein of SARS-CoV-2, whilst the knockdown of mGluR2, even though it does not affect binding to the plasma membrane, reduces the clathrin-mediated endocytosis of this virus (35). Moreover, in vivo studies corroborated these findings, where viral infectivity in lungs of mice was significantly reduced, when mGluR2 was knocked out (35). Of note, angiotensin II receptor type 2 is another G protein-coupled receptor, which, based on an in silico simulation data has revealed an interaction with the S protein of SARS-CoV-2 (36). However, further in vitro and/or in vivo studies are required to validate these initial modelling data.

Apart from direct cell entry mediators of SARS-CoV-2 into host cells, there have been molecules which appear to function as co-factors in this process, such as HS and ADAM17. Of note, HS is a co-factor for ACE2-mediated viral entry and a potential therapeutic target (e.g., using mitoxantrone) (37). On the other hand, ADAM17 has been shown to regulate the shedding of the ACE2 ectodomain (38). This is in line with its role in releasing ectodomains of a wide repertoire of cytokines, enzymes or other molecules (38,39). However, due to a potential redundancy in viral cell entry, it has been demonstrated that TMPRSS2 can compete with ADAM17 for ACE2 processing (40). Furthermore, SP-D is an innate immune molecule with a main role in clearing pathogens and apoptotic/necrotic cells from pulmonary and extra-pulmonary mucosal sites (41). Given that human SP-D can recognise the spike glycoprotein of SARS-CoV, a recent study demonstrated that the treatment of 293T cells overexpressing ACE2 with a recombinant fragment of human SP-D (rfhSP-D) inhibited the interaction of the S1 spike proteins with the cell membrane, raising the possibility of an alternative therapeutic target (42). Indeed, in another study, when clinical samples were used, treatment with rfhSP-D inhibited viral replication, and appeared to be more efficient than the antiviral drug remdesivir, when Vero cells were treated in vitro (43). Currently, a Phase 1 clinical trial (ClinicalTrials.gov identifier: NCT04659122) is ongoing, entitled: 'Phase 1b Open-label, Single Arm, Cohort Dose Escalation Study Evaluating the Safety, Tolerability, and Feasibility of Intervention With AT-100 (rhSP-D) in Intubated Patients Receiving Invasive Mechanical Ventilation With Severe COVID-19 Infection'. Finally, in a previous observational study on 39 patients with COVID-19, SP-D levels were found to be significantly higher in severe compared to mild cases, suggesting the possibility of its use as a biomarker of severity as well (44).

Finally, another innate immune molecule that has gained increasing interest in the context of COVID-19 is TLR4, which represents a key cell surface receptor that induces the secretion of pro-inflammatory cytokines and interferons to fight infection (45). Indeed, there is accumulating evidence to indicate that SARS-CoV-2 can bind to TLR4, whilst treatment with resatorvid (a TLR4 specific inhibitor) has been shown to abolish the secretion of IL1B by SARS-CoV-2 in THP-1 cells (46,47).

Overall, identifying the underlying mechanisms that facilitate the entry of SARS-CoV-2 into host cells (Fig. 1) is currently the focus of intensive research which is expected to provide further valuable insight into the pathophysiology of COVID-19, identifying the human tissues/organs that are more vulnerable to a SARS-CoV-2 infection. Ultimately, this may also aid in the development of more precise treatments against COVID-19 sequelae. A list of the 'canonical' and potential cell entry mediators is provided in Table I.

Table I

Genes related to SARS-CoV-2 entry into host cells.

Table I

Genes related to SARS-CoV-2 entry into host cells.

Cell entry mediatorsAbbreviationProposed action(Refs.)
Angiotensin converting enzyme 2ACE2Canonical receptor for the S glycoprotein (S1 domain)(4,48,49)
Transmembrane protease, serine 2 and serine 4TMPRSS2 TMPRSS4Mediate fusion of viral and host cell membranes after proteolytic cleavage at the S1/S2 region(7,19,50,51)
Neuropilin-1NRP1Additional SARS-CoV-2 host cell receptor, increasing its infectivity and contributing to its tropism(20,23-25,48)
Toll-like receptor 4TLR4Potential alternative receptor for SARS-CoV-2(46,47,52)
Kidney injury molecule-1KIM1Potential interaction with SARS-CoV-2 receptor-binding domain(33,34)
BasiginBSG/CD147Potential facilitator of the entry of SARS-CoV-2 into host cells by endocytosis(30,31)
Heat shock protein A5 or glucose regulating protein 78HSPA5/GRP78Possible route for SARS-CoV-2 cell attachment and entry(28,29)
ADAM metallopeptidase domain 17 or TNF-α convertase enzymeADAM17Regulates shedding of the ACE2 ectodomain(10,38,39)

[i] SARS-CoV2, severe acute respiratory syndrome coronavirus 2.

5. Identification of miRNAs that can target and regulate the gene expression of SARS-CoV-2 cell entry mediators

miRNAs are single-strand, endogenous, non-coding, short RNAs of 22-25 nucleotides in length, which can regulate gene expression post-transcriptionally and subsequently alter signalling pathways (53). A series of review articles have elegantly summarised the functions of miRNAs in health and disease (54-57). Of note, miRNAs regulate the expression of key inflammatory cytokines involved in the massive recruitment of immune cells to the lungs, such as IL6 and TNFα. Several studies have demonstrated that changes in the expression of miRNAs in patients with COVID-19 may be a predictor of tissue damage and lung inflammation, whilst miRNAs can also be potent diagnostic biomarkers for COVID-19 (58-60).

Several miRNAs have been either predicted or shown to target SARS-CoV-2 RNAs (61). Indeed, a number of miRNAs have been found to target the RNA S glycoprotein sequence of SARS-CoV-2, which interacts with ACE2 for viral entry into host cells, including hsa-miR-4661-3p (62), hsa-miR-510-3p, hsa-miR-624-5p, hsa-miR-497-5p (63), hsa-miR-622, hsa-miR-761, miR-A3r, hsa-miR-15b-5p, miR-A2r, hsa-miR-196a-5p (64), and miR-338-3p, miR-4661-3p, miR-4761-5p, hsa-miR-4464, hsa-miR-1234-3p, hsa-miR-7107-5p and hsa-miR-885-5p, which have been shown to bind to the receptor binding domain of the S gene (62).

As such, a number of studies have identified host cell miRNAs that target different SARS-CoV-2 proteins. Considering the aforementioned significance of ACE2 and TMPRSS2 in SARS-CoV-2 infection, in a previous study, a TargetScan analysis revealed several miRNAs that could directly target these two receptors, including hsa-mi-200b-3p, hsa-miR-200c-3p and hsa-miR-429 for ACE2, and hsa-let7a-5p, hsa-let7b-5p, hsa-let7c-5p, hsa-let7d-5p, hsa-let7e-5p, hsa-let7f-5p, hsa-let7g-5p, hsa-let7i-5p, hsa-miR-98-5p, has-miR-4458 and hsa-miR-4500 for TMPRSS2; thus, these miRNAs may be utilised as potent therapeutic molecules to regulate key proteins that are required for viral entry to the host airway/lung epithelial cells (59). Moreover, TMPRSS2, which activates the spike protein of SARS-CoV-2 to promote viral infection, has been predicted to be regulated by MR147-3p in the gut (62).

Overall, several cell host miRNAs that target the ACE2 and TMPRSS2 genes can prevent attachment and the entry of SARS-CoV-2 into different tissues. For example, hsa-miR-98-5p has been shown to directly target the 3′-UTR of TMPRSS2, both in human lung microvascular endothelial cells (HMVEC-L) and human umbilical vein endothelial cells (65). Furthermore, the hsa-let-7e/hsa-mir-125a and hsa-mir-141/hsa-miR-200 miRNA families inhibit ACE2 and TMPRSS2 expression (66). It has also been shown that miR-200c is essential for SARS-CoV-2 binding to the ACE2 receptor and entry into cardiomyocytes (67), while miR-98-5p can inhibit TMPRSS2 expression in human endothelial cells (65). In a previous study, bioinformatics analysis revealed that lysine-specific demethylase 5B can regulate ACE2 and TMPRSS2 via the transcriptional repression of let-7e/miR-125a and miR-141/miR-200 miRNAs; thus, these miRNAs are crucial for the function of ACE2 and TMPRSS2 (66).

Notably, a recent study provided novel evidence of an interplay between host miRNAs and SARS-CoV-2, as well as the mechanisms through which these interactions can drive pathogenesis by dysregulating certain immune pathways (68). A recent mini-review, provided an excellent overview of the role of COVID-19-associated miRNAs as potential therapeutic targets and biomarkers (69). Currently, there are a number of clinical trials/studies investigating miRNAs in the context of COVID-19. For example, in an observational study on the expression of cytokines, transcriptome and miRNAs in COVID-19 patients is currently recruiting (ClinicalTrials.gov identifier: NCT04583566). Similarly, 'The Effect of miRNA and Epigenetic Modifications on Prognosis in Covid-19 Infection' will also be investigated (ClinicalTrials.gov Identifier: NCT04411563), whilst another study aims to assess the mechanisms through which miRNA levels relate to viral infection (ClinicalTrials.gov Identifier: NCT04346160). In terms of therapeutics, a phase 2 trial is currently enrolling by invitation, with miRNA-containing exosomes as an aerosol inhaler (ClinicalTrials.gov Identifier: NCT04602442).

Of note, the targeting of PARP-1 by miRNAs has been suggested to hold therapeutic value for COVID-19-related pathologies (70). In a recent in silico study, a number of hypothalamic miRNAs that can bind and regulate ACE2 and TMPRSS2 were identified (71). The authors of that study concluded that these miRNAs may be used in the search for novel therapies for the neurological symptoms in patients with COVID-19. The present study expanded on these initial observations by identifying potential miRNA modulators of all remaining SARS-CoV-2 cell entry mediators, using miRDB, an online database for miRNA target prediction and functional annotations (mirdb.org) (72,73). For the present study, the target prediction score was set >80 for higher confidence in the interactions. As such, Table II provides a list of five potential candidate miRNAs against TMPRRS4; Table III provides a list of 69 miRNAs for NRP1; Table IV indicates 36 potential miRNA interactions with ADAM17; Table V presents five interactions with KIM1; Table VI presents a list of 27 interactions with GRP78 (HSPA5); Table VII presents only one possible interaction identified between a miRNA and CD147 (BSG); Table VIII presents 12 interactions identified between miRNAs and TLR4; and finally, Table IX outlines five possible interactions of these regulatory RNAs with mGluR2.

Table II

List of miRNAs that indicate strong binding potential against transmembrane serine protease 4.

Table II

List of miRNAs that indicate strong binding potential against transmembrane serine protease 4.

Target rankTarget scoremiRNA namemiRNA sequence
188 hsa-miR-6721-5p 5′-ugggcaggggcuuauuguaggag-3′
285hsa-miR-3671 5′-aucaaauaaggacuagucugca-3′
382hsa-miR-496 5′-ugaguauuacauggccaaucuc-3′
481 hsa-miR-551b-3p 5′-gcgacccauacuugguuucag-3′
581hsa-miR-551a 5′-gcgacccacucuugguuucca-3′

Table III

List of miRNAs that indicate strong binding potential against neuropilin-1.

Table III

List of miRNAs that indicate strong binding potential against neuropilin-1.

Target rankTarget scoremiRNA namemiRNA sequence
198hsa-miR-3646 5′-aaaaugaaaugagcccagccca-3′
297hsa-miR-7977 5′-uucccagccaacgcacca-3′
396hsa-miR-6844 5′-uucuuuguuuuuaauucacag-3′
495hsa-miR-124-3p 5′-uaaggcacgcggugaaugccaa-3′
595hsa-miR-3133 5′-uaaagaacucuuaaaacccaau-3′
695hsa-miR-5094 5′-aaucagugaaugccuugaaccu-3′
795hsa-miR-506-3p 5′-uaaggcacccuucugaguaga-3′
895hsa-miR-153-5p 5′-ucauuuuugugauguugcagcu-3′
994 hsa-miR-4755-5p 5′-uuucccuucagagccuggcuuu-3′
1094 hsa-miR-148a-3p 5′-ucagugcacuacagaacuuugu-3′
1194hsa-miR-152-3p 5′-ucagugcaugacagaacuugg-3′
1294 hsa-miR-148b-3p 5′-ucagugcaucacagaacuuugu-3′
1394hsa-miR-3148 5′-uggaaaaaacuggugugugcuu-3′
1494 hsa-miR-5006-3p 5′-uuucccuuuccauccuggcag-3′
1593 hsa-miR-1285-3p 5′-ucugggcaacaaagugagaccu-3′
1693hsa-miR-548an 5′-aaaaggcauugugguuuuug-3′
1793hsa-miR-3686 5′-aucuguaagagaaaguaaauga-3′
1893hsa-miR-651-3p 5′-aaaggaaaguguauccuaaaag-3′
1993 hsa-miR-5189-5p 5′-ucugggcacaggcggauggacagg-3′
2092hsa-let-7a-3p 5′-cuauacaaucuacugucuuuc-3′
2192hsa-miR-98-3p 5′-cuauacaacuuacuacuuuccc-3′
2292hsa-miR-6860 5′-acugggcagggcuguggugagu-3′
2392hsa-miR-1-3p 5′-uggaauguaaagaaguauguau-3′
2492hsa-miR-612 5′-gcugggcagggcuucugagcuccuu-3′
2592 hsa-miR-3187-5p 5′-ccugggcagcguguggcugaagg-3′
2692hsa-let-7b-3p 5′-cuauacaaccuacugccuuccc-3′
2792hsa-miR-206 5′-uggaauguaaggaagugugugg-3′
2892 hsa-miR-5004-5p 5′-ugaggacagggcaaauucacga-3′
2992 hsa-let-7f-1-3p 5′-cuauacaaucuauugccuuccc-3′
3091hsa-miR-510-3p 5′-auugaaaccucuaagagugga-3′
3190hsa-miR-320d 5′-aaaagcuggguugagagga-3′
3290hsa-miR-613 5′-aggaauguuccuucuuugcc-3′
3390hsa-miR-4429 5′-aaaagcugggcugagaggcg-3′
3490hsa-miR-320c 5′-aaaagcuggguugagagggu-3′
3590 hsa-miR-6806-3p 5′-ugaagcucugacauuccugcag-3′
3690 hsa-miR-3928-5p 5′-ugaagcucuaagguuccgccugc-3′
3790 hsa-miR-320a-3p 5′-aaaagcuggguugagagggcga-3′
3890hsa-miR-338-3p 5′-uccagcaucagugauuuuguug-3′
3990hsa-miR-9-5p 5′-ucuuugguuaucuagcuguauga-3′
4090hsa-miR-1322 5′-gaugaugcugcugaugcug-3′
4190hsa-miR-320b 5′-aaaagcuggguugagagggcaa-3′
4289hsa-miR-150-3p 5′-cugguacaggccugggggacag-3′
4389hsa-miR-4261 5′-aggaaacagggaccca-3′
4488 hsa-miR-6733-3p 5′-ucagugucuggauuuccuag-3′
4588hsa-miR-5701 5′-uuauugucacguucugauu-3′
4687hsa-miR-3920 5′-acugauuaucuuaacucucuga-3′
4787 hsa-miR-147b-5p 5′-uggaaacauuucugcacaaacu-3′
4887 hsa-miR-4724-5p 5′-aacugaaccaggagugagcuucg-3′
4987hsa-miR-4251 5′-ccugagaaaagggccaa-3′
5086hsa-miR-137-3p 5′-uuauugcuuaagaauacgcguag-3′
5186 hsa-miR-4789-5p 5′-guauacaccugauauguguaug-3′
5286hsa-miR-570-3p 5′-cgaaaacagcaauuaccuuugc-3′
5385hsa-miR-4801 5′-uacacaagaaaaccaaggcuca-3′
5484hsa-miR-24-3p 5′-uggcucaguucagcaggaacag-3′
5584hsa-miR-587 5′-uuuccauaggugaugagucac-3′
5683hsa-miR-4324 5′-cccugagacccuaaccuuaa-3′
5783 hsa-miR-181a-2-3p 5′-accacugaccguugacuguacc-3′
5882hsa-miR-186-5p 5′-caaagaauucuccuuuugggcu-3′
5982 hsa-let-7f-2-3p 5′-cuauacagucuacugucuuucc-3′
6082 hsa-miR-6843-3p 5′-auggucuccuguucucugcag-3′
6182 hsa-miR-1185-1-3p 5′-auauacagggggagacucuuau-3′
6282 hsa-miR-6848-3p 5′-guggucucuuggcccccag-3′
6382 hsa-miR-6853-3p 5′-uguucauuggaacccugcgcag-3′
6482 hsa-miR-1185-2-3p 5′-auauacagggggagacucucau-3′
6581 hsa-miR-6736-3p 5′-ucagcuccucucuacccacag-3′
6681hsa-miR-4277 5′-gcaguucugagcacaguacac-3′
6781hsa-miR-3662 5′-gaaaaugaugaguagugacugaug-3′
6881hsa-miR-1289 5′-uggaguccaggaaucugcauuuu-3′
6981hsa-miR-1825 5′-uccagugcccuccucucc-3′

Table IV

List of miRNAs that indicate strong binding potential against ADAM metallopeptidase domain 17.

Table IV

List of miRNAs that indicate strong binding potential against ADAM metallopeptidase domain 17.

Target rankTarget scoremiRNA namemiRNA sequence
199hsa-miR-3163 5′-uauaaaaugagggcaguaagac-3′
297 hsa-miR-548ah-3p 5′-caaaaacugcaguuacuuuugc-3′
397 hsa-miR-548am-3p 5′-caaaaacugcaguuacuuuugu-3′
497 hsa-miR-548x-3p 5′-uaaaaacugcaauuacuuuc-3′
597hsa-miR-4465 5′-cucaaguagucugaccagggga-3′
697 hsa-miR-548aj-3p 5′-uaaaaacugcaauuacuuuua-3′
796 hsa-miR-548ae-3p 5′-caaaaacugcaauuacuuuca-3′
896 hsa-miR-548aq-3p 5′-caaaaacugcaauuacuuuugc-3′
996hsa-miR-1297 5′-uucaaguaauucaggug-3′
1096hsa-miR-26a-5p 5′-uucaaguaauccaggauaggcu-3′
1196hsa-miR-26b-5p 5′-uucaaguaauucaggauaggu-3′
1296 hsa-miR-548j-3p 5′-caaaaacugcauuacuuuugc-3′
1389hsa-miR-507 5′-uuuugcaccuuuuggagugaa-3′
1488hsa-miR-5697 5′-ucaaguaguuucaugauaaagg-3′
1588 hsa-miR-4762-5p 5′-ccaaaucuugaucagaagccu-3′
1688hsa-miR-4719 5′-ucacaaaucuauaauaugcagg-3′
1788 hsa-miR-5197-5p 5′-caauggcacaaacucauucuuga-3′
1888hsa-miR-4686 5′-uaucugcugggcuuucugguguu-3′
1987hsa-miR-95-5p 5′-ucaauaaaugucuguugaauu-3′
2087hsa-miR-8064 5′-agcacacugagcgagcggac-3′
2186 hsa-miR-7109-3p 5′-caagccucuccugcccuuccag-3′
2286 hsa-miR-513a-5p 5′-uucacagggaggugucau-3′
2384 hsa-miR-5195-3p 5′-auccaguucucugagggggcu-3′
2484hsa-miR-557 5′-guuugcacgggugggccuugucu-3′
2584hsa-miR-145-5p 5′-guccaguuuucccaggaaucccu-3′
2684hsa-miR-12136 5′-gaaaaagucauggaggcc-3′
2784hsa-miR-6126 5′-gugaaggcccggcggaga-3′
2883 hsa-miR-4799-5p 5′-aucuaaaugcagcaugccaguc-3′
2983 hsa-miR-148a-3p 5′-ucagugcacuacagaacuuugu-3′
3083hsa-miR-152-3p 5′-ucagugcaugacagaacuugg-3′
3183hsa-miR-4464 5′-aagguuuggauagaugcaaua-3′
3283hsa-miR-4748 5′-gagguuuggggaggauuugcu-3′
3383hsa-miR-605-3p 5′-agaaggcacuaugagauuuaga-3′
3483 hsa-miR-148b-3p 5′-ucagugcaucacagaacuuugu-3′
3583hsa-miR-767-5p 5′-ugcaccaugguugucugagcaug-3′
3681hsa-miR-548l 5′-aaaaguauuugcggguuuuguc-3′

Table V

List of miRNAs that indicate strong binding potential against hepatitis A virus cellular receptor 1 (HAVCR1, KIM1).

Table V

List of miRNAs that indicate strong binding potential against hepatitis A virus cellular receptor 1 (HAVCR1, KIM1).

Target rankTarget scoremiRNA namemiRNA sequence
190hsa-miR-4255 5′-caguguucagagaugga-3′
287 hsa-miR-4738-3p 5′-ugaaacuggagcgccuggagga-3′
386 hsa-miR-3117-3p 5′-auaggacucauauagugccag-3′
485hsa-miR-3171 5′-agauguauggaaucuguauauauc-3′
583hsa-miR-383-5p 5′-agaucagaaggugauuguggcu-3′

Table VI

List of miRNAs that indicate strong binding potential against heat shock protein family A (Hsp70) member 5 (HSPA5 or GRP78).

Table VI

List of miRNAs that indicate strong binding potential against heat shock protein family A (Hsp70) member 5 (HSPA5 or GRP78).

Target rankTarget scoremiRNA namemiRNA sequence
199 hsa-miR-3121-3p 5′-uaaauagaguaggcaaaggaca-3′
297hsa-miR-5688 5′-uaacaaacaccuguaaaacagc-3′
396hsa-miR-635 5′-acuugggcacugaaacaaugucc-3′
496 hsa-miR-6774-5p 5′-acuugggcaggagggacccuguaug-3′
595hsa-miR-495-3p 5′-aaacaaacauggugcacuucuu-3′
693hsa-miR-338-5p 5′-aacaauauccuggugcugagug-3′
791hsa-miR-7-2-3p 5′-caacaaaucccagucuaccuaa-3′
891hsa-miR-7-1-3p 5′-caacaaaucacagucugccaua-3′
990hsa-miR-30d-3p 5′-cuuucagucagauguuugcugc-3′
1089 hsa-miR-199a-5p 5′-cccaguguucagacuaccuguuc-3′
1189 hsa-miR-199b-5p 5′-cccaguguuuagacuaucuguuc-3′
1289 hsa-miR-4699-3p 5′-aauuuacucugcaaucuucucc-3′
1388 hsa-miR-7854-3p 5′-ugaggugaccgcagaugggaa-3′
1487 hsa-miR-4650-3p 5′-agguagaaugaggccugacau-3′
1585hsa-miR-222-5p 5′-cucaguagccaguguagauccu-3′
1684 hsa-miR-16-1-3p 5′-ccaguauuaacugugcugcuga-3′
1784hsa-miR-30e-3p 5′-cuuucagucggauguuuacagc-3′
1884 hsa-miR-148a-5p 5′-aaaguucugagacacuccgacu-3′
1984hsa-miR-30a-3p 5′-cuuucagucggauguuugcagc-3′
2084hsa-miR-4451 5′-ugguagagcugaggaca-3′
2184 hsa-miR-7162-3p 5′-ucugagguggaacagcagc-3′
2283 hsa-miR-6777-3p 5′-uccacucuccuggcccccag-3′
2382hsa-miR-770-5p 5′-uccaguaccacgugucagggcca-3′
2482 hsa-miR-4712-5p 5′-uccaguacaggucucucauuuc-3′
2581 hsa-miR-3606-3p 5′-aaaauuucuuucacuacuuag-3′
2681 hsa-miR-513c-3p 5′-uaaauuucaccuuucugagaaga-3′
2781 hsa-miR-513a-3p 5′-uaaauuucaccuuucugagaagg-3′

Table VII

One miRNA which exhibits strong binding potential against basigin (BSG, CD147).

Table VII

One miRNA which exhibits strong binding potential against basigin (BSG, CD147).

Target rankTarget scoremiRNA namemiRNA sequence
193 hsa-miR-1252-5p 5′-agaaggaaauugaauucauuua-3′

Table VIII

List of miRNAs that indicate strong binding potential against Toll-like receptor 4.

Table VIII

List of miRNAs that indicate strong binding potential against Toll-like receptor 4.

Target rankTarget scoremiRNA namemiRNA sequence
193hsa-miR-448 5′-uugcauauguaggaugucccau-3′
291hsa-miR-3924 5′-auauguauaugugacugcuacu-3′
389hsa-miR-627-3p 5′-ucuuuucuuugagacucacu-3′
488hsa-miR-338-5p 5′-aacaauauccuggugcugagug-3′
588hsa-miR-4272 5′-cauucaacuagugauugu-3′
687 hsa-miR-10397-5p 5′-uccuugaccugaugcuguaggg-3′
787 hsa-miR-5583-3p 5′-gaauauggguauauuaguuugg-3′
885 hsa-miR-4760-3p 5′-aaauucauguucaaucuaaacc-3′
985 hsa-miR-4652-3p 5′-guucuguuaacccauccccuca-3′
1084hsa-miR-1243 5′-aacuggaucaauuauaggagug-3′
1181hsa-miR-587 5′-uuuccauaggugaugagucac-3′
1281hsa-miR-4678 5′-aagguauuguucagacuuauga-3′

Table IX

List of miRNAs that indicate strong binding potential against metabotropic glutamate receptor subtype 2.

Table IX

List of miRNAs that indicate strong binding potential against metabotropic glutamate receptor subtype 2.

Target rankTarget scoremiRNA namemiRNA sequence
186hsa-miR-4492 5′-ggggcugggcgcgcgcc-3′
284hsa-miR-555 5′-aggguaagcugaaccucugau-3′
383 hsa-miR-4436b-3p 5′-cagggcaggaagaaguggacaa-3′
483hsa-miR-497-3p 5′-caaaccacacugugguguuaga-3′
582hsa-miR-4498 5′-ugggcuggcagggcaagugcug-3′

Further analysis using Venn diagrams (Fig. 2), revealed that there were three common miRNAs amongst NRP1 and ADAM17, namely hsa-miR-148a-3p, hsa-miR-152-3p and hsa-miR-148b-3p. One common miRNA, i.e., hsa-miR-587, was also shared between TLR4 and NRP1, and another miRNA (hsa-miR-338-5p) between TLR4 and GRP78. Of note, hsa-miR-148a-3p was also shared with ACE2, as previously reported (71). Moreover, hsa-miR-587 was recently identified as a top miRNA regulating ACE2 networks; it was thus concluded that these miRNAs can be used in the personalized diagnosis of patients with COVID-19 (74).

Moreover, making use of the miRNA atlas (ccb-web. cs.uni-saarland.de/tissueatlas), it was evident that the miRNAs listed in Tables I-VIII have a wide distribution in the human body and co-localise with SARS-CoV-2 cell entry mediators (Fig. 3). For example, hsa-miR-152-3p is primarily expressed in the epididymis, colon, artery, oesophagus, thyroid, whereas hsa-miR-338-5p is mostly present in the kidneys, brain and spleen. This co-localisation provides a higher order of complexity on the mechanisms through which these mediators may be regulated by miRNAs. Future studies are required to evaluate their therapeutic or diagnostic potential.

6. Conclusion

The pathophysiology of COVID-19 is mainly dependent on the underlying mechanisms that mediate the entry of SARS-CoV-2 into the host cells of the various human tissues/organs. Based on the growing body of evidence regarding these mechanisms, it is evident that there is an array of factors/mediators which play crucial roles in these processes, often in synergistic/complimentary ways. The complexity of these viral-host interactions appears to be further increased when the co-localization of SARS-CoV-2 host cell entry mediators in different human tissues/organs is considered, as well as when considering the potential modulatory effects that several miRNAs can have on the local expression of these mediators. Accordingly, intensive research is still warranted in this field in order to precisely characterise these underlying mechanisms, with the ultimate objective of aiding in the development of novel treatments, which will act by blocking the entry of SARS-CoV-2 into susceptible human host cells.

Availability of data and materials

The data used and/or analysed during the current study are available from the corresponding author on reasonable request.

Authors' contributions

PK, EK, SS, HSR and IK contributed to the conceptualization, methodology, data curation, investigation, visualization, drafting, editing and reviewing of the manuscript. HSR, DAS and SS contributed to the literature search, and the drafting and critical revision of the manuscript. All authors have read and approved the final manuscript. SS and EK confirm the authenticity of all the raw data.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

DAS is the Editor-in-Chief for the journal, but had no personal involvement in the reviewing process, or any influence in terms of adjudicating on the final decision, for this article. The other authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank Dr Evangelos Kolettas, Laboratory of Biology, School of Medicine, University of Ioannina, and Biomedical Research Division, Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology, Ioannina, Greece, for critically reading and commenting on the manuscript.

Funding

No funding was received.

Abbreviations:

COVID-19

coronavirus disease 2019

SARS-CoV2

severe acute respiratory syndrome coronavirus 2

ACE2

angiotensin converting enzyme 2

NPR1

neuropilin-1

TMPRSS2 and 4

transmembrane protease serine 2 and 4

ADAM17

ADAM metallopeptidase domain 17 (also termed tumour necrosis factor-αconvertase)

KIM1

kidney injury molecule-1

TLR4

Toll-like receptor 4

GRP78

glucose-regulated protein 78

miRNAs

microRNAs

References

1 

Osuchowski MF, Winkler MS, Skirecki T, Cajander S, Shankar-Hari M, Lachmann G, Monneret G, Venet F, Bauer M, Brunkhorst FM, et al: The COVID-19 puzzle: Deciphering pathophysiology and phenotypes of a new disease entity. Lancet Respir Med. 9:622–642. 2021. View Article : Google Scholar : PubMed/NCBI

2 

Singh S, Pandey R, Tomar S, Varshney R, Sharma D and Gangenahalli G: A brief molecular insight of COVID-19: Epidemiology, clinical manifestation, molecular mechanism, cellular tropism and immuno-pathogenesis. Mol Cell Biochem. 476:3987–4002. 2021. View Article : Google Scholar : PubMed/NCBI

3 

Murgolo N, Therien AG, Howell B, Klein D, Koeplinger K, Lieberman LA, Adam GC, Flynn J, McKenna P, Swaminathan G, et al: SARS-CoV-2 tropism, entry, replication, and propagation: Considerations for drug discovery and development. PLoS Pathog. 17:e10092252021. View Article : Google Scholar : PubMed/NCBI

4 

Gao S and Zhang L: ACE2 partially dictates the host range and tropism of SARS-CoV-2. Comput Struct Biotechnol J. 18:4040–4047. 2020. View Article : Google Scholar : PubMed/NCBI

5 

Hoffmann M, Kleine-Weber H, Schroeder S, Krüger N, Herrler T, Erichsen S, Schiergens TS, Herrler G, Wu NH, Nitsche A, et al: SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell. 181:271–280.e8. 2020. View Article : Google Scholar : PubMed/NCBI

6 

Cai L, Guo X, Cao Y, Ying P, Hong L, Zhang Y, Yi G and Fu M: Determining available strategies for prevention and therapy: Exploring COVID-19 from the perspective of ACE2 (Review). Int J Mol Med. 47:432021. View Article : Google Scholar :

7 

Glowacka I, Bertram S, Müller MA, Allen P, Soilleux E, Pfefferle S, Steffen I, Tsegaye TS, He Y, Gnirss K, et al: Evidence that TMPRSS2 activates the severe acute respiratory syndrome coronavirus spike protein for membrane fusion and reduces viral control by the humoral immune response. J Virol. 85:4122–4134. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Piva F, Sabanovic B, Cecati M and Giulietti M: Expression and Co-expression analyses of TMPRSS2, a key element in COVID-19. Eur J Clin Microbiol Infect Dis. 40:451–455. 2021. View Article : Google Scholar

9 

Hou Y, Zhao J, Martin W, Kallianpur A, Chung MK, Jehi L, Sharifi N, Erzurum S, Eng C and Cheng F: New insights into genetic susceptibility of COVID-19: An ACE2 and TMPRSS2 polymorphism analysis. BMC Med. 18:2162020. View Article : Google Scholar : PubMed/NCBI

10 

Zipeto D, Palmeira JDF, Argañaraz GA and Argañaraz ER: ACE2/ADAM17/TMPRSS2 interplay may be the main risk factor for COVID-19. Front Immunol. 11:5767452020. View Article : Google Scholar : PubMed/NCBI

11 

Shang J, Wan Y, Luo C, Ye G, Geng Q, Auerbach A and Li F: Cell entry mechanisms of SARS-CoV-2. Proc Natl Acad Sci USA. 117:11727–11734. 2020. View Article : Google Scholar : PubMed/NCBI

12 

Magrone T, Magrone M and Jirillo E: Focus on receptors for coronaviruses with special reference to Angiotensin-converting Enzyme 2 as a potential drug Target-A perspective. Endocr Metab Immune Disord Drug Targets. 20:807–811. 2020. View Article : Google Scholar

13 

Jia H, Neptune E and Cui H: Targeting ACE2 for COVID-19 therapy: Opportunities and challenges. Am J Respir Cell Mol Biol. 64:416–425. 2021. View Article : Google Scholar :

14 

Imai Y, Kuba K, Rao S, Huan Y, Guo F, Guan B, Yang P, Sarao R, Wada T, Leong-Poi H, et al: Angiotensin-converting enzyme 2 protects from severe acute lung failure. Nature. 436:112–116. 2005. View Article : Google Scholar : PubMed/NCBI

15 

Schuler BA, Habermann AC, Plosa EJ, Taylor CJ, Jetter C, Negretti NM, Kapp ME, Benjamin JT, Gulleman P, Nichols DS, et al: Age-determined expression of priming protease TMPRSS2 and localization of SARS-CoV-2 in lung epithelium. J Clin Invest. 131:e1407662021. View Article : Google Scholar :

16 

Musso N, Falzone L, Stracquadanio S, Bongiorno D, Salerno M, Esposito M, Sessa F, Libra M, Stefani S and Pomara C: Post-mortem detection of SARS-CoV-2 RNA in Long-buried lung samples. Diagnostics (Basel). 11:11582021. View Article : Google Scholar

17 

Deinhardt-Emmer S, Wittschieber D, Sanft J, Kleemann S, Elschner S, Haupt KF, Vau V, Häring C, Rödel J, Henke A, et al: Early postmortem mapping of SARS-CoV-2 RNA in patients with COVID-19 and the correlation with tissue damage. Elife. 10:e603612021. View Article : Google Scholar : PubMed/NCBI

18 

Yao XH, Luo T, Shi Y, He ZC, Tang R, Zhang PP, Cai J, Zhou XD, Jiang DP, Fei XC, et al: A cohort autopsy study defines COVID-19 systemic pathogenesis. Cell Res. 31:836–846. 2021. View Article : Google Scholar : PubMed/NCBI

19 

Zang R, Gomez Castro MF, McCune BT, Zeng Q, Rothlauf PW, Sonnek NM, Liu Z, Brulois KF, Wang X, Greenberg HB, et al: TMPRSS2 and TMPRSS4 promote SARS-CoV-2 infection of human small intestinal enterocytes. Sci Immunol. 5:eabc35822020. View Article : Google Scholar : PubMed/NCBI

20 

Kyrou I, Randeva HS, Spandidos DA and Karteris E: Not only ACE2-the quest for additional host cell mediators of SARS-CoV-2 infection: Neuropilin-1 (NRP1) as a novel SARS-CoV-2 host cell entry mediator implicated in COVID-19. Signal Transduct Target Ther. 6:212021. View Article : Google Scholar : PubMed/NCBI

21 

Katopodis P, Kerslake R, Davies J, Randeva HS, Chatha K, Hall M, Spandidos DA, Anikin V, Polychronis A, Robertus JL, et al: COVID-19 and SARS-CoV-2 host cell entry mediators: Expression profiling of TMRSS4 in health and disease. Int J Mol Med. 47:642021. View Article : Google Scholar :

22 

Cuervo NZ and Grandvaux N: ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. Elife. 9:e613902020. View Article : Google Scholar

23 

Davies J, Randeva HS, Chatha K, Hall M, Spandidos DA, Karteris E and Kyrou I: Neuropilin-1 as a new potential SARS-CoV-2 infection mediator implicated in the neurologic features and central nervous system involvement of COVID-19. Mol Med Rep. 22:4221–4226. 2020.PubMed/NCBI

24 

Daly JL, Simonetti B, Antón-Plágaro C, Williamson MK, Shoemark DK, Simón-Gracia L, Klein K, Bauer M, Hollandi R, Greber UF, et al: Neuropilin-1 is a host factor for SARS-CoV-2 infection. Science. 370:861–865. 2020. View Article : Google Scholar : PubMed/NCBI

25 

Cantuti-Castelvetri L, Ojha R, Pedro LD, Djannatian M, Franz J, Kuivanen S, van der Meer F, Kallio K, Kaya T, Anastasina M, et al: Neuropilin-1 facilitates SARS-CoV-2 cell entry and infectivity. Science. 370:856–860. 2020. View Article : Google Scholar : PubMed/NCBI

26 

Kielian M: Enhancing host cell infection by SARS-CoV-2. Science. 370:765–766. 2020. View Article : Google Scholar : PubMed/NCBI

27 

Cantuti-Castelvetri L, Ojha R, Pedro LD, Djannatian M, Franz J, Kuivanen S, Kallio K, Kaya T, Anastasina M, Smura T, et al: Neuropilin-1 facilitates SARS-CoV-2 cell entry and provides a possible pathway into the central nervous system. bioRxiv. Jul 15–2020.Epub ahead of print. View Article : Google Scholar

28 

Elfiky AA: SARS-CoV-2 Spike-heat shock protein A5 (GRP78) recognition may be related to the immersed human coronaviruses. Front Pharmacol. 11:5774672020. View Article : Google Scholar : PubMed/NCBI

29 

Ibrahim IM, Abdelmalek DH, Elshahat ME and Elfiky AA: COVID-19 spike-host cell receptor GRP78 binding site prediction. J Infect. 80:554–562. 2020. View Article : Google Scholar : PubMed/NCBI

30 

Wang K, Chen W, Zhang Z, Deng Y, Lian JQ, Du P, Wei D, Zhang Y, Sun XX, Gong L, et al: CD147-spike protein is a novel route for SARS-CoV-2 infection to host cells. Signal Transduct Target Ther. 5:2832020. View Article : Google Scholar : PubMed/NCBI

31 

Shilts J, Crozier TWM, Greenwood EJD, Lehner PJ and Wright GJ: No evidence for basigin/CD147 as a direct SARS-CoV-2 spike binding receptor. Sci Rep. 11:4132021. View Article : Google Scholar : PubMed/NCBI

32 

Minami T, Iwata Y and Wada T: Renal complications in coronavirus disease 2019: A systematic review. Inflamm Regen. 40:312020. View Article : Google Scholar : PubMed/NCBI

33 

Yang C, Zhang Y, Zeng X, Chen H, Chen Y, Yang D, Shen Z, Wang X, Liu X, Xiong M, et al: Kidney injury molecule-1 is a potential receptor for SARS-CoV-2. J Mol Cell Biol. 13:185–196. 2021. View Article : Google Scholar : PubMed/NCBI

34 

Wan C and Zhang C: Kidney injury molecule-1: A novel entry factor for SARS-CoV-2. J Mol Cell Biol. 13:159–160. 2021. View Article : Google Scholar : PubMed/NCBI

35 

Bu Z, Wang J, Yang G, Wang X, Wen Z, Shuai L, Luo J, Wang C, Sun Z, Liu R, et al: Metabotropic glutamate receptor subtype 2 is a receptor of SARS-CoV-2. Res Sq. April 21–2021.Epub ahead of print. View Article : Google Scholar

36 

Cui C, Huang C, Zhou W, Ji X, Zhang F, Wang L, Zhou Y and Cui Q: AGTR2, one possible novel key gene for the entry of SARS-CoV-2 into human cells. IEEE/ACM Trans Comput Biol Bioinforma. 18:1230–1233. 2021. View Article : Google Scholar

37 

Zhang Q, Chen CZ, Swaroop M, Xu M, Wang L, Lee J, Wang AQ, Pradhan M, Hagen N, Chen L, et al: Heparan sulfate assists SARS-CoV-2 in cell entry and can be targeted by approved drugs in vitro. Cell Discov. 6:802020. View Article : Google Scholar : PubMed/NCBI

38 

Lambert DW, Yarski M, Warner FJ, Thornhill P, Parkin ET, Smith AI, Hooper NM and Turner AJ: Tumor necrosis factor-alpha convertase (ADAM17) mediates regulated ectodomain shedding of the severe-acute respiratory syndrome-coronavirus (SARS-CoV) receptor, angiotensin-converting enzyme-2 (ACE2). J Biol Chem. 280:30113–30119. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Palau V, Riera M and Soler MJ: ADAM17 inhibition may exert a protective effect on COVID-19. Nephrol Dial Transplant. 35:1071–1072. 2020. View Article : Google Scholar : PubMed/NCBI

40 

Heurich A, Hofmann-Winkler H, Gierer S, Liepold T, Jahn O and Pöhlmann S: TMPRSS2 and ADAM17 cleave ACE2 differentially and only proteolysis by TMPRSS2 augments entry driven by the severe acute respiratory syndrome coronavirus spike protein. J Virol. 88:1293–1307. 2014. View Article : Google Scholar :

41 

Kumar J, Murugaiah V, Sotiriadis G, Kaur A, Jeyaneethi J, Sturniolo I, Alhamlan FS, Chatterjee J, Hall M, Kishore U and Karteris E: Surfactant Protein D as a potential biomarker and therapeutic target in ovarian cancer. Front Oncol. 9:5422019. View Article : Google Scholar : PubMed/NCBI

42 

Hsieh MH, Beirag N, Murugaiah V, Chou YC, Kuo WS, Kao HF, Madan T, Kishore U and Wang JY: Human surfactant Protein D binds spike protein and acts as an entry inhibitor of SARS-CoV-2 pseudotyped viral particles. Front Immunol. 12:6413602021. View Article : Google Scholar : PubMed/NCBI

43 

Madan T, Biswas B, Varghese PM, Subedi R, Pandit H, Idicula-Thomas S, Kundu I, Rooge S, Agarwal R, Tripathi DM, et al: A recombinant fragment of human surfactant Protein D binds spike protein and inhibits infectivity and replication of SARS-CoV-2 in clinical samples. Am J Respir Cell Mol Biol. 65:41–53. 2021. View Article : Google Scholar : PubMed/NCBI

44 

Tong M, Xiong Y, Zhu C, Xu H, Zheng Q, Jiang Y, Zou L, Xiao X, Chen F, Yan X, et al: Serum surfactant protein D in COVID-19 is elevated and correlated with disease severity. BMC Infect Dis. 21:7372021. View Article : Google Scholar : PubMed/NCBI

45 

Aboudounya MM and Heads RJ: COVID-19 and toll-like receptor 4 (TLR4): SARS-CoV-2 may bind and activate TLR4 to increase ACE2 expression, facilitating entry and causing hyper-inflammation. Mediators Inflamm. 2021:88743392021. View Article : Google Scholar

46 

Gadanec LK, McSweeney KR, Qaradakhi T, Ali B, Zulli A and Apostolopoulos V: Can SARS-CoV-2 virus use multiple receptors to enter host cells? Int J Mol Sci. 22:9922021. View Article : Google Scholar : PubMed/NCBI

47 

Zhao Y, Kuang M, Li J, Zhu L, Jia Z, Guo X, Hu Y, Kong J, Yin H, Wang X and You F: SARS-CoV-2 spike protein interacts with and activates TLR41. Cell Res. 31:818–820. 2021. View Article : Google Scholar : PubMed/NCBI

48 

Zamorano Cuervo N and Grandvaux N: ACE2: Evidence of role as entry receptor for SARS-CoV-2 and implications in comorbidities. Elife. 9:e613902020. View Article : Google Scholar : PubMed/NCBI

49 

Lan J, Ge J, Yu J, Shan S, Zhou H, Fan S, Zhang Q, Shi X, Wang Q, Zhang L and Wang X: Structure of the SARS-CoV-2 spike receptor-binding domain bound to the ACE2 receptor. Nature. 581:215–220. 2020. View Article : Google Scholar : PubMed/NCBI

50 

Shrimp JH, Kales SC, Sanderson PE, Simeonov A, Shen M and Hall MD: An enzymatic TMPRSS2 assay for assessment of clinical candidates and discovery of inhibitors as potential treatment of COVID-19. ACS Pharmacol Transl Sci. 3:997–1007. 2020. View Article : Google Scholar : PubMed/NCBI

51 

Lee JJ, Kopetz S, Vilar E, Shen JP, Chen K and Maitra A: Relative abundance of SARS-CoV-2 entry genes in the enterocytes of the lower gastrointestinal tract. Genes (Basel). 11:6452020. View Article : Google Scholar :

52 

Choudhury A and Mukherjee S: In silico studies on the comparative characterization of the interactions of SARS-CoV-2 spike glycoprotein with ACE-2 receptor homologs and human TLRs. J Med Virol. 92:2105–2113. 2020. View Article : Google Scholar : PubMed/NCBI

53 

Dexheimer PJ and Cochella L: MicroRNAs: From mechanism to organism. Front Cell Dev Biol. 8:4092020. View Article : Google Scholar : PubMed/NCBI

54 

Peng Y and Croce CM: The role of MicroRNAs in human cancer. Signal Transduct Target Ther. 1:150042016. View Article : Google Scholar : PubMed/NCBI

55 

Chen K and Rajewsky N: The evolution of gene regulation by transcription factors and microRNAs. Nat Rev Genet. 8:93–103. 2007. View Article : Google Scholar : PubMed/NCBI

56 

Correia de Sousa M, Gjorgjieva M, Dolicka D, Sobolewski C and Foti M: Deciphering miRNAs' Action through miRNA Editing. Int J Mol Sci. 20:62492019. View Article : Google Scholar

57 

Cai Y, Yu X, Hu S and Yu J: A brief review on the mechanisms of miRNA regulation. Genomics Proteomics Bioinformatics. 7:147–154. 2009. View Article : Google Scholar

58 

Liu Z, Wang J, Ge Y, Xu Y, Guo M, Mi K, Xu R, Pei Y, Zhang Q, Luan X, et al: SARS-CoV-2 encoded microRNAs are involved in the process of virus infection and host immune response. J Biomed Res. 35:216–227. 2021. View Article : Google Scholar : PubMed/NCBI

59 

Chauhan N, Jaggi M, Chauhan SC and Yallapu MM: COVID-19: Fighting the invisible enemy with microRNAs. Expert Rev Anti Infect Ther. 19:137–145. 2021. View Article : Google Scholar

60 

Fani M, Zandi M, Ebrahimi S, Soltani S and Abbasi S: The role of miRNAs in COVID-19 disease. Future Virol. 16:301–306. 2021. View Article : Google Scholar

61 

Bugnon LA, Raad J, Merino GA, Yones C, Ariel F, Milone DH and Stegmayer G: Deep Learning for the discovery of new pre-miRNAs: Helping the fight against COVID-19. Mach Learn with Appl. 6:1001502021. View Article : Google Scholar

62 

Abedi F, Rezaee R, Hayes AW, Nasiripour S and Karimi G: MicroRNAs and SARS-CoV-2 life cycle, pathogenesis, and mutations: Biomarkers or therapeutic agents? Cell Cycle. 20:143–153. 2021. View Article : Google Scholar : PubMed/NCBI

63 

Haddad H and Al-Zyoud W: miRNA target prediction might explain the reduced transmission of SARS-CoV-2 in Jordan, Middle East. Noncoding RNA Res. 5:135–143. 2020. View Article : Google Scholar : PubMed/NCBI

64 

Sardar R, Satish D, Birla S and Gupta D: Integrative analyses of SARS-CoV-2 genomes from different geographical locations reveal unique features potentially consequential to host-virus interaction, pathogenesis and clues for novel therapies. Heliyon. 6:e046582020. View Article : Google Scholar : PubMed/NCBI

65 

Matarese A, Gambardella J, Sardu C and Santulli G: miR-98 regulates TMPRSS2 expression in human endothelial cells: Key implications for COVID-19. Biomedicines. 8:4622020. View Article : Google Scholar :

66 

Nersisyan S, Shkurnikov M, Turchinovich A, Knyazev E and Tonevitsky A: Integrative analysis of miRNA and mRNA sequencing data reveals potential regulatory mechanisms of ACE2 and TMPRSS2. PLoS One. 15:e02359872020. View Article : Google Scholar : PubMed/NCBI

67 

Lu D, Chatterjee S, Xiao K, Riedel I, Wang Y, Foo R, Bär C and Thum T: MicroRNAs targeting the SARS-CoV-2 entry receptor ACE2 in cardiomyocytes. J Mol Cell Cardiol. 148:46–49. 2020. View Article : Google Scholar : PubMed/NCBI

68 

Khan MA, Sany MRU, Islam MS and Islam ABMMK: Epigenetic regulator miRNA pattern differences among SARS-CoV, SARS-CoV-2, and SARS-CoV-2 World-Wide isolates delineated the mystery behind the epic pathogenicity and distinct clinical characteristics of pandemic COVID-19. Front Genet. 11:7652020. View Article : Google Scholar : PubMed/NCBI

69 

Paul S, Bravo Vázquez LA, Reyes-Pérez PR, Estrada-Meza C, Aponte Alburquerque RA, Pathak S, Banerjee A, Bandyopadhyay A, Chakraborty S and Srivastava A: The role of microRNAs in solving COVID-19 puzzle from infection to therapeutics: A mini-review. Virus Res. 308:1986312021. View Article : Google Scholar : PubMed/NCBI

70 

Dash S, Dash C and Pandhare J: Therapeutic significance of microRNA-mediated regulation of PARP-1 in SARS-CoV-2 infection. Noncoding RNA. 7:602021.PubMed/NCBI

71 

Mukhopadhyay D and Mussa BM: Identification of novel hypothalamic MicroRNAs as promising therapeutics for SARS-CoV-2 by regulating ACE2 and TMPRSS2 expression: An in silico analysis. Brain Sci. 10:6662020. View Article : Google Scholar :

72 

Wang X: miRDB: A microRNA target prediction and functional annotation database with a wiki interface. RNA. 14:1012–1017. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Wong N and Wang X: miRDB: An online resource for microRNA target prediction and functional annotations. Nucleic Acids Res. 43:D146–D152. 2015. View Article : Google Scholar :

74 

Wicik Z, Eyileten C, Jakubik D, Simões SN, Martins DC Jr, Pavão R, Siller-Matula JM and Postula M: ACE2 interaction networks in COVID-19: A physiological framework for prediction of outcome in patients with cardiovascular risk factors. J Clin Med. 9:37432020. View Article : Google Scholar :

Related Articles

Journal Cover

February-2022
Volume 49 Issue 2

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Katopodis P, Randeva HS, Spandidos DA, Saravi S, Kyrou I and Karteris E: Host cell entry mediators implicated in the cellular tropism of SARS‑CoV‑2, the pathophysiology of COVID‑19 and the identification of microRNAs that can modulate the expression of these mediators (Review). Int J Mol Med 49: 20, 2022
APA
Katopodis, P., Randeva, H.S., Spandidos, D.A., Saravi, S., Kyrou, I., & Karteris, E. (2022). Host cell entry mediators implicated in the cellular tropism of SARS‑CoV‑2, the pathophysiology of COVID‑19 and the identification of microRNAs that can modulate the expression of these mediators (Review). International Journal of Molecular Medicine, 49, 20. https://doi.org/10.3892/ijmm.2021.5075
MLA
Katopodis, P., Randeva, H. S., Spandidos, D. A., Saravi, S., Kyrou, I., Karteris, E."Host cell entry mediators implicated in the cellular tropism of SARS‑CoV‑2, the pathophysiology of COVID‑19 and the identification of microRNAs that can modulate the expression of these mediators (Review)". International Journal of Molecular Medicine 49.2 (2022): 20.
Chicago
Katopodis, P., Randeva, H. S., Spandidos, D. A., Saravi, S., Kyrou, I., Karteris, E."Host cell entry mediators implicated in the cellular tropism of SARS‑CoV‑2, the pathophysiology of COVID‑19 and the identification of microRNAs that can modulate the expression of these mediators (Review)". International Journal of Molecular Medicine 49, no. 2 (2022): 20. https://doi.org/10.3892/ijmm.2021.5075