Next Article in Journal
Lymphocytic Choriomeningitis Virus Alters the Expression of Male Mouse Scent Proteins
Previous Article in Journal
The Viral Capsid: A Master Key to Access the Host Nucleus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Broad Impact of Exchange Protein Directly Activated by cAMP 2 (EPAC2) on Respiratory Viral Infections

1
Department of Pediatrics, The University of Texas Medical Branch, Galveston, TX 77555-0372, USA
2
Department of Pharmacology & Toxicology, The University of Texas Medical Branch, Galveston, TX 77555-0372, USA
3
Department of Kinesiology, Rice University, Houston, TX 77005, USA
4
College of Natural Sciences, The University of Texas at Austin, Austin, TX 78712, USA
5
Sealy Center for Molecular Medicine, The University of Texas Medical Branch, Galveston, TX 77555, USA
6
The Institute of Translational Sciences, The University of Texas Medical Branch, Galveston, TX 77555, USA
7
The Institute for Human Infections and Immunity, The University of Texas Medical Branch, Galveston, TX 77555, USA
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 28 April 2021 / Revised: 27 May 2021 / Accepted: 7 June 2021 / Published: 21 June 2021
(This article belongs to the Section Human Virology and Viral Diseases)

Abstract

:
The recently discovered exchange protein directly activated by cAMP (EPAC), compared with protein kinase A (PKA), is a fairly new family of cAMP effectors. Soon after the discovery, EPAC has shown its significance in many diseases including its emerging role in infectious diseases. In a recent study, we demonstrated that EPAC, but not PKA, is a promising therapeutic target to regulate respiratory syncytial virus (RSV) replication and its associated inflammation. In mammals, there are two isoforms of EPAC—EPAC1 and EPAC2. Unlike other viruses, including Middle East respiratory syndrome coronavirus (MERS-CoV) and Ebola virus, which use EPAC1 to regulate viral replication, RSV uses EPAC2 to control its replication and associated cytokine/chemokine responses. To determine whether EPAC2 protein has a broad impact on other respiratory viral infections, we used an EPAC2-specific inhibitor, MAY0132, to examine the functions of EPAC2 in human metapneumovirus (HMPV) and adenovirus (AdV) infections. HMPV is a negative-sense single-stranded RNA virus belonging to the family Pneumoviridae, which also includes RSV, while AdV is a double-stranded DNA virus. Treatment with an EPAC1-specific inhibitor was also included to investigate the impact of EPAC1 on these two viruses. We found that the replication of HMPV, AdV, and RSV and the viral-induced immune mediators are significantly impaired by MAY0132, while an EPAC1-specific inhibitor, CE3F4, does not impact or slightly impacts, demonstrating that EPAC2 could serve as a novel common therapeutic target to control these viruses, all of which do not have effective treatment and prevention strategies.

1. Introduction

Acute lower respiratory tract infections (RTIs) have been one of the top five causes of death globally in adults and children, which is estimated to lead to nearly 4 million deaths annually [1,2]. Young children, the elderly, and immunocompromised patients are particularly susceptible to severe RTIs. It has been reported that two-thirds to three-fourths of cases of acute respiratory illness are caused by viruses [3]. More than other factors, viral-induced respiratory diseases have a significant impact on public health due to their rapid spread across the globe. For example, since the emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in December 2019 [4,5], there were about 91 million confirmed infections and 2 million deaths worldwide as of 14 January 2021 [6]. The COVID-19 pandemic has been generating health, economic, and social crises worldwide [7]. Given that multiple viruses are the causative agents of human respiratory infection and co-infection has been continuously reported, the development of a novel treatment modality that has a broad-spectrum antiviral effect is urgently needed.
Human metapneumovirus (HMPV), which belongs to the Pneumoviridae family, is a negative-sense single-stranded RNA virus. Similar to respiratory syncytial virus (RSV), it is also the leading cause of lower RTI in young children, high-risk adults, and the elderly [8,9,10]. In some regions, HMPV was reported to be only secondary to RSV infection [11]. Overall, these viruses are responsible for a significant health burden of disease in children under five years of age in terms of hospitalizations, emergency/urgent care visits, and clinic visits [12,13]. Co-infection of HMPV and RSV has been demonstrated to be associated with a higher risk of ICU admission in children under five years [14] and increased disease severity in infants [15]. In addition to these RNA viruses, double-stranded DNA adenovirus (AdV) is also a common cause of RTI in children and adults [16,17]. Adenovirus infection can cause significant morbidity and mortality in young children and immunocompromised patients [18,19,20]. Unfortunately, there are no effective antivirals or vaccines available for the prevention or treatment of these viral infections. Palivizumab, a monoclonal antibody (mAb) targeting fusion protein of RSV, was licensed in 1998 for prophylactic use in high-risk infants. However, it is not very cost effective with a limited application to high-risk infants in their first period of exposure to the RSV season [21].
Exchange protein directly activated by cyclic AMP (cAMP) (EPAC) is a downstream effector of cAMP, consisting of two isoforms, EPAC1 and EPAC2, in mammals [22,23]. Compared with protein kinase A (PKA), EPAC is a relatively new effector family. Cumulative findings have shown the important function of EPAC in various diseases including heart failure, cancer, neurological disorders, diabetes, inflammation, and bacterial and viral infections [24,25,26,27,28,29]. For this reason, several activators and inhibitors, targeting EPAC1, EPAC2, or both, have been continuously developed [30,31,32]. We have recently investigated the effect of EPAC on RSV infection and found that EPAC2 regulates RSV replication and its associated innate immune response [33]. Earlier than our findings, another group demonstrated the impact of EPAC1 on Middle East respiratory syndrome coronavirus (MERS-CoV) infection [28], with more recent publications demonstrating the impact of EPAC1 on other viral infections [34,35]. However, the significance of EPAC2 in viral infections is still limitedly reported. Overall, research on the function and significance of EPAC in virus infection is recently being explored and remains to be investigated in many other viral infections.
Herein, we aim to examine the broad-spectrum effect of EPAC on respiratory virus infection. Since both HMPV and AdV are major causes of RTI in humans, we selected HMPV and AdV as a representative RNA and DNA viruses, respectively, for the study. Using EPAC1- and EPAC2-specific inhibitors [36,37], we found that the EPAC2-specific inhibitor MAY0132 significantly suppressed HMPV and AdV replication and cytokine/chemokine induction in both immortalized and primary epithelial cells, while the EPAC1-specific inhibitor CE3F4 played a minimal role. Furthermore, EPAC2 inhibitors were highly effective in interfering with the polyinosinic–polycytidylic acid (poly I:C)- or TNF-α-induced NF-κB activation. Taken together, our results propose the potential of EPAC2 as a novel therapeutic target against multiple respiratory viral infections to control not only virus replication but also cytokine/chemokine induction.

2. Materials and Methods

2.1. Cells, Virus, and Reagents

A549 (human alveolar type II-like epithelial), HEp-2 (human epithelial type 2), LLC-MK2, and 293 (human embryonic kidney epithelial) cells were obtained from the ATCC (Manassas, VA, USA) and maintained as previously described [33]. Primary cultured human SAE (small airway epithelial) cells were purchased from Lonza (distributed via Fisher Scientific, Pittsburgh, PA, USA). The isolate HMPV CAN-83 was propagated in LLC-MK2 cells at 35 °C in the absence of serum and the presence of 1 μg/mL of trypsin and was sucrose-purified, as previously described [38,39]. HMPV titer was determined by immunostaining in LLC-MK2 cells using polyclonal rabbit anti-HMPV antibody and streptavidin-peroxidase-conjugated secondary antibody (Sigma-Aldrich, St. Louis, MO, USA), as previously described [40]. Human AdV serotype 5 was kindly provided by Dr. Matthew D. Weitzman from the Children’s Hospital of Philadelphia. AdV yield was determined by plaque assay using crystal violet staining as previously described [41]. RSV long strain was propagated in HEp-2 cells at 37 °C and purified by sucrose gradient as described previously [33,42]. RSV titer was determined by immunostaining in HEp-2 cells using polyclonal biotin-conjugated goat anti-RSV antibody (Bio-Rad, Hercules, CA, USA) and streptavidin-peroxidase-conjugated secondary antibody, as previously described [33,42]. CE3F4 and H89 were purchased from Tocris (Lille, France) and LC Laboratories (Woburn, MA, USA), respectively. MAY0132 and ESI-05 were synthesized by the laboratory of co-author J.Z. [43]. Poly I:C and recombinant human TNF-α were purchased from Sigma-Aldrich and R and D Systems (Minneapolis, MN, USA), respectively.

2.2. Cytokine and Chemokine Quantification

A549 and SAE cells were inoculated with HMPV, AdV, or RSV at a multiplicity of infection (MOI) of 1. After 2 h, cells were treated with dimethyl sulfoxide (DMSO, vehicle control), 20 μM CE3F4, or 20 μM MAY0132 in fresh media. At 15 h post-treatment, cytokines/chemokines from collected supernatant were quantified using a Bio-Plex Pro Human Cytokine 27-plex Assay kit from Bio-Rad according to the manufacturer’s instructions. Data were analyzed using the multiplex analysis software from Bio-Rad.

2.3. Quantitative Real-Time PCR (qRT-PCR)

For quantitation of cytokine/chemokine mRNAs or the mRNA of AdV-DNA-binding protein (DBP), total cellular RNA was extracted using TRIzol reagents (Thermo Fisher Scientific, Waltham, MA, USA), followed by reverse transcription into cDNA using iScriptTM cDNA Synthesis Kit (Bio-Rad, Hercules, CA, USA) according to the manufacturer’s instructions. qRT-PCR was performed by using iTaq™ Universal SYBR Green Supermix (Bio-Rad, Hercules, CA, USA) in the CFX Connect Real-Time PCR System (Bio-Rad, Hercules, CA, USA). To quantify the viral genome or N gene of HMPV or RSV, total cellular RNA was extracted and cDNA was synthesized using HMPV- or RSV-specific reverse transcription primers with TaqMan™ Reverse Transcription Reagents from Thermo Fisher Scientific, followed by qRT-PCR, as described in [44,45]. The expression of IP-10, RANTES, AdV-DBP, and HMPV- or RSV-N was calculated by the 2ΔΔct method. 18S ribosomal RNA or glyceraldehyde 3-phosphate dehydrogenase (GAPDH) was used for normalization. The amount of HMPV or RSV genome was calculated by the absolute quantitation method. The information on primers for reverse transcription and qRT-PCR is shown in Supplementary Table S1.

2.4. Reporter Gene Assay

293 cells in triplicate were transfected with a luciferase reporter gene plasmid containing multiple copies of NF-κB binding sites (NF-κB-luc) using X-tremeGENE 9 (Roche, Indianapolis, IN, USA), as described previously [33,40], treated with or without the EPAC-isoform-specific inhibitors. Dimethyl sulfoxide (DMSO) was used as a vehicle control. At 15 h post-transfection, cells were transfected with poly I:C using Lipofectamine 2000 (Life Technologies, Grand Island, NY, USA) or treated with TNF-α with or without the inhibitor. After 6 h, cells were lysed to measure luciferase reporter activity. Cells without poly I:C or TNF-α treatment were used as negative controls.

2.5. Statistical Analysis

Statistical significance was determined using analysis of variance (ANOVA). A p value less than 0.05 was considered significant. One and two asterisks designate a p value less than 0.05 and 0.01, respectively. Data are presented as means ± standard error (SE).

3. Results

3.1. EPAC2 Promotes HMPV Replication

Our early observation on the EPAC2′s significance in RSV infection prompted us to investigate the broad effect of EPAC on other respiratory viral infections [33]. To the best of our knowledge, the function of EPAC2 in viral infection has not been extensively demonstrated. EPAC1, on the other hand, has been shown to be involved in MERS-CoV, Ebola virus, and recently vesicular stomatitis virus (VSV) infections [28,34,35]. However, the overall impact of EPAC1 on viral infections, especially the regulatory mechanisms, is still largely unknown.
Although HMPV and RSV belong to the same family (Pneumoviridae) and share many things in common, they also carry their distinct features [46,47]. To investigate whether and how EPAC plays a critical role in HMPV infection, we decided to investigate the impact of CE3F4 and MAY0132 on HMPV replication. We also tested a PKA inhibitor (H89) to explore whether cAMP/PKA signaling is involved in viral replication as well. As shown in Figure 1A, the MAY0132 treatment of A549 cells, a human alveolar epithelial cell line, led to a significant decrease in HMPV titer, by one and a half log, compared with DMSO-treated cells. Either H89 or CE3F4 treatment seemed uninfluential on HMPV replication. We also observed the inhibitory effect of MAY0132 on HMPV replication in primary small airway epithelial (SAE) cells, a more physiologically relevant cell model for the infection. Similar to the result demonstrated in Figure 1A, MAY0132-treated SAE cells showed remarkable suppression of infectious virus particles, compared with DMSO treatment, while CE3F4 treatment had no effect (Figure 1B). Furthermore, we confirmed the anti-HMPV effect of MAY0132 by investigating HMPV genome copies and N gene expression using qRT-PCR. Treatment with MAY0132, but not CE3F4, resulted in a considerable suppression in viral genome copies (Figure 1C) and N expression (Figure 1D) in comparison with the infected samples with DMSO treatment. Overall, these results demonstrate that EPAC2 is the isoform that is involved in the regulation of HMPV replication in airway epithelial cells (ACEs), a cell line, or primarily cultured cells.

3.2. EPAC2 Regulates HMPV-Induced Cytokines/Chemokines

The innate immune response is the first line of defense of hosts against viral infection and cytokines/chemokines are key molecules in innate immunity. HMPV has been demonstrated as a strong inducer of cytokines/chemokines [47,48] and several pro-inflammatory mediators are associated with increased disease severity in HMPV-infected patients [49,50]. To examine whether EPAC2 regulates virus-induced cytokines/chemokines, we also compared HMPV-induced cytokines/chemokines among cells, treated with/without chemical compounds. We found that, in A549 cells, HMPV-induced pro-inflammatory immune mediators such as IP-10, RANTES, MCP-1, TNF-α, MIP-1β, and IL-6, were significantly decreased by MAY0132 treatment, compared with that of DMSO (Figure 2A). Figure 2A also showed that CE3F4 suppressed the production of these cytokines/chemokines in response to HMPV infection but to a lesser extent than that of MAY0132. Consistent with the anti-inflammatory impact of MAY0132 on HMPV-infected A549, MAY0132-treated SAE cells also showed significantly suppressed production of IL-6, IP-10, RANTES, TNF-α, and MIP-1β, compared with DMSO control cells (Figure 2B). However, CE3F4 did not suppress the immune mediators’ induction in SAE cells, demonstrating a slight difference in cellular responses between an infected airway epithelial cell line and primary cultured airway epithelial cells and also highlighting the importance of EPAC2 in HMPV infection.
In the context of viral infection, the production of cytokines/chemokines is often dependent on viral replication [46,51], but sometimes the induction could also be replication independent [52,53]. To determine whether MAY0132-decreased cytokine/chemokine induction completely resulted from decreased viral replication, we measured cytokines/chemokines at 4 h post-infection (p.i.), an early time point when the viral replication (Figure 2C) and the N expression (Figure 2D) in the DMSO-, CE3F4-, and MAY0132-treated cells were comparable. At the early time point post-infection, most HMPV-induced cytokines/chemokines are too little to be detected by real-time PCR. IP-10, however, is a very inducible molecule and is the most inducible protein at 15 h p.i. Therefore, we used the IP-10 expression as a representative to study the impact of EPAC on HMPV-induced inflammatory gene expression. As shown in Figure 2E, at the early time point p.i., HMPV-induced IP-10 was detectable and significantly suppressed by MAY0132 treatment. CE3F4 also decreased virus-induced IP-10 transcription but less effectively than MAY0132. IP-10 is a suitable representative of important immune mediators for the study, as it is associated with the severity of acute respiratory infection in healthy adults [54]. In addition, IP-10 production has also been shown in rhinovirus-infected monocytic and reovirus-infected epithelial cells [55,56]. Overall, our results suggested that EPAC2 could control early inflammatory responses to HMPV infection in a replication-independent fashion.

3.3. EPAC2 Is Responsible for AdV Replication

As RSV, HMPV, Ebola, VSV, and MERS-CoV are all RNA viruses, we then decided to take a look at whether EPAC plays a role in respiratory DNA viral infection. Herein, AdV, which is a double-stranded DNA virus and also one of the predominant viral pathogens causing lower RTI in humans, was used for the study [16]. As shown in Figure 3A, the AdV titer was not affected by treatment with H89 and CE3F4, while there was a significant drop in viral replication in the MAY0132-treated A549 cells, compared with DMSO-treated infected cells. Furthermore, MAY0132 showed an inhibitory effect on the expression of the AdV-DBP gene, which is essential for AdV DNA replication and gene expression [57] (Figure 3B). CE3F4 slightly decreased AdV-DBP gene expression, although it did not affect AdV titer. The anti-AdV function of MAY0132 was also confirmed in virus-infected SAE cells, which shows the suppression in infectious progeny virus (Figure 3C). These results demonstrate that EPAC2 inhibition also suppresses the replication of AdV, which is a respiratory DNA virus.

3.4. EPAC2 Regulates AdV-Induced Cytokines/Chemokines

Next, we examined whether MAY0132 affects cytokine/chemokine production upon AdV infection by measuring the expression of RANTES and IP-10, two pro-inflammatory chemokines accounting for the severity of acute respiratory infection [54,58]. Compared with RSV and HMPV, AdV is not a great inducer of cytokines/chemokines. Our AdV belongs to type 5, which has been reported as a weak inducer of cytokines/chemokines compared to other AdV serotypes [59,60], but can induce RANTES and IP-10 in human respiratory cells [61]. As shown in Figure 4, AdV infection of SAE cells led to the induction of RANTES and IP-10. MAY0132 significantly blocked both AdV-induced pro-inflammatory mediators, whereas CE3F4 also had a minor impact on IP-10 induction. Overall, these results demonstrate that EPAC2 inhibition has a significant anti-inflammatory effect on AdV infection.

3.5. EPAC2 Regulates RSV Replication and Virus-Induced Cytokines/Chemokines

We have previously revealed that treatment with ESI-05, another EPAC2-specific inhibitor, suppresses RSV replication and pro-inflammatory responses [33]. Consistently, we found that treatment with MAY0132 led to the remarkable suppression of RSV replication in A549 cells, which is comparable to that of ESI-05 (Figure 5A). Herein, we also found that CE3F4 treatment did not show an antiviral effect, consistent with our publication on an unessential role of EPAC1 in RSV replication based on EPAC1 knockdown experiments [33]. As shown in Figure 5B, MAY0132 also reduced the production of virus-induced immune mediators such as IP-10, RANTES, and TNF-α [33]. EPAC2-mediated RSV replication and cytokine/chemokine induction by RSV were also confirmed in SAE cells (Figure 5C,D). We also found that IP-10 induction was impaired by MAY0132 treatment early after RSV infection (Figure 5G) when the change of viral genome (Figure 5E) and N gene (Figure 5F) by inhibitors could not be detected, suggesting replication-independent direct suppression of MAY0132 on cytokine induction. Taken together, these results support our previous findings on the importance of EPAC2, but not EPAC1, in regulating RSV infection.

3.6. EPAC2 Regulates Poly I:C- and TNF-α-Induced Inflammatory Signaling Pathways

NF-κB is one of the major transcription factors that control the expression of cytokines/chemokines [62,63]. In response to viral infections, the induction of various inflammatory genes requires NF-κB activation for their transcription [64,65]. Since EPAC2 seems to play a dominant role in HMPV, AdV, and RSV infection, and the EPAC2-mediated cytokine/chemokine induction likely has a component that is replication independent, we used MAY0132 to investigate the role of EPAC2 in poly I:C/ TNF-α-activated NF-κB. As shown in Figure 6A,B, when cells were transfected with a luciferase reporter plasmid containing the binding sites of NF-κB, treatment with poly I:C or TNF-α efficiently increased the luciferase activity, indicating NF-κB activation. The addition of ESI-05 or MAY0132 significantly decreased poly I:C- and TNF-α-enhanced signals, supporting that EPAC2 mediated poly I:C/ TNF-α-activated inflammatory signaling.

4. Discussion

Respiratory viral-induced illnesses are among the most common and severe diseases in the pediatric age group and high-risk populations, resulting in a great need for effective treatment and preventive vaccines. This study aims to evaluate the function of EPAC in both RNA and DNA respiratory viral infections using isoform-specific inhibitors. Herein, we demonstrated that EPAC2 plays a dominant role in regulating HMPV, AdV, and RSV infections. Treatment with MAY0132, an EPAC2-specific inhibitor, suppressed not only viral replication but also the induction of pro-inflammatory mediators in response to these three viruses. Moreover, the anti-inflammatory effect of MAY0132 was also observed in the context of poly I:C and TNF-α stimulation. Taken together, this study provides the first evidence on the potential of EPAC2 as a broad-spectrum drug target of multiple respiratory viral infections.
As discussed, EPAC has two major forms: EPAC1 and EPAC2. They exhibit distinct expression patterns and interacting partners, suggesting the discrete involvement in diverse biological functions [66,67]. The development of EPAC-isoform-specific pharmacological modulators led to significant progress in studying EPAC-mediated signaling pathways [30,31,32]. CE3F4 and its derivative have been well known as EPAC1 inhibitors [68], and AM-001 was recently identified an EPAC1 inhibitor as well [69]. In addition, EPAC2-specific inhibitors including ESI-05, HJC0350, and MAY0132 have been also developed [67]. Among them, MAY0132 has been well-characterized as a specific and potent EPAC2 inhibitor with an apparent 50% inhibitory concentration (IC50) value of 0.4 μM [43]. It selectively inhibits EPAC2 with no effect on EPAC1 activity at up to 100 μM. Herein, we used EPAC inhibitors to our benefit to discover EPAC2 as a new determinant to control the viral replication and host innate responses to HMPV and AdV. In this study, we also found that the EPAC1 inhibitor CE3F4 also suppressed some immune mediators in HMPV and AdV infections, however, to a lesser extent and significance than the EPAC2 inhibitors. Using EPAC1/2-specific inhibitors we also confirmed the role of EPAC2 and excluded the involvement of EPAC1 in RSV infection [33]. Therefore, these studies demonstrated that EPAC2 has an overall prevalent function in HMPV, AdV, and RSV infections. As discussed, some viruses, including MERS-CoV and Ebola, use EPAC1 to promote entry [28,35], supporting that EPAC isoforms and associated signaling pathways are also viral specific. How viruses select EPAC isoforms to evade host defense needs to be further elucidated in the future.
The development of diagnostic methods such as next-generation sequencing and multiplex PCR has allowed for the simultaneous identification of multiple causative agents from one sample in rapid and accurate ways. Based on this technology, there have been accumulating observations that respiratory diseases are attributed to multiple respiratory pathogens and a significant portion of cases shows co-infections with more than one virus [70,71]. Currently, it is controversial whether viral co-infections are associated with the severity of clinical outcomes. Several groups have reported that co-infection is less severe than a single infection and even beneficial, possibly due to viral competition for resources [72,73,74,75]. On the contrary, other studies have reported the association between co-infection and severe disease outcomes including pediatric intensive care unit (PICU) admission [15,76,77,78]. Despite the controversy, it is evident that drugs targeting multipathogens during viral co-infections should be beneficial for respiratory illness control. HMPV, AdV, and RSV are among the most common contributors to prevailing human respiratory disease, with co-infections that occur quite often as well [79,80]. Since EPAC2-specific inhibitors have a broad antiviral and anti-inflammation impact against all three major respiratory viruses, it is a promising treatment of modality against HMPV, AdV, and RSV.
As mentioned above, the activation of NF-κB is essential for the induction of inflammatory and antiviral genes [62]. The NF-κB pathway is activated in response to diverse stimuli including pattern-recognition receptors (PRRs) such as Toll-like receptors (TLRs) and retinoic acid-inducible gene I (RIG-I)/MDA5, as well as, TNF receptor family members [81]. Previous findings have revealed that HMPV and RSV infections of AECs induce the first wave of immune mediators through the NF-κB signaling pathway, which is activated via RIG-I/mitochondrial antiviral signaling protein (MAVS)-dependent signaling pathways [81,82]. On the other hand, poly I:C, a well-characterized inducer of NF-κB, can be recognized through two different pathways of TLR-3 and RIG-I/MDA5 [83,84,85]. In this study, since 293 cells do not express any TLRs, NF-κB activation by poly I:C treatment of 293 cells (Figure 6) is possibly from RIG-I/MDA5 or other unrevealed signaling [86]. Given the fact that MAY0132 efficiently blocked NF-κB activation, EPAC2-mediated signaling might either interact with the downstream molecules along with NF-κB-activating pathways or directly target NF-κB. In the future, we will investigate the molecules associated with EPAC2 and understand the consequence of such an interaction in regulating viral replication and host innate immune responses to respiratory viral infections. We are also planning to expand our research to different virus serotypes to investigate whether the impact of EPAC on the virus is serotype specific.

Supplementary Materials

The following are available online at https://0-www-mdpi-com.brum.beds.ac.uk/article/10.3390/v13061179/s1. Table S1. Primers for reverse transcription and qRT-PCR.

Author Contributions

E.-J.C. and X.B. wrote the manuscript; E.-J.C., W.W., X.C., and X.B. contributed to experimental design and data analysis, E.-J.C., W.W., X.C., K.Z., J.L., S.Y., A.S., and U.M.U. performed the experiments; P.W. and J.Z. synthesized EPAC2-specific inhibitors; and X.B. was responsible for project oversight. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the NIH R01 AI116812 and R21 AI113771, and FAMRI Clinical Innovator Award 160020 to X.B., J.Z. is partly supported by the John, D., Stobo, M.D. Distinguished Chair Endowment Fund at UTMB.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data is available upon the request.

Acknowledgments

We also thank Cynthia Tribble for the manuscript editing and Teodora Ivanciuc for performing the Bio-Plex to quantify the cytokines and chemokines.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Forum of International Respiratory Societies. The Global Impact of Respiratory Disease; European Respiratory Society: Lausanne, Switzerland, 2017. [Google Scholar]
  2. Wardlaw, T.M.; Johansson, E.W.; Hodge, M.; World Health Organization; UNICEF. Pneumonia: The Forgotten Killer of Children; UNICEF: New York, NY, USA; WHO: Geneva, Switzerland, 2006; p. 42.
  3. Kasper, D.; Fauci, A.; Hauser, S.; Longo, D.; Jameson, J.L.; Loscalzo, J. Infections Due to DNA and RNA Respiratory Viruses. In Harrison’s Principles of Internal Medicine, 19th ed.; McGraw-Hill Education: New York, NY, USA, 2014. [Google Scholar]
  4. Lu, R.; Zhao, X.; Li, J.; Niu, P.; Yang, B.; Wu, H.; Wang, W.; Song, H.; Huang, B.; Zhu, N.; et al. Genomic characterisation and epidemiology of 2019 novel coronavirus: Implications for virus origins and receptor binding. Lancet 2020, 395, 565–574. [Google Scholar] [CrossRef] [Green Version]
  5. World Health Organization. Disease Outbreak News. Novel Coronavirus—China; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  6. World Health Organization. WHO Coronavirus (COVID-19) Dashboard; WHO: Geneva, Switzerland, 2020. [Google Scholar]
  7. Singh, K.; Kondal, D.; Mohan, S.; Jaganathan, S.; Deepa, M.; Venkateshmurthy, N.S.; Jarhyan, P.; Anjana, R.M.; Narayan, K.M.V.; Mohan, V.; et al. Health, psychosocial, and economic impacts of the COVID-19 pandemic on people with chronic conditions in India: A mixed methods study. BMC Public Health 2021, 21, 1–15. [Google Scholar] [CrossRef]
  8. Falsey, A.R.; Erdman, D.; Anderson, L.J.; Walsh, E.E. Human Metapneumovirus Infections in Young and Elderly Adults. J. Infect. Dis. 2003, 187, 785–790. [Google Scholar] [CrossRef] [PubMed]
  9. Madhi, S.A.; Ludewick, H.; Abed, Y.; Klugman, K.P.; Boivin, G. Human Metapneumovirus-Associated Lower Respiratory Tract Infections among Hospitalized Human Immunodeficiency Virus Type 1 (HIV-1)-Infected and HIV-1-Uninfected African Infants. Clin. Infect. Dis. 2003, 37, 1705–1710. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  10. Viazov, S.; Ratjen, F.; Scheidhauer, R.; Fiedler, M.; Roggendorf, M. High Prevalence of Human Metapneumovirus Infection in Young Children and Genetic Heterogeneity of the Viral Isolates. J. Clin. Microbiol. 2003, 41, 3043–3045. [Google Scholar] [CrossRef] [Green Version]
  11. Crowe, J.E. Human Metapneumovirus as a Major Cause of Human Respiratory Tract Disease. Pediatr. Infect. Dis. J. 2004, 23, S215–S221. [Google Scholar] [CrossRef] [PubMed]
  12. Edwards, K.M.; Zhu, Y.; Griffin, M.R.; Weinberg, G.A.; Hall, C.B.; Szilagyi, P.G.; Staat, M.A.; Iwane, M.; Prill, M.M.; Williams, J.V. Burden of Human Metapneumovirus Infection in Young Children. N. Engl. J. Med. 2013, 368, 633–643. [Google Scholar] [CrossRef] [Green Version]
  13. Hall, C.B.; Weinberg, G.A.; Iwane, M.K.; Blumkin, A.K.; Edwards, K.M.; Staat, M.A.; Auinger, P.; Griffin, M.R.; Poehling, K.A.; Erdman, D.; et al. The Burden of Respiratory Syncytial Virus Infection in Young Children. N. Engl. J. Med. 2009, 360, 588–598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Li, Y.; Pillai, P.; Miyake, F.; Nair, H. The role of viral co-infections in the severity of acute respiratory infections among children infected with respiratory syncytial virus (RSV): A systematic review and meta-analysis. J. Glob. Health 2020, 10, 010426. [Google Scholar] [CrossRef]
  15. Semple, M.G.; Cowell, A.; Dove, W.F.; Greensill, J.; McNamara, P.; Halfhide, C.P.; Shears, P.; Smyth, R.L.; Hart, C.A. Dual Infection of Infants by Human Metapneumovirus and Human Respiratory Syncytial Virus Is Strongly Associated with Severe Bronchiolitis. J. Infect. Dis. 2005, 191, 382–386. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  16. Wen, S.; Lin, Z.; Zhang, Y.; Lv, F.; Li, H.; Zhang, X.; Lin, L.; Zhu, H.-H.; Xu, Z.; Li, C.; et al. The Epidemiology, Molecular, and Clinical of Human Adenoviruses in Children Hospitalized with Acute Respiratory Infections. Front. Microbiol. 2021, 12, 629971. [Google Scholar] [CrossRef]
  17. Scott, M.K.; Chommanard, C.; Lu, X.; Appelgate, D.; Grenz, L.; Schneider, E.; Gerber, S.I.; Erdman, D.D.; Thomas, A. Human Adenovirus Associated with Severe Respiratory Infection, Oregon, USA, 2013–2014. Emerg. Infect. Dis. 2016, 22, 1044–1051. [Google Scholar] [CrossRef] [Green Version]
  18. Alharbi, S.; Van Caeseele, P.; Consunji-Araneta, R.; Zoubeidi, T.; Fanella, S.; Souid, A.-K.; Alsuwaidi, A.R. Epidemiology of severe pediatric adenovirus lower respiratory tract infections in Manitoba, Canada, 1991–2005. BMC Infect. Dis. 2012, 12, 55. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Gray, G.C.; McCarthy, T.; Lebeck, M.G.; Schnurr, D.P.; Russell, K.L.; Kajon, A.E.; Landry, M.L.; Leland, D.S.; Storch, G.A.; Ginocchio, C.C.; et al. Genotype prevalence and risk factors for severe clinical adenovirus infection, United States 2004–2006. Clin. Infect. Dis. 2007, 45, 1120–1131. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Echavarría, M. Adenoviruses in Immunocompromised Hosts. Clin. Microbiol. Rev. 2008, 21, 704–715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  21. Resch, B. Product review on the monoclonal antibody palivizumab for prevention of respiratory syncytial virus infection. Hum. Vaccines Immunother. 2017, 13, 2138–2149. [Google Scholar] [CrossRef] [Green Version]
  22. De Rooij, J.; Zwartkruis, F.J.; Verheijen, M.H.; Cool, R.H.; Nijman, S.M.; Wittinghofer, A.; Bos, J.L. Epac is a Rap1 gua-nine-nucleotide-exchange factor directly activated by cyclic AMP. Nature 1998, 396, 474–477. [Google Scholar] [CrossRef]
  23. Kawasaki, H.; Springett, G.M.; Mochizuki, N.; Toki, S.; Nakaya, M.; Matsuda, M.; Housman, D.E.; Graybiel, A.M. A family of cAMP-binding proteins that directly activate Rap1. Science 1998, 282, 2275–2279. [Google Scholar] [CrossRef] [Green Version]
  24. Metrich, M.; Lucas, A.; Gastineau, M.; Samuel, J.L.; Heymes, C.; Morel, E.; Lezoualc’’h, F. Epac mediates beta-adrenergic receptor-induced cardiomyocyte hypertrophy. Circ. Res. 2008, 102, 959–965. [Google Scholar] [CrossRef] [Green Version]
  25. Kumar, N.; Gupta, S.; Dabral, S.; Singh, S.; Sehrawat, S. Role of exchange protein directly activated by cAMP (EPAC1) in breast cancer cell migration and apoptosis. Mol. Cell. Biochem. 2017, 430, 115–125. [Google Scholar] [CrossRef]
  26. Suzuki, S.; Yokoyama, U.; Abe, T.; Kiyonari, H.; Yamashita, N.; Kato, Y.; Kurotani, R.; Sato, M.; Okumura, S.; Ishikawa, Y. Differential Roles of Epac in Regulating Cell Death in Neuronal and Myocardial Cells. J. Biol. Chem. 2010, 285, 24248–24259. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Griggs, R.B.; Santos, D.F.; Laird, D.E.; Doolen, S.; Donahue, R.R.; Wessel, C.R.; Fu, W.; Sinha, G.P.; Wang, P.; Zhou, J.; et al. Methylglyoxal and a spinal TRPA1-AC1-Epac cascade facilitate pain in the db/db mouse model of type 2 diabetes. Neurobiol. Dis. 2019, 127, 76–86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Tao, X.; Mei, F.; Agrawal, A.; Peters, C.J.; Ksiazek, T.G.; Cheng, X.; Tseng, C.-T.K. Blocking of Exchange Proteins Directly Activated by cAMP Leads to Reduced Replication of Middle East Respiratory Syndrome Coronavirus. J. Virol. 2014, 88, 3902–3910. [Google Scholar] [CrossRef] [Green Version]
  29. Chen, Y.-F.; Huang, G.; Wang, Y.-M.; Cheng, M.; Zhu, F.-F.; Zhong, J.-N.; Gao, Y.-D. Exchange protein directly activated by cAMP (Epac) protects against airway inflammation and airway remodeling in asthmatic mice. Respir. Res. 2019, 20, 1–14. [Google Scholar] [CrossRef] [Green Version]
  30. Parnell, E.; Palmer, T.M.; Yarwood, S.J. The future of EPAC-targeted therapies: Agonism versus antagonism. Trends Pharmacol. Sci. 2015, 36, 203–214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  31. Xu, Y.; Schwede, F.; Wienk, H.; Tengholm, A.; Rehmann, H. A Membrane Permeable Prodrug of S223 for Selective Epac2 Activation in Living Cells. Cells 2019, 8, 1589. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  32. Lezoualc’H, F.; Fazal, L.; Laudette, M.; Conte, C. Cyclic AMP Sensor EPAC Proteins and Their Role in Cardiovascular Function and Disease. Circ. Res. 2016, 118, 881–897. [Google Scholar] [CrossRef]
  33. Choi, E.-J.; Ren, Y.; Chen, Y.; Liu, S.; Wu, W.; Ren, J.; Wang, P.; Garofalo, R.P.; Zhou, J.; Bao, X. Exchange Proteins Directly Activated by cAMP and Their Roles in Respiratory Syncytial Virus Infection. J. Virol. 2018, 92, 22. [Google Scholar] [CrossRef] [Green Version]
  34. Zhang, C.; Yan, Y.; He, H.; Wang, L.; Zhang, N.; Zhang, J.; Huang, H.; Wu, N.; Ren, H.; Qian, M.; et al. IFN-stimulated P2Y13 protects mice from viral infection by suppressing the cAMP/EPAC1 signaling pathway. J. Mol. Cell Biol. 2018, 11, 395–407. [Google Scholar] [CrossRef] [Green Version]
  35. Drelich, A.; Judy, B.; He, X.; Chang, Q.; Yu, S.; Li, X.; Lu, F.; Wakamiya, M.; Popov, V.; Zhou, J.; et al. Exchange Protein Directly Activated by cAMP Modulates Ebola Virus Uptake into Vascular Endothelial Cells. Viruses 2018, 10, 563. [Google Scholar] [CrossRef] [Green Version]
  36. Courilleau, D.; Bouyssou, P.; Fischmeister, R.; Lezoualc’H, F.; Blondeau, J.-P. The (R)-enantiomer of CE3F4 is a preferential inhibitor of human exchange protein directly activated by cyclic AMP isoform 1 (Epac1). Biochem. Biophys. Res. Commun. 2013, 440, 443–448. [Google Scholar] [CrossRef]
  37. Liu, Z.; Zhu, Y.; Chen, H.; Wang, P.; Mei, F.C.; Ye, N.; Cheng, X.; Zhou, J. Structure-activity relationships of 2-substituted phenyl-N-phenyl-2-oxoacetohydrazonoyl cyanides as novel antagonists of exchange proteins directly activated by cAMP (EPACs). Bioorganic Med. Chem. Lett. 2017, 27, 5163–5166. [Google Scholar] [CrossRef] [PubMed]
  38. Bao, X.; Liu, T.; Shan, Y.; Li, K.; Garofalo, R.P.; Casola, A. Human Metapneumovirus Glycoprotein G Inhibits Innate Immune Responses. PLoS Pathog. 2008, 4, e1000077. [Google Scholar] [CrossRef] [PubMed]
  39. Bao, X.; Sinha, M.; Liu, T.; Hong, C.; Luxon, B.A.; Garofalo, R.P.; Casola, A. Identification of human metapneu-movirus-induced gene networks in airway epithelial cells by microarray analysis. Virology 2008, 374, 114–127. [Google Scholar] [CrossRef] [Green Version]
  40. Bao, X.; Kolli, D.; Liu, T.; Shan, Y.; Garofalo, R.P.; Casola, A. Human metapneumovirus small hydrophobic protein inhibits NF-kappaB transcriptional activity. J. Virol. 2008, 82, 8224–8229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Reyes, E.D.; Kulej, K.; Pancholi, N.J.; Akhtar, L.; Avgousti, D.C.; Kim, E.T.; Bricker, D.K.; Spruce, L.A.; Koniski, S.A.; Seeholzer, S.H.; et al. Identifying Host Factors Associated with DNA Replicated During Virus Infection. Mol. Cell. Proteom. 2017, 16, 2079–2097. [Google Scholar] [CrossRef] [Green Version]
  42. Bao, X.; Indukuri, H.; Liu, T.; Liao, S.L.; Tian, B.; Brasier, A.R.; Garofalo, R.P.; Casola, A. IKKepsilon modulates RSV-induced NF-kappaB-dependent gene transcription. Virology 2010, 408, 224–231. [Google Scholar] [CrossRef] [Green Version]
  43. Wild, C.T.; Zhu, Y.; Na, Y.; Mei, F.; Ynalvez, M.A.; Chen, H.; Cheng, X.; Zhou, J. Functionalized N,N-Diphenylamines as Potent and Selective EPAC2 Inhibitors. ACS Med. Chem. Lett. 2016, 7, 460–464. [Google Scholar] [CrossRef] [Green Version]
  44. Ren, J.; Wang, Q.; Kolli, D.; Prusak, D.J.; Tseng, C.-T.K.; Chen, Z.; Li, K.; Wood, T.G.; Bao, X. Human Metapneumovirus M2-2 Protein Inhibits Innate Cellular Signaling by Targeting MAVS. J. Virol. 2012, 86, 13049–13061. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  45. Li, H.; Ma, Y.; Escaffre, O.; Ivanciuc, T.; Komaravelli, N.; Kelley, J.P.; Coletta, C.; Szabo, C.; Rockx, B.; Garofalo, R.P.; et al. Role of Hydrogen Sulfide in Paramyxovirus Infections. J. Virol. 2015, 89, 5557–5568. [Google Scholar] [CrossRef] [Green Version]
  46. Guerrero-Plata, A.; Casola, A.; Suarez, G.; Yu, X.; Spetch, L.; Peeples, M.E.; Garofalo, R.P. Differential Response of Dendritic Cells to Human Metapneumovirus and Respiratory Syncytial Virus. Am. J. Respir. Cell Mol. Biol. 2006, 34, 320–329. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Guerrero-Plata, A.; Casola, A.; Garofalo, R.P. Human Metapneumovirus Induces a Profile of Lung Cytokines Distinct from That of Respiratory Syncytial Virus. J. Virol. 2005, 79, 14992–14997. [Google Scholar] [CrossRef] [Green Version]
  48. Huck, B.; Neumann-Haefelin, D.; Schmitt-Graeff, A.; Weckmann, M.; Mattes, J.; Ehl, S.; Falcone, V. Human metapneumovirus induces more severe disease and stronger innate immune response in BALB/c mice as compared with respiratory syncytial virus. Respir. Res. 2007, 8, 6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. Malmo, J.; Moe, N.; Krokstad, S.; Ryan, L.; Loevenich, S.; Johnsen, I.B.; Espevik, T.; Nordbø, S.A.; Døllner, H.; Anthonsen, M.W. Cytokine Profiles in Human Metapneumovirus Infected Children: Identification of Genes Involved in the Antiviral Response and Pathogenesis. PLoS ONE 2016, 11, e0155484. [Google Scholar] [CrossRef] [PubMed]
  50. Pancham, K.; Perez, G.F.; Huseni, S.; Jain, A.; Kurdi, B.; Rodriguez-Martinez, C.E.; Preciado, D.; Rose, M.C.; Nino, G. Prem-ature infants have impaired airway antiviral IFNgamma responses to human metapneumovirus compared to respiratory syncytial virus. Pediatr. Res. 2015, 78, 389–394. [Google Scholar] [CrossRef] [Green Version]
  51. Melchjorsen, J.; Siren, J.; Julkunen, I.; Paludan, S.R.; Matikainen, S. Induction of cytokine expression by herpes simplex virus in human monocyte-derived macrophages and dendritic cells is dependent on virus replication and is counteracted by ICP27 targeting NF-kappaB and IRF-3. J. Gen. Virol. 2006, 87, 1099–1108. [Google Scholar] [CrossRef]
  52. Saba, T.G.; Chung, Y.; Hong, J.Y.; Sajjan, U.S.; Bentley, J.K.; Hershenson, M.B. Rhinovirus-Induced Macrophage Cytokine Expression Does Not Require Endocytosis or Replication. Am. J. Respir. Cell Mol. Biol. 2014, 50, 974–984. [Google Scholar] [CrossRef]
  53. Forbester, J.L.; Clement, M.; Wellington, D.; Yeung, A.; Dimonte, S.; Marsden, M.; Chapman, L.; Coomber, E.L.; Tolley, C.; Lees, E.; et al. IRF5 Promotes Influenza Virus-Induced Inflam-matory Responses in Human Induced Pluripotent Stem Cell-Derived Myeloid Cells and Murine Models. J. Virol. 2020, 94. [Google Scholar] [CrossRef] [Green Version]
  54. Hayney, M.S.; Henriquez, K.M.; Barnet, J.H.; Ewers, T.; Champion, H.M.; Flannery, S.; Barrett, B. Serum IFN-gamma-induced protein 10 (IP-10) as a biomarker for severity of acute respiratory infection in healthy adults. J. Clin. Virol. 2017, 90, 32–37. [Google Scholar] [CrossRef]
  55. Korpi-Steiner, N.L.; Bates, M.E.; Lee, W.-M.; Hall, D.J.; Bertics, P.J. Human rhinovirus induces robust IP-10 release by monocytic cells, which is independent of viral replication but linked to type I interferon receptor ligation and STAT1 activation. J. Leukoc. Biol. 2006, 80, 1364–1374. [Google Scholar] [CrossRef]
  56. Hamamdzic, D.; Phillips-Dorsett, T.; Altman-Hamamdzic, S.; London, S.D.; London, L. Reovirus triggers cell type-specific proinflammatory responses dependent on the autocrine action of IFN-beta. Am. J. Physiol. Lung Cell. Mol. Physiol. 2001, 280, L18–L29. [Google Scholar] [CrossRef] [PubMed]
  57. Kruijer, W.; van Schaik, F.M.; Speijer, J.G.; Sussenbach, J.S. Structure and function of adenovirus DNA binding protein: Comparison of the amino acid sequences of the Ad5 and Ad12 proteins derived from the nucleotide sequence of the corre-sponding genes. Virology 1983, 128, 140–153. [Google Scholar] [CrossRef]
  58. Hornsleth, A.; Loland, L.; Larsen, L.B. Cytokines and chemokines in respiratory secretion and severity of disease in infants with respiratory syncytial virus (RSV) infection. J. Clin. Virol. 2001, 21, 163–170. [Google Scholar] [CrossRef]
  59. Teigler, J.E.; Iampietro, M.J.; Barouch, D.H. Vaccination with Adenovirus Serotypes 35, 26, and 48 Elicits Higher Levels of Innate Cytokine Responses than Adenovirus Serotype 5 in Rhesus Monkeys. J. Virol. 2012, 86, 9590–9598. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Booth, J.L.; Metcalf, J.P. Type-Specific Induction of Interleukin-8 by Adenovirus. Am. J. Respir. Cell Mol. Biol. 1999, 21, 521–527. [Google Scholar] [CrossRef]
  61. Tamanini, A.; Nicolis, E.; Bonizzato, A.; Bezzerri, V.; Melotti, P.; Assael, B.M.; Cabrini, G. Interaction of Adenovirus Type 5 Fiber with the Coxsackievirus and Adenovirus Receptor Activates Inflammatory Response in Human Respiratory Cells. J. Virol. 2006, 80, 11241–11254. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Oeckinghaus, A.; Ghosh, S. The NF-kappaB family of transcription factors and its regulation. Cold Spring Harb. Perspect. Biol. 2009, 1, a000034. [Google Scholar] [CrossRef] [PubMed]
  63. Song, K.; Li, S. The Role of Ubiquitination in NF-kappaB Signaling during Virus Infection. Viruses 2021, 13, 145. [Google Scholar] [CrossRef]
  64. Bao, X.; Liu, T.; Spetch, L.; Kolli, D.; Garofalo, R.; Casola, A. Airway epithelial cell response to human metapneumovirus infection. J. Virol. 2007, 368, 91–101. [Google Scholar] [CrossRef] [Green Version]
  65. Yoboua, F.; Martel, A.; Duval, A.; Mukawera, E.; Grandvaux, N. Respiratory syncytial virus-mediated NF-kappa B p65 phosphorylation at serine 536 is dependent on RIG-I, TRAF6, and IKK beta. J. Virol. 2010, 84, 7267–7277. [Google Scholar] [CrossRef] [Green Version]
  66. Schmidt, M.; Dekker, F.; Maarsingh, H. Exchange Protein Directly Activated by cAMP (epac): A Multidomain cAMP Mediator in the Regulation of Diverse Biological Functions. Pharmacol. Rev. 2013, 65, 670–709. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Wang, P.; Liu, Z.; Chen, H.; Ye, N.; Cheng, X.; Zhou, J. Exchange proteins directly activated by cAMP (EPACs): Emerging therapeutic targets. Bioorganic Med. Chem. Lett. 2017, 27, 1633–1639. [Google Scholar] [CrossRef] [PubMed]
  68. Courilleau, D.; Bisserier, M.; Jullian, J.C.; Lucas, A.; Bouyssou, P.; Fischmeister, R.; Blondeau, J.P.; Lezoualc’’h, F. Identification of a tetrahydroquinoline analog as a pharmacological inhibitor of the cAMP-binding protein Epac. J. Biol. Chem. 2012, 287, 44192–44202. [Google Scholar] [CrossRef] [Green Version]
  69. Laudette, M.; Coluccia, A.; Sainte-Marie, Y.; Solari, A.; Fazal, L.; Sicard, P.; Silvestri, R.; Mialet-Perez, J.; Pons, S.; Ghaleh, B.; et al. Identification of a pharmacological inhibitor of Epac1 that protects the heart against acute and chronic models of cardiac stress. Cardiovasc. Res. 2019, 115, 1766–1777. [Google Scholar] [CrossRef]
  70. Waner, J.L. Mixed viral infections: Detection and management. Clin. Microbiol. Rev. 1994, 7, 143–151. [Google Scholar] [CrossRef]
  71. Calvo, C.; García-García, M.L.; Blanco, C.; Vázquez, M.C.; Frías, M.E.; Pérez-Breña, P.; Casas, I. Multiple simultaneous viral infections in infants with acute respiratory tract infections in Spain. J. Clin. Virol. 2008, 42, 268–272. [Google Scholar] [CrossRef]
  72. Brand, H.K.; de Groot, R.; Galama, J.M.; Brouwer, M.L.; Teuwen, K.; Hermans, P.W.; Melchers, W.J.; Warris, A. Infection with multiple viruses is not associated with increased disease severity in children with bronchiolitis. Pediatr. Pulmonol. 2012, 47, 393–400. [Google Scholar] [CrossRef]
  73. Martin, E.T.; Fairchok, M.P.; Stednick, Z.J.; Kuypers, J.; Englund, J.A. Epidemiology of Multiple Respiratory Viruses in Childcare Attendees. J. Infect. Dis. 2013, 207, 982–989. [Google Scholar] [CrossRef] [Green Version]
  74. Martin, E.T.; Kuypers, J.; Wald, A.; Englund, J.A. Multiple versus single virus respiratory infections: Viral load and clinical disease severity in hospitalized children. Influenza Other Respir. Viruses 2012, 6, 71–77. [Google Scholar] [CrossRef] [PubMed]
  75. Pinky, L.; Dobrovolny, H.M. Coinfections of the Respiratory Tract: Viral Competition for Resources. PLoS ONE 2016, 11, e0155589. [Google Scholar] [CrossRef]
  76. Chan, K.C.; Yu, M.W.; Cheung, T.W.Y.; Lam, D.S.Y.; Leung, T.N.H.; Tsui, T.K.; Ip, K.I.; Chau, C.S.K.; Lee, S.L.; Yip, A.Y.F.; et al. Childhood bronchiolitis obliterans in Hong Kong-case series over a 20-year period. Pediatr. Pulmonol. 2021, 56, 153–161. [Google Scholar] [CrossRef] [PubMed]
  77. Goka, E.A.; Vallely, P.J.; Mutton, K.J.; Klapper, P.E. Single, dual and multiple respiratory virus infections and risk of hospi-talization and mortality. Epidemiol. Infect. 2015, 143, 37–47. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Richard, N.; Komurian-Pradel, F.; Javouhey, E.; Perret, M.; Rajoharison, A.; Bagnaud, A.; Billaud, G.; Vernet, G.; Lina, B.; Floret, D.; et al. The Impact of Dual Viral Infection in Infants Admitted to a Pediatric Intensive Care Unit Associated with Severe Bronchiolitis. Pediatr. Infect. Dis. J. 2008, 27, 213–217. [Google Scholar] [CrossRef]
  79. Meskill, S.D.; O’Bryant, S.C. Respiratory Virus Co-infection in Acute Respiratory Infections in Children. Curr. Infect. Dis. Rep. 2020, 22, 3. [Google Scholar] [CrossRef] [PubMed]
  80. Coiras, M.; Pérez-Breña, P.; García, M.; Casas, I. Simultaneous detection of influenza A, B, and C viruses, respiratory syncytial virus, and adenoviruses in clinical samples by multiplex reverse transcription nested-PCR assay. J. Med. Virol. 2003, 69, 132–144. [Google Scholar] [CrossRef]
  81. Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-kappaB signaling in inflammation. Signal Transduct. Target Ther. 2017, 2. [Google Scholar] [CrossRef] [Green Version]
  82. Liao, S.; Bao, X.; Liu, T.; Lai, S.; Li, K.; Garofalo, R.P.; Casola, A. Role of retinoic acid inducible gene-I in human metapneu-movirus-induced cellular signalling. J. Gen. Virol. 2008, 89 Pt 8, 1978–1986. [Google Scholar] [CrossRef]
  83. Alexopoulou, L.; Holt, A.C.; Medzhitov, R.; Flavell, R.A. Recognition of double-stranded RNA and activation of NF-kappaB by Toll-like receptor 3. Nature 2001, 413, 732–738. [Google Scholar] [CrossRef]
  84. Kato, H.; Takeuchi, O.; Sato, S.; Yoneyama, M.; Yamamoto, M.; Matsui, K.; Uematsu, S.; Jung, A.; Kawai, T.; Ishii, K.J.; et al. Differential roles of MDA5 and RIG-I helicases in the recognition of RNA viruses. Nat. Cell Biol. 2006, 441, 101–105. [Google Scholar] [CrossRef]
  85. Yoneyama, M.; Kikuchi, M.; Natsukawa, T.; Shinobu, N.; Imaizumi, T.; Miyagishi, M.; Taira, K.; Akira, S.; Fujita, T. The RNA helicase RIG-I has an essential function in double-stranded RNA-induced innate antiviral responses. Nat. Immunol. 2004, 5, 730–737. [Google Scholar] [CrossRef]
  86. Wang, J.; Shao, Y.; Bennett, T.A.; Shankar, R.A.; Wightman, P.D.; Reddy, L.G. The Functional Effects of Physical Interactions among Toll-like Receptors 7, 8, and 9. J. Biol. Chem. 2006, 281, 37427–37434. [Google Scholar] [CrossRef] [PubMed] [Green Version]
Figure 1. The impact of EPAC2 on HMPV replication. (A,B) A549 (A) or SAE (B) cells in triplicate were infected with HMPV at an MOI of 1. At 2 h post-infection (p.i.), cells were washed and subsequently treated with 20 μM CE3F4 or MAY0132. Next, 20 μM H89 was used to examine the role of cAMP/PKA signaling in HMPV infection. DMSO was used as vehicle control for EPAC inhibitors. At 15 h post-treatment, total viruses were harvested, and viral titer was determined by immunostaining using an anti-HMPV antibody. Data are representative of three independent experiments. ** p < 0.01, relative to the DMSO-treated group. (C,D) A549 cells in triplicate were infected with HMPV, followed by drug treatment as described in (A). After 15 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure HMPV genome copies (C) and N gene expression (D). 18S ribosomal RNA (rRNA) was used for normalization to N gene level. DMSO-treated mock infection was used as a negative control for DMSO-treated HMPV infection. ** p < 0.01, relative to the DMSO-treated and HMPV-infected group.
Figure 1. The impact of EPAC2 on HMPV replication. (A,B) A549 (A) or SAE (B) cells in triplicate were infected with HMPV at an MOI of 1. At 2 h post-infection (p.i.), cells were washed and subsequently treated with 20 μM CE3F4 or MAY0132. Next, 20 μM H89 was used to examine the role of cAMP/PKA signaling in HMPV infection. DMSO was used as vehicle control for EPAC inhibitors. At 15 h post-treatment, total viruses were harvested, and viral titer was determined by immunostaining using an anti-HMPV antibody. Data are representative of three independent experiments. ** p < 0.01, relative to the DMSO-treated group. (C,D) A549 cells in triplicate were infected with HMPV, followed by drug treatment as described in (A). After 15 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure HMPV genome copies (C) and N gene expression (D). 18S ribosomal RNA (rRNA) was used for normalization to N gene level. DMSO-treated mock infection was used as a negative control for DMSO-treated HMPV infection. ** p < 0.01, relative to the DMSO-treated and HMPV-infected group.
Viruses 13 01179 g001
Figure 2. EPAC2-mediated cytokine/chemokine induction by HMPV. (A,B) A549 (A) or SAE (B) cells in triplicate were infected with HMPV at an MOI of 1. At 2 h p.i., cells were treated with DMSO, 20 μM CE3F4, or 20 μM MAY0132. At 15 h post-treatment, the supernatant was collected, and the amount of diluted or undiluted cytokines/chemokines was measured by Bio-Plex. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (CE) A549 cells in triplicate were infected with HMPV, followed by the drug treatment as described in (A). At 4 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure HMPV genome copies (C), N gene expression (D), and IP-10 production (E). DMSO-treated mock infection was used as a negative control for DMSO-treated HMPV infection. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated and HMPV-infected group.
Figure 2. EPAC2-mediated cytokine/chemokine induction by HMPV. (A,B) A549 (A) or SAE (B) cells in triplicate were infected with HMPV at an MOI of 1. At 2 h p.i., cells were treated with DMSO, 20 μM CE3F4, or 20 μM MAY0132. At 15 h post-treatment, the supernatant was collected, and the amount of diluted or undiluted cytokines/chemokines was measured by Bio-Plex. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (CE) A549 cells in triplicate were infected with HMPV, followed by the drug treatment as described in (A). At 4 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure HMPV genome copies (C), N gene expression (D), and IP-10 production (E). DMSO-treated mock infection was used as a negative control for DMSO-treated HMPV infection. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated and HMPV-infected group.
Viruses 13 01179 g002
Figure 3. The impact of EPAC2 on AdV replication. (A) A549 cells in triplicate were infected with AdV at an MOI of 1. At 2 h p.i., cells were washed, followed by treatment with DMSO, 10 μM H89, 10 μM CE3F4, or 10 μM MAY0132. At 48 h post-treatment, total viruses were harvested, and infectious AdV particles were quantified by the plaque assay using crystal violet staining. Data are representative of three independent experiments. ** p < 0.01, relative to the DMSO-treated group. (B) A549 cells in triplicate were infected with AdV and treated with drugs as described in (A). At 48 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure AdV-DBP gene expression. 18S rRNA was used for normalization. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (C) SAE cells were infected with AdV at an MOI of 1 and then treated with DMSO, 10 μM CE3F4, or 10 μM MAY0132. At 48 h post-treatment, total viruses were prepared, and infectious AdV particles were quantified by the plaque assay. Data are representative of three independent experiments. *p < 0.05 and **p < 0.01, relative to the DMSO-treated group.
Figure 3. The impact of EPAC2 on AdV replication. (A) A549 cells in triplicate were infected with AdV at an MOI of 1. At 2 h p.i., cells were washed, followed by treatment with DMSO, 10 μM H89, 10 μM CE3F4, or 10 μM MAY0132. At 48 h post-treatment, total viruses were harvested, and infectious AdV particles were quantified by the plaque assay using crystal violet staining. Data are representative of three independent experiments. ** p < 0.01, relative to the DMSO-treated group. (B) A549 cells in triplicate were infected with AdV and treated with drugs as described in (A). At 48 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure AdV-DBP gene expression. 18S rRNA was used for normalization. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (C) SAE cells were infected with AdV at an MOI of 1 and then treated with DMSO, 10 μM CE3F4, or 10 μM MAY0132. At 48 h post-treatment, total viruses were prepared, and infectious AdV particles were quantified by the plaque assay. Data are representative of three independent experiments. *p < 0.05 and **p < 0.01, relative to the DMSO-treated group.
Viruses 13 01179 g003
Figure 4. EPAC2-mediated chemokine induction by AdV. (A,B) SAE cells in triplicate were mock-infected or infected with AdV at an MOI of 1. At 2 h p.i., cells were treated with DMSO, 10 μM CE3F4, or 10 μM MAY0132. At 48 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure the expression of RANTES (A) and IP-10 (B). GAPDH was used for normalization. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated and AdV-infected group.
Figure 4. EPAC2-mediated chemokine induction by AdV. (A,B) SAE cells in triplicate were mock-infected or infected with AdV at an MOI of 1. At 2 h p.i., cells were treated with DMSO, 10 μM CE3F4, or 10 μM MAY0132. At 48 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure the expression of RANTES (A) and IP-10 (B). GAPDH was used for normalization. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated and AdV-infected group.
Viruses 13 01179 g004
Figure 5. The impact of EPAC2 on RSV infection. (A,B) A549 cells in triplicate were infected with RSV at an MOI of 1. After 2 h p.i., cells were treated with DMSO, 20 μM CE3F4, 20 μM ESI-05, or 20 μM MAY0132. At 15 h post-treatment, (A) total viruses were collected, and viral titer was determined by immunostaining using an anti-RSV antibody. (B) The supernatant was collected, and diluted or undiluted cytokines/chemokines were measured by Bio-Plex. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (C,D) SAE cells were infected with RSV and treated with the indicated drugs as described in (A). At 15 h post-treatment, (C) virus titration was determined. ** p < 0.01, relative to the DMSO-treated group. (D) Cytokines/chemokines of SAE cells were also measured by Bio-Plex. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (EG) A549 cells in triplicate were infected with RSV and treated with drugs as described in (A). At 4 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure RSV genome copies (E), N gene expression (F), and IP-10 transcription (G). DMSO-treated mock infection was used as a negative control for DMSO-treated RSV infection. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated and RSV-infected group.
Figure 5. The impact of EPAC2 on RSV infection. (A,B) A549 cells in triplicate were infected with RSV at an MOI of 1. After 2 h p.i., cells were treated with DMSO, 20 μM CE3F4, 20 μM ESI-05, or 20 μM MAY0132. At 15 h post-treatment, (A) total viruses were collected, and viral titer was determined by immunostaining using an anti-RSV antibody. (B) The supernatant was collected, and diluted or undiluted cytokines/chemokines were measured by Bio-Plex. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (C,D) SAE cells were infected with RSV and treated with the indicated drugs as described in (A). At 15 h post-treatment, (C) virus titration was determined. ** p < 0.01, relative to the DMSO-treated group. (D) Cytokines/chemokines of SAE cells were also measured by Bio-Plex. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated group. (EG) A549 cells in triplicate were infected with RSV and treated with drugs as described in (A). At 4 h post-treatment, total RNA was extracted and subjected to qRT-PCR to measure RSV genome copies (E), N gene expression (F), and IP-10 transcription (G). DMSO-treated mock infection was used as a negative control for DMSO-treated RSV infection. * p < 0.05 and ** p < 0.01, relative to the DMSO-treated and RSV-infected group.
Viruses 13 01179 g005
Figure 6. The EPAC2-mediated NF-κB activation by poly I:C or TNF-α. (A,B) 293 cells in triplicate were transiently transfected with a luciferase reporter plasmid containing the NF-κB promoter (NF-κB-luc) in the presence of the indicated drugs. At 15 h post-transfection, cells were transfected with 1 μg/mL poly I:C (A) or treated with 10 ng/mL of TNF-α (B) for an additional 6 h. Cells were then lysed to measure luciferase activities. Cells without poly I:C or TNF-α treatment were used as a negative control. Data are representative of three independent experiments. * p < 0.05 and ** p < 0.01, relative to the poly I:C-transfected and DMSO-treated group or the TNF-α- and DMSO-treated group, respectively.
Figure 6. The EPAC2-mediated NF-κB activation by poly I:C or TNF-α. (A,B) 293 cells in triplicate were transiently transfected with a luciferase reporter plasmid containing the NF-κB promoter (NF-κB-luc) in the presence of the indicated drugs. At 15 h post-transfection, cells were transfected with 1 μg/mL poly I:C (A) or treated with 10 ng/mL of TNF-α (B) for an additional 6 h. Cells were then lysed to measure luciferase activities. Cells without poly I:C or TNF-α treatment were used as a negative control. Data are representative of three independent experiments. * p < 0.05 and ** p < 0.01, relative to the poly I:C-transfected and DMSO-treated group or the TNF-α- and DMSO-treated group, respectively.
Viruses 13 01179 g006
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Choi, E.-J.; Wu, W.; Cong, X.; Zhang, K.; Luo, J.; Ye, S.; Wang, P.; Suresh, A.; Ullah, U.M.; Zhou, J.; et al. Broad Impact of Exchange Protein Directly Activated by cAMP 2 (EPAC2) on Respiratory Viral Infections. Viruses 2021, 13, 1179. https://0-doi-org.brum.beds.ac.uk/10.3390/v13061179

AMA Style

Choi E-J, Wu W, Cong X, Zhang K, Luo J, Ye S, Wang P, Suresh A, Ullah UM, Zhou J, et al. Broad Impact of Exchange Protein Directly Activated by cAMP 2 (EPAC2) on Respiratory Viral Infections. Viruses. 2021; 13(6):1179. https://0-doi-org.brum.beds.ac.uk/10.3390/v13061179

Chicago/Turabian Style

Choi, Eun-Jin, Wenzhe Wu, Xiaoyan Cong, Ke Zhang, Jiaqi Luo, Sha Ye, Pingyuan Wang, Adarsh Suresh, Uneeb Mohammad Ullah, Jia Zhou, and et al. 2021. "Broad Impact of Exchange Protein Directly Activated by cAMP 2 (EPAC2) on Respiratory Viral Infections" Viruses 13, no. 6: 1179. https://0-doi-org.brum.beds.ac.uk/10.3390/v13061179

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop