Next Article in Journal
Polymorphisms Analysis of BMP15, GDF9 and BMPR1B in Tibetan Cashmere Goat (Capra hircus)
Previous Article in Journal
ACE-I/D Allele Modulates Improvements of Cardiorespiratory Function and Muscle Performance with Interval-Type Exercise
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Molecular Profiling of Inflammatory Mediators in Human Respiratory Syncytial Virus and Human Bocavirus Infection

by
Noorah A. Alkubaisi
1,
Ibrahim M. Aziz
1,
Asma N. Alsaleh
1,
Abdulkarim F. Alhetheel
2 and
Fahad N. Almajhdi
1,*
1
Department of Botany and Microbiology, College of Science, King Saud University, Riyadh 11451, Saudi Arabia
2
Department of Pathology and Laboratory Medicine, College of Medicine, King Saud University, Riyadh 11451, Saudi Arabia
*
Author to whom correspondence should be addressed.
Genes 2023, 14(5), 1101; https://doi.org/10.3390/genes14051101
Submission received: 11 April 2023 / Revised: 13 May 2023 / Accepted: 14 May 2023 / Published: 17 May 2023
(This article belongs to the Section Viral Genomics)

Abstract

:
Infections due to human respiratory syncytial virus (HRSV) and human bocavirus (HBoV) can mediate the release of several pro-inflammatory cytokines such as IL-6, IL-8, and TNF-α, which are usually associated with disease severity in children. In this study, the change in the expression profile of cytokines and chemokines were determined during HRSV, HBoV, and HRSV coinfection with HBoV in 75 nasopharyngeal aspirates (NPAs) samples, positive real-time reverse transcriptase PCR Assay (rRT-PCR) for HRSV (n = 36), HBoV (n = 23) infection alone or HRSV coinfection with HBoV (n = 16). The samples were collected from hospitalized children. qPCR-based detection revealed that the levels of IL-6, IL-8, IL-10, IL-13, IL-33, and G-CSF were significantly (p < 0.05) greater in patients than in controls. IL-4, IL-17, GM-CSF, and CCL-5 were significantly elevated in children with HRSV coinfection with HBoV than in other groups (p < 0.05). TNF-α, IL-6, IL-8, IL-10, IL-13, and IL-33 in children with HRSV were significantly increased in severe infections compared to mild infections. Whereas, IL-10, IL-13, and IL-33 were significantly increased in severe infection in compared a mild infection in children with HBoV. Further large-scale investigations involving isolates are needed to enhance our knowledge of the association between viral infections and cytokine expression patterns during the different stages of HRSV and HBoV infection.

1. Introduction

The majority of lower respiratory tract infections (LRTI) in children worldwide are caused by the human respiratory syncytial virus (HRSV), formerly known as the human orthopneumovirus (HOPV) [1,2]. HRSV is a member of the genus Orthopneumovirus within the family Pneumoviridae, and order Mononegavirales [3]. HRSV is an enveloped virus that consists of a negative-sense single-stranded RNA genome with a 15.2-kb size. The genome contains 10 genes encoding 11 proteins that some of which have a significant role in both pathogenicity and classification [4]. The attachment protein (G), for instance, plays an important role in viral entry to the host. Based on the antigenic variations observed in it along with its interactions with monoclonal antibodies (mAbs), circulating viruses have been classified into two antigenic subgroups (A and B) [5,6]. As reported widely, both HRSV-A and B can co-circulate during an epidemic year with a slight predominance of type A [7]. Our group has previously shown that a similar pattern is occurring in Saudi Arabia. Both subtypes, HRSV-A and B, were circulating in some epidemic years with a slight predominance of HRSV-A [8,9,10]. Another etiological agent for LRTIs in infants and children is human bocavirus (HBoV). The virus, which was first identified in September 2005, was frequently detected in respiratory samples that were collected from children with acute wheezing episodes [11]. HBoV is a putative member of the family Parvoviridae, subfamily Parvovirinae, genus Bocavirus, that is spherical in shape, with icosahedral symmetry and non-enveloped nucleocapsid [11,12]. The virus is single-stranded DNA genome comprises three open reading frames (ORFs), the first two of which are nuclear phosphoprotein (NP1) and non-structural protein 1 (NS1), and the third of which is viral capsid proteins 1 and 2 (VP1 and VP2) [12]. There are four known human genotypes of this virus: type HBoV (1 to 4). HBoV1 is associated with LRTIs and, while the three additional genotypes HBoV2, 3, and 4 are associated with gastroenteritis and primarily seen in stools [13].
In response to localized inflammation infected by the virus, cytokines, such as interleukin-6 (IL-6), IL-8, tumor necrosis factor-α (TNF-α), CXCL2, and CXCL10 produced by various cell types in response to invading virus to promote the mobilization of macrophages and DCs can be linked to the severity of the illness in children [14,15,16]. It has been shown that children with HRSV ARTI have higher levels of pro-inflammatory cytokines including IL-4, IL-6, IL-10, and IL-13 [17,18,19,20,21]. According to another study, compared to influenza A virus infections, the concentrations of IL-4, IL-5, and CCL-5 are higher during HRSV with LRTI [22]. Furthermore, high levels of IL-6, IL-8, and IL-10 levels were correlated with disease severity in HRSV LRTI, the severity was determined by the requirement for supplemental oxygen [20,23,24]. The levels of IL-6, IL-1β, and IL-8 were also shown to be increased in patients infected with HRSV only or with both HRSV and human rhinovirus (HRV) [25]. Whereas, of HBoV-infected individuals TNF-a, IL-2, IL-5 IL-8, and IL-10 were found to be dramatically higher in HBoV-positive children in the acute stage of respiratory infection. [26,27].
Typically, cytokines have been evaluated by serological-based approaches including enzyme-linked immunosorbent assay (ELISA), Luminex, and cytometric bead array (CBA) [28]. However, cytokine mRNA quantification is widely used to investigate cytokine profiles via molecular-based assays, particularly in small samples. In comparison with serological-based approaches, real-time reverse transcriptase polymerase chain reaction (rRT-PCR) is the method of choice for rapid and reproducible, and flexibility to measure cytokine from cells, tissues, or tissue biopsies [29,30,31,32]. Therefore, the primary purpose of the current study is to identify the change in inflammatory mediators during HRSV, HBoV, and HRSV coinfection with HBoV.

2. Materials and Methods

2.1. Study Population

A total of 75 nasopharyngeal aspirate samples (NPAs) were previously identified using rRT-PCR assay by King Khalid University Hospital (KKUH) (36 positives HRSV, 23 positives HBoV and 16 HRSV coinfection with HBoV). These samples were collected from infants and young children less than 5 years’ old. The patients were admitted to KKUH in Riyadh, Saudi Arabia during the winter seasons of 2019/20 and 2021/22 with acute respiratory symptoms including such as fever, dry cough, and fatigue. The number of days of supplementary oxygen was used to determine the severity for the purposes of this study. Patients in hospitals who required less than two days of oxygen supplementation were considered to have a severe illness. Patients hospitalized with 1 day of oxygen supplements or with no shortness of breath were classified as having a mild infection. Children who were reported positive for any other respiratory viruses were not included in the study. The healthy control NPAs samples (n = 25) were collected from infants without ALRTI symptoms and age-matched children of whom NPA samples were negative for HRSV and HBoV using rRT-PCR.
Control samples (n = 25) were collected from a group of healthy, asymptomatic age-matched children of whom NPA samples were negative for HRSV and HBoV using rRT-PCR. Clinical samples were collected following the protocols approved by the institutional review board (IRB) at King Khalid University Hospital, Riyadh (ref, No. 4/67/352665/IRB).

2.2. Virus Studies

2.2.1. Viral RNA Extraction

QIAamp Viral RNA Extraction Kit (Qiagen, Hilden, Germany) was used to extract viral RNA from clinical samples. HRSV and HboV were confirmed using one-step reverse transcriptase polymerase chain reaction (RT-PCR) Kit (Qiagen, Hilden, Germany) with the following set of primers forward primer HRSV-U-F (5′-GGAACAAGTTGTTGAGGTTTATGAATATGC-3′) and reverse primer HRSV-U-R (5′-CTTCTGCTGTCAAGTCTAGTACACTGTAGT-3′) [33]. 75 nasopharyngeal aspirate samples For the detection of HBoV, the same RT-PCR kit was used with the HboV-specific primer set: Boca-VP/NC-F (5′-AGCTGTGAGATTGTATGGGAAG-3′) and PanBoca-R (5′-AAAACAGCTCCCCCCACAAT-3′), the final 1/10 of the VP1 gene’s 3′ and the last 1/4 of the 3′ of the non-coding regions were targeted using this primer set, which flanks a 378-bp highly conserved sequence [34]. The reaction was performed in a GeneAmp 9700 thermal cycler (GeneAmp PCR system 9700, Applied Biosystems, Waltham, Massachusetts, United States) using the following cycling conditions: reverse transcription step at 50 °C for 30 min followed by initial denaturation at 95 °C for 15 min then 35 cycles of denaturation at 94 °C for 30 s, annealing at 52 °C for 30 s, and extension at 72 °C for 2 min; and a final extension step of 72 °C for 10 min. The amplified products were compared to a 100 bp DNA ladder (Qiagen, Hilden, Germany) and observed on an agarose gel stained with 1% ethidium-bromide.

2.2.2. Quantitative Real-Time PCR

RNeasy® Mini Kit (Qiagen, Hilden, Germany) was used to extract total RNA from clinical samples in accordance with the manufacturer’s instructions. The quantification of cytokines and chemokines in NPAs was determined by performing a quantitative qPCR using one step RT² SYBR® Green/ROX™ qPCR Master Mix. The reactions were carried out in 7500 Fast real-time PCR (7500 Fast; Applied Biosystems, Waltham, MA, USA) using the following cycling conditions: reverse transcription at 37 °C for 15 min, initial PCR denaturation at 95 °C for 10 min, followed by annealing at 60 °C for 30 s, and extension at 72 °C for 30 s. The data were expressed as mean fold changes ± standard error for three independent amplifications. The sequences of the utilized primers used in PCR are presented in Table S1 [35,36,37,38,39,40,41,42,43,44,45,46,47,48,49].

2.2.3. Analysis of Real-Time PCR Array Data

All arrays were run under the same conditions and Ct values were obtained using a constant baseline threshold for all PCR runs. All samples were analyzed in duplicate at least three separate experiments. Melting curve analysis was done to confirm the specificity of amplification and the lack of primer dimers. The results were obtained using the 2−ΔΔCq method [50], and delta Cq (ΔCq) values obtained for the different genes were normalized based on the value of GAPDH amplified from the same genes and the fold-change in expression was calculated as referenced to the expression of the control (non-infected cells or negative control).

2.3. Statistical Analysis

The non-parametric one-way analysis of variance (ANOVA) with posthoc Dennet’s test was used for data analysis). Categorical data were presented as frequencies and percentages (%). Non-Gaussian variables were presented as the median. Independent Sample t-test and The Chi-Square independence test were used to indicate differences as all variables are nominal. The results with p < 0.05 were considered significant.

3. Results

3.1. General Patient Characteristics

Infants under one year of age were found to be the age group most affected, accounting for 44.5%, 65.2%, and 50.0% of all cases of HRSV, HBoV, and HRSV coinfection with HBoV, respectively. Based on gender, HRSV with 61.1% and HBoV with 60.9% were predominant in males than in females. Based on severity were found 55.6%, 78.3% of HRSV and HBoV (Table 1).

3.2. Cytokine and Chemokine Profile

IL-6, IL-8, IL-10, IL-13, IL-33, and G-CSF in the NPA were significantly greater (p < 0.05) in children with HRSV, HBoV and HRSV coinfection with HBoV than in controls (Table 2).
(Table 3) showed that children with HRSV had significantly higher levels of IL-4, IL-13, IL-17, GM-CSF, and CCL-5 (p < 0.05) than children with HBoV alone or HRSV coinfection with HBoV, whereas, children with HBoV infection had significantly greater levels of TNF-α, IL-2, IL-6, IL-8, IL-10, IL-33, and CCL-3 compare to HRSV alone or HRSV coinfection with HBoV. IL-4, IL-17, GM-CSF, and CCL-5 were significantly elevated (p < 0.05) in children with HRSV coinfection with HBoV than HRSV or HBoV alone.
We next compared the proinflammatory cytokines and chemokines markers in the NPAs among children with HRSV and/or HBoV and disease severity. As depicted in (Figure 1), TNF-α, IL-6, IL-8, IL-10, IL-13, and IL-33 in children with HRSV were significantly increased in severe infection compared to mild infection (p < 0.05). Whereas, IL-10, IL-13, and IL-33 were significantly (p < 0.05) increased in severe infection compared to a mild infection in children with HBoV. TNF-α, IL-10, IL-13, and IL-33 were significantly (p < 0.05) increased in severe infection comparison to a mild infection in children with HRSV coinfection with HBoV.

4. Discussion

HRSV is the major cause of LRTI hospitalizations in young children, immunocompromised adults, and the elderly. Currently, there is no effective treatment or vaccine available [51]. Several pro-inflammatory cytokines can be released by innate immune cells during severe lung responses to RSV infection leading to lung immunopathology [52]. Concurrently, pro-inflammatory cytokines (e.g., CXCL8, CCL-3, CCL-2, and CCL5) and chemokines (e.g., IL-6, IL-8, IL-33, TNF-) are induced and secreted to the extracellular medium, which can be related to the severity of the disease in children. Therefore, the pro-inflammatory cytokines mediator may represent a strategy that contributes to HRSV pathogenesis and is associated with the disease severity of respiratory viral infections in children. In this report, the pro-inflammatory cytokines elicited by HRSV and HBoV were analyzed. In addition, to identify the correlation between these biomarkers and the development of lung fibrosis and the severity of immunopathological responses and comparison of pulmonary and in vitro gene expression.
Infants under one year of age were found to be the age group most affected, comprising 44.5% of all cases of HRSV. Males had a higher prevalence of HRSV than females according to gender. According to our earlier research, HRSV-A was somewhat more prevalent than HRSV-B between 2008 and 2016 in Saudi Arabia, causing lower respiratory tract infections in hospitalized children at a rate of 19.3 to 45.4% [8,9,10,53]. A similar predominance of HRSV-A over HRSV-B was reported earlier in Germany [54], Spain [55], France [56], Kuwait [57], and Lebanon [58]. Infants < 6 months of age were found to be the age group most affected age group, accounting for 59.32% of all cases. According to gender, men (18%) had more cases of HRSV than women (7%) [10].
The age group most afflicted, accounting for 59.32% of all cases, was infants under 6 months old. Males (18%) had a higher prevalence of HRSV than females (7%) when comparing the two genders.
In this study, qPCR-based detection revealed that the levels of IL-6, IL-8, IL-10, IL-13, IL-33, and G-CSF were significantly (p < 0.05) greater in children with HRSV, HBoV and HRSV coinfection with HBoV than in controls. TNF-α, IL-6, IL-8, IL-10, IL-13, and IL-33 in children with HRSV were significantly increased in severe infection compared to mild infection (p < 0.05) in children with HRSV. Children with HRSV have higher amounts of IL-8, IFN-a, IL-6, IL-3, IL-33, IL-1a, IL-2, IL-22, G-CSF, CCL-2, and CCL-3, which were associated with children’s disease severity [59,60,61]. According to Sheeran et al., all children with HRSV infection had significantly higher levels of IL-6, IL-8, and IL-10 in their nasal wash and tracheal aspirate samples than did control children [62]. Another study indicated that patient NPAs had higher levels of cytokines (IL-1, IL-2, IL-12, IFN-, TNF-a, IL-13, IL-4, IL-6, IL-10), chemokines (IP-10, IL-8, MIP1-a, MIP-1), growth factors (FGFb, PDGFbb, G-CSF), and cytokines (IL-1RA, IL-17) in nasal washes [18]. Further, early research also revealed elevated IL-4 levels, particularly in very young newborns, during RSV infection. However, another study found that only children with acute RSV-induced bronchiolitis had significantly elevated levels of IL-l0, but not IL-12 or IFN, possibly explaining the ineffectiveness of the cell-mediated immune response. According to other studies, nasal washes include high levels of IL-4, IL-6, IL-9, IL-10, and IL-13 [63,64,65], and in the lung in children with HRSV LRTI [66].
The current study also reported that IL-10, IL-13, and IL-33 were significantly increased in severe infection compared to mild infection in children with HBoV. Early study reported that cytokines including GM-CSF, Leptin, IL-1β, TNF-α, IL-8, IL-16, CCL-5, and IL-3 were clearly upregulated in the HBoV-positive cohort [67]. This study also revealed that IL-10 and IL-33 were markedly elevated in children with HBoV as well as with HRSV coinfection with HBoV coinfection. Further, IL-4, IL-17, GM-CSF, and CCL-5 were significantly elevated in children with HRSV coinfection with HBoV than in other groups (p < 0.05). Early research revealed that two or more respiratory viruses are co-infected in 10–30% of individuals under age 5 years [68,69,70]. According to a previous investigation, patients with HRSV coinfection with HBoV had greater rates of pneumonia than those with HBoV or HRSV infection alone [71].
IL-6 is a soluble mediator that is produced by macrophages and epithelial cells that has a pleiotropic influence on inflammation and immune response [72,73]. IL-6 is frequently associated with innate and adaptive immunity and regulates antimicrobial defense by promoting the differentiation of naive CD4+ and CD8+ T cells. During HRSV infections, Tabarani et al. reported that the level of IL-6 was evaluated in NPAs samples from children with HRSV. It’s interesting to note that among other inflammatory mediators (IFN-α, CCL3, CCL-4, and CCL-2), they have discovered an association between the severity of the clinical manifestations elicited by HRSV infection and high levels of IL-6 [74]. However, Brown et al. have proposed that high plasma levels of IL-6 could suggest a higher likelihood of infant hospitalization and severe bronchiolitis caused by HRSV [75]. Other studies demonstrated a positive correlation between high levels of IL-6 found in the NPAs of HRSV-infected patients and more severe forms of the disease, and that at the period of hospital admission, the levels of IL-6 in the serum could be used as indicators of the severity in patients with HRSV-caused LRTI [24,59].
The chemotactic factor known as IL-8 is known to be released by activated monocytes and macrophages and to encourage the directional migration of neutrophils, basophils, and T lymphocytes [76]. Disease severity caused by HRSV infection has been closely correlated with elevated IL-8 levels in NPAs samples, including the risk of mechanical ventilation [75,77]. Significantly, children’s higher NPA IL-6 and IL-8 levels have been associated with more severe HRSV infections [78]. Another study reported that the elevated concentrations of IL-8 in NPAs washes were associated with severe HRSV infection [74]. Similarly, the levels of IL-8 and CCL-5 were elevated in plasma and NPAs with disease severity in HRSV-infected children as compared to children with mild or moderate disease [79].
IL-33 is a member of the IL-1 family, which is constitutively produced from the structural and lining cells including fibroblasts, endothelial cells, and epithelial cells of the skin, gastrointestinal tract, and lungs that are exposed to the environment [80]. In respiratory tract epithelia, the main function of this cytokine is involved in early immune and inflammatory responses following HRSV infection. IL-33 is constitutively expressed by endothelial and epithelial cells. Unlike other interleukin 1 family members, IL-33 has been associated with the initiation and development of the innate and adaptive Th2-type immune response [81]. Clinically, patients with mild to moderately severe asthma show increased nasal IL-33 levels following HRSV inoculation and this increase correlates with symptom severity and viral load [82].
It is believed that IL-5 and IL-13 play important roles in the etiology of asthma, with IL-13 acting on a different pathway from IL-5 to cause airway hyperreactivity (AHR) in the allergic lung [83]. Importantly, IL-13 is generated and secreted by a variety of cells, including alveolar macrophages, basophils, mast cells, eosinophils, and CD4+ T cells, in response to IL-33 signaling. IL-33 directly stimulates in response to IL-33 signaling produced and secreted through various cells, including alveolar macrophages, and basophils [81]. Infants hospitalized with HRSV-caused bronchiolitis had high levels of IL-13 and IL-33, which were associated with the need for ventilation [65].
We acknowledge the shortcomings and limitations in terms of the methodology we used in our present study. Since it is easier to measure mRNA levels across the entire genome than protein levels, mRNA expression levels are frequently used as a proxy for estimating functional differences that occur at the protein levels [84]. Therefore, further work should be measured on HRSV and/or HBoV at protein levels of proinflammatory cytokines and chemokines markers for disease severity. In addition, rRT-PCR, which is becoming the method used for respiratory diagnosis, yields result as cycle threshold (CT) values, which is a semi-quantitative value that can broadly categorize the concentration of viral genetic material in a patient sample, therefore, further research is required to examine the relationship between viral load or CT values and inflammatory markers. Since in vivo study is not sufficient for proving the induction of chemokines and cytokines by HRSV and/or HBoV, in vitro model is required to support the in vivo observations.

5. Conclusions

In conclusion, this is an excellent and very promising study and really well done to identify the change in inflammatory mediators during HRSV, HBoV, and HRSV coinfection with HBoV. Results from this study revealed that qPCR-based detection revealed that the levels of IL-6, IL-8, IL-10, IL-13, IL-33, and G-CSF were significantly (p < 0.05) greater in children with HRSV, HBoV, and HRSV coinfection with HBoV than in controls. TNF-α, IL-6, IL-8, IL-10, IL-13, and IL-33 in children with HRSV were significantly increased in severe infections compared to mild infections. Whereas, IL-10, IL-13, and IL-33 were significantly increased in severe infection compared to mild infection in children with HBoV. IL-4, IL-17, GM-CSF, and CCL-5 were significantly elevated in children with HRSV coinfection with HBoV than in other groups (p < 0.05). Further investigations including patients during the various stages of HRSV infection are needed to understanding our knowledge of the biological impact of pro-inflammatory cytokine and cytokine expression patterns.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/genes14051101/s1, Table S1. Primers for cytokines and chemokines detection.

Author Contributions

Conceptualization, N.A.A. and I.M.A.; methodology, N.A.A.; software, N.A.A. and I.M.A.; validation, N.A.A., I.M.A. and F.N.A.; formal analysis, A.F.A.; investigation, F.N.A.; resources, F.N.A.; data curation, N.A.A. and I.M.A.; writing original draft preparation, N.A.A. and I.M.A.; writing review and editing, A.N.A. and F.N.A.; review and editing, visualization, I.M.A.; supervision, F.N.A.; project administration, F.N.A.; funding acquisition, F.N.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

The study was conducted in accordance with the Declaration of Helsinki and approved by Research Ethics Committee (RES) at the King Saud University, Riyadh, Saudi Arabia (Ethics Reference No. 4/67/352665/IRB).

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Data Availability Statement

All the data is provided.

Acknowledgments

The authors thank the Researchers Supporting Project number (RSPD2023R801), King Saud University, Riyadh, Saudi Arabia.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Mazur, N.I.; Martinón-Torres, F.; Baraldi, E.; Fauroux, B.; Greenough, A.; Heikkinen, T.; Manzoni, P.; Mejias, A.; Nair, H.; Papadopoulos, N.; et al. Lower respiratory tract infection caused by respiratory syncytial virus: Current management and new therapeutics. Lancet Respir. Med. 2015, 3, 888–900. [Google Scholar] [CrossRef] [PubMed]
  2. Mazur, N.I.; Bont, L.; Cohen, A.L.; Cohen, C.; Von Gottberg, A.; Groome, M.J.; Hellferscee, O.; Klipstein-Grobusch, K.; Mekgoe, O.; Naby, F.; et al. Severity of respiratory syncytial virus lower respiratory tract infection with viral coinfection in HIV-uninfected children. Clin. Infect. Dis. 2017, 64, 443–450. [Google Scholar] [CrossRef] [PubMed]
  3. Afonso, C.L.; Amarasinghe, G.K.; Bányai, K.; Bào, Y.; Basler, C.F.; Bavari, S.; Bejerman, N.; Blasdell, K.R.; Briand, F.-X.; Briese, T.; et al. Taxonomy of the order Mononegavirales: Update 2016. Arch. Virol. 2016, 161, 2351–2360. [Google Scholar] [CrossRef]
  4. Ogunsemowo, O.; Olaleye, D.O.; Odaibo, G.N. Genetic diversity of human respiratory syncytial virus circulating among children in Ibadan, Nigeria. PLoS ONE 2018, 13, e0191494. [Google Scholar] [CrossRef]
  5. Sullender, W.M. Respiratory syncytial virus genetic and antigenic diversity. Clin. Microbiol. Rev. 2000, 13, 1–15. [Google Scholar] [CrossRef] [PubMed]
  6. Obodai, E.; Odoom, J.K.; Adiku, T.; Goka, B.; Wolff, T.; Biere, B.; Chweiger, B.; Reiche, J. The significance of human respiratory syncytial virus (HRSV) in children from Ghana with acute lower respiratory tract infection: A molecular epidemiological analysis, 2006 and 2013–2014. PLoS ONE 2018, 13, e0221315. [Google Scholar] [CrossRef] [PubMed]
  7. Esposito, S.; Piralla, A.; Zampiero, A.; Bianchini, S.; Di Pietro, G.; Scala, A.; Pinzani, R.; Fossali, E.; Baldanti, F.; Principi, N.; et al. Characteristics and their clinical relevance of respiratory syncytial virus types and genotypes circulating in Northern Italy in five consecutive winter seasons. PLoS ONE 2015, 10, e0129369. [Google Scholar] [CrossRef] [PubMed]
  8. Al-Majhdi, F.N.; Al-Jarallah, A.; Elaeed, M.; Latif, A.; Gissmann, L.; Amer, H.M. Prevalence of respiratory syncytial virus infection in Riyadh during the winter season 2007–2008 and different risk factors impact. Int. J. Virol. 2009, 5, 154–163. [Google Scholar] [CrossRef]
  9. Almajhdi, F.N.; Farrag, M.A.; Amer, H.M. Genetic diversity in the G protein gene of group A human respiratory syncytial viruses circulating in Riyadh, Saudi Arabia. Arch. Virol. 2014, 159, 73–81. [Google Scholar] [CrossRef] [PubMed]
  10. Farrag, M.A.; Amer, H.M.; Aziz, I.M.; Alsaleh, A.N.; Almajhdi, F.N. The emergence of subgenotype ON-1 of Human orthopneumovirus type A in Riyadh, Saudi Arabia: A new episode of the virus epidemiological dynamic. J. Med. Virol. 2020, 92, 1133–1140. [Google Scholar] [CrossRef]
  11. Allander, T.; Tammi, M.T.; Eriksson, M.; Bjerkner, A.; Tiveljung-Lindell, A.; Andersson, B. Cloning of a human parvovirus by molecular screening of respiratory tract samples. Proc. Natl. Acad. Sci. USA 2005, 102, 12891–12896. [Google Scholar] [CrossRef] [PubMed]
  12. Cashman, O.; O’Shea, H. Detection of human bocaviruses 1, 2 and 3 in Irish children presenting with gastroenteritis. Arch. Virol. 2012, 157, 1767–1773. [Google Scholar] [CrossRef] [PubMed]
  13. Abdelqader, R.; Hasan, H.; Alanagreh, L. Epidemiology of Human Bocavirus in the Middle East and North Africa: Systematic Review. Pathogens 2021, 10, 1456. [Google Scholar] [CrossRef] [PubMed]
  14. Yui, I.; Hoshi, A.; Shigeta, Y.; Takami, T.; Nakayama, T. Detection of human respiratory syncytial virus sequences in peripheral blood mononuclear cells. J. Med. Virol. 2003, 70, 481–489. [Google Scholar] [CrossRef]
  15. de Graaff, P.M.; de Jong, E.C.; van Capel, T.M.; van Dijk, M.E.; Roholl, P.J.; Boes, J.; Luytjes, W.; Kimpen, J.L.; van Bleek, G.M. Respiratory syncytial virus infection of monocyte-derived dendritic cells decreases their capacity to activate CD4 T cells. J. Immunol. 2005, 175, 5904–5911. [Google Scholar] [CrossRef] [PubMed]
  16. Zidek, Z.; Anzenbacher, P.; Kmoníčková, E. Current status and challenges of cytokine pharmacology. Br. J. Pharmacol. 2009, 157, 342–361. [Google Scholar] [CrossRef]
  17. Chung, H.L.; Park, H.J.; Kim, S.Y.; Kim, S.G. Age-related difference in immune responses to respiratory syncytial virus infection in young children. Pediatr. Allergy Immunol. 2007, 18, 94–99. [Google Scholar] [CrossRef] [PubMed]
  18. Bermejo-Martin, J.F.; Garcia-Arevalo, M.C.; De Lejarazu, R.O.; Ardura, J.; Eiros, J.M.; Alonso, A.; Matías, V.; Pino, M.; Bernardo, D.; Arranz, E.; et al. Predominance of Th2 cytokines, CXC chemokines and innate immunity mediators at the mucosal level during severe respiratory syncytial virus infection in children. Eur. Cytokine Netw. 2007, 18, 162–167. [Google Scholar]
  19. Hassan, M.A.; Eldin, A.M.; Ahmed, M.M. T–helper2/T–helper1 imbalance in respiratory syncytial virus bronchiolitis in relation to disease severity and outcome. Egypt J. Immunol. 2008, 15, 153–160. [Google Scholar]
  20. Diaz, P.V.; Pinto, R.A.; Mamani, R.; Uasapud, P.A.; Bono, M.R.; Gaggero, A.A.; Guerrero, J.; Goecke, A. Increased expression of the glucocorticoid receptor β in infants with RSV bronchiolitis. Pediatrics 2012, 130, e804–e811. [Google Scholar] [CrossRef]
  21. Ye, Q.; Shao, W.X.; Shang, S.Q.; Pan, Y.X.; Shen, H.Q.; Chen, X.J. Epidemiological characteristics and immune status of children with Respiratory Syncytial Virus. J. Med. Virol. 2015, 87, 323–329. [Google Scholar] [CrossRef] [PubMed]
  22. Sung, R.Y.; Hui, S.H.; Wong, C.K.; Lam, C.W.; Yin, J. A comparison of cytokine responses in respiratory syncytial virus and influenza A infections in infants. Eur. J. Pediatr. 2001, 160, 117–122. [Google Scholar] [CrossRef] [PubMed]
  23. Alonso Fernández, J.; Roine, I.; Vasquez, A.; Cáneo, M. Soluble interleukin-2 receptor (sCD25) and interleukin-10 plasma concentrations are associated with severity of primary respiratory syncytial virus (RSV) infection. Eur. Cytokine Netw. 2005, 16, 81–90. [Google Scholar] [PubMed]
  24. Vieira, R.A.; Diniz, E.M.; Ceccon, M.E. Correlation between inflammatory mediators in the nasopharyngeal secretion and in the serum of children with lower respiratory tract infection caused by respiratory syncytial virus and disease severity. J. Bras. Pneumol. 2010, 36, 59–66. [Google Scholar] [CrossRef]
  25. Díaz, P.V.; Valdivia, G.; Gaggero, A.A.; Bono, M.; Zepeda, G.; Rivas, M.; Uasapud, P.; Pinto, R.A.; Boza, M.L.; Guerrero, J. Pro-inflammatory cytokines in nasopharyngeal aspirate from hospitalized children with respiratory syncytial virus infection with or without rhinovirus bronchiolitis, and use of the cytokines as predictors of illness severity. Medicine 2015, 94, e1512. [Google Scholar] [CrossRef]
  26. Kumar, A.; Filippone, C.; Lahtinen, A.; Hedman, L.; Söderlund-Venermo, M.; Hedman, K.; Franssila, R. Comparison of Th-cell immunity against human bocavirus and parvovirus B19: Proliferation and cytokine responses are similar in magnitude but more closely interrelated with human bocavirus. Scand. J. Immunol. 2011, 73, 135–140. [Google Scholar] [CrossRef]
  27. Hirose, Y.; Hamada, H.; Wakui, T.; Ogawa, T.; Terai, M. Characteristic systemic cytokine responses in children with human bocavirus-positive lower respiratory tract infection. Microbiol. Immunol. 2014, 58, 215–218. [Google Scholar] [CrossRef]
  28. Chiswick, E.L.; Duffy, E.; Japp, B.; Remick, D. Detection and quantification of cytokines and other biomarkers. Methods Protoc. 2012, 844, 15–30. [Google Scholar]
  29. Yin, J.L.; Shackel, N.A.; Zekry, A.; McGuinness, P.H.; Richards, C.; Van Der Putten, K.; McCaughan, G.W.; Eris, J.M.; Bishop, G.A. Real-time reverse transcriptase–polymerase chain reaction (RT–PCR) for measurement of cytokine and growth factor mRNA expression with fluorogenic probes or SYBR Green I. Immunol. Cell Biol. 2001, 79, 213–221. [Google Scholar] [CrossRef] [PubMed]
  30. Overbergh, L.; Giulietti, A.; Valckx, D.; Decallonne, B.; Bouillon, R.; Mathieu, C. The use of real-time reverse transcriptase PCR for the quantification of cytokine gene expression. J. Biomol. Tech. 2003, 14, 33. [Google Scholar]
  31. Ramos-Payán, R.; Aguilar-Medina, M.; Estrada-Parra, S.; González-y-Merchand, J.; Favila-Castillo, L.; Monroy-Ostria, A.; Estrada-Garcia ICE. Quantification of cytokine gene expression using an economical real-time polymerase chain reaction method based on SYBR® green I. Scand. J. Immunol. 2003, 57, 439–445. [Google Scholar] [CrossRef] [PubMed]
  32. Noh, I.C.; Avoi, R.; Nurul, A.A.; Ahmad, I.; Bakar, R.A. Analysis of serum and gene expression profile of cytokines (IL-6, TNF-α and TGF-β1) in chronic hepatitis C virus infection. PeerJ 2022, 10, e13330. [Google Scholar]
  33. Cane, P.; Pringle, C. Respiratory syncytial virus heterogeneity during an epidemic: Analysis by limited nucleotide sequencing (SH gene) and restriction mapping (N gene). J. Gen. Virol. 1991, 72, 349–357. [Google Scholar] [CrossRef] [PubMed]
  34. Abdel-Moneim, A.S.; EMahfouz, M.; Zytouni, D.M. Detection of human bocavirus in Saudi healthy blood donors. PLoS ONE 2018, 13, e0193594. [Google Scholar] [CrossRef]
  35. Munk, R.B.; Sugiyama, K.; Ghosh, P.; Sasaki, C.Y.; Rezanka, L.; Banerjee, K.; Takahashi, H.; Sen, R.; Longo, D.L. Antigen-independent IFN-γ production by human naive CD4+ T cells activated by IL-12 plus IL-18. PLoS ONE 2011, 6, e18553. [Google Scholar] [CrossRef]
  36. Carneiro, J.R.M.; Fuzii, H.T.; Kayser, C.; Alberto, F.L.; Soares, F.A.; Sato, E.I.; Andrade LEC. IL-2, IL-5, TNF-α and IFN-γ mRNA expression in epidermal keratinocytes of systemic lupus erythematosus skin lesions. Clinics 2011, 66, 77–82. [Google Scholar] [CrossRef] [PubMed]
  37. Huynh, N.C.; Everts, V.; Pavasant, P.; Ampornaramveth, R.S. Interleukin-1β induces human cementoblasts to support osteoclastogenesis. Int. J. Oral Sci. 2017, 9, e5. [Google Scholar] [CrossRef]
  38. Stott, B.; Lavender, P.; Lehmann, S.; Pennino, D.; Durham, S.; Schmidt-Weber, C.B. Human IL-31 is induced by IL-4 and promotes TH2-driven inflammation. J. Allergy Clin. Immunol. 2013, 132, 446–454.e445. [Google Scholar] [CrossRef]
  39. Ansari, N.A.; Ramesh, V.; Salotra, P. Interferon (IFN)–γ, tumor necrosis factor–α, interleukin-6, and IFN-γ receptor 1 are the major immunological determinants associated with post–kala azar dermal leishmaniasis. J. Infect. Dis. 2006, 194, 958–965. [Google Scholar] [CrossRef]
  40. Chen, P.M.; Liu, K.J.; Hsu, P.J.; Wei, C.F.; Bai, C.H.; Ho, L.J.; Sytwu, H.K.; Yen, B.L. Induction of immunomodulatory monocytes by human mesenchymal stem cell-derived hepatocyte growth factor through ERK1/2. J. Leukoc. Biol. 2014, 96, 295–303. [Google Scholar] [CrossRef]
  41. Nitsche, A.; Zhang, M.; Clauss, T.; Siegert, W.; Brune, K.; Pahl, A. Cytokine profiles of cord and adult blood leukocytes: Differences in expression are due to differences in expression and activation of transcription factors. BMC Immunol. 2007, 8, 18. [Google Scholar] [CrossRef] [PubMed]
  42. Polumuri, S.K.; Jayakar, G.G.; Shirey, K.A.; Roberts, Z.J.; Perkins, D.J.; Pitha, P.M.; Vogel, S.N. Transcriptional regulation of murine IL-33 by TLR and non-TLR agonists. J. Immunol. 2012, 189, 50–60. [Google Scholar] [CrossRef]
  43. Hansen, L.K.; Sevelsted-Møller, L.; Rabjerg, M.; Larsen, D.; Hansen, T.P.; Klinge, L.; Wulff, H.; Knudsen, T.; Kjeldsen, J.; Köhler, R. Expression of T-cell KV1. 3 potassium channel correlates with pro-inflammatory cytokines and disease activity in ulcerative colitis. J. Crohns Colitis 2014, 8, 1378–1391. [Google Scholar] [CrossRef] [PubMed]
  44. Marotte, H.; Salahuddin Ahmed, J.H.R.; Koch, A.E. Blocking ERK1/2 reduces TNF-α-induced-IL-18 bioactivity in rheumatoid arthritis synovial fibroblasts by induction of IL-18BPa Role of ERK1/2 in IL-18 bioactivity regulation. Arthritis Rheum. 2010, 62, 722. [Google Scholar] [CrossRef] [PubMed]
  45. Kong, J.; Grando, S.A.; Li, Y.C. Regulation of IL-1 family cytokines IL-1α, IL-1 receptor antagonist, and IL-18 by 1, 25-dihydroxyvitamin D3 in primary keratinocytes. J. Immunol. 2006, 176, 3780–3787. [Google Scholar] [CrossRef] [PubMed]
  46. Giribaldi, G.; Prato, M.; Ulliers, D.; Gallo, V.; Schwarzer, E.; Akide-Ndunge, O.B.; Valente, E.; Saviozzi, S.; Calogero, R.A.; Arese, P. Involvement of inflammatory chemokines in survival of human monocytes fed with malarial pigment. Infect. Immun. 2010, 78, 4912–4921. [Google Scholar] [CrossRef]
  47. Li, Z.; Wang, X.; Pan, H.; Yang, H.; Li, X.; Zhang, K.; Wang, H.; Zheng, Z.; Liu, H.; Wang, J. Resistin promotes CCL4 expression through toll-like receptor-4 and activation of the p38-MAPK and NF-κB signaling pathways: Implications for intervertebral disc degeneration. Osteoarthr. Cartil. 2017, 25, 341–350. [Google Scholar] [CrossRef]
  48. Pharoah, D.S.; Varsani, H.; Tatham, R.W.; Newton, K.R.; de Jager, W.; Prakken, B.J.; Klein, N.; Wedderburn, L.R. Expression of the inflammatory chemokines CCL5, CCL3 and CXCL10 in juvenile idiopathic arthritis, and demonstration of CCL5 production by an atypical subset of CD8+ T cells. Arthritis Res. Ther. 2006, 8, 1–11. [Google Scholar] [CrossRef]
  49. Johnson, B.V.; Bert, A.G.; Ryan, G.R.; Condina, A.; Cockerill, P.N. Granulocyte-macrophage colony-stimulating factor enhancer activation requires cooperation between NFAT and AP-1 elements and is associated with extensive nucleosome reorganization. Mol. Cell. Biol. 2004, 24, 7914–7930. [Google Scholar] [CrossRef]
  50. Schmittgen, T.D.; Livak, K.J. Analyzing real-time PCR data by the comparative CT method. Nat. Protoc. 2008, 3, 1101–1108. [Google Scholar] [CrossRef]
  51. Rayavara, K.; Kurosky, A.; Stafford, S.J.; Garg, N.J.; Brasier, A.R.; Garofalo, R.P.; Hosakote, Y.M. Proinflammatory effects of respiratory syncytial virus–induced epithelial HMGB1 on human innate immune cell activation. J. Immunol. 2018, 201, 2753–2766. [Google Scholar] [CrossRef]
  52. Taleb, S.A.; Al Thani, A.A.; Al Ansari, K.; Yassine, H.M. Human respiratory syncytial virus: Pathogenesis, immune responses, and current vaccine approaches. Eur. J. Clin. Microbiol. Infect. Dis. 2018, 37, 1817–1827. [Google Scholar] [CrossRef] [PubMed]
  53. Almajhdi, F.N.; Farrag, M.A.; Amer, H.M. Group B strains of human respiratory syncytial virus in Saudi Arabia: Molecular and phylogenetic analysis. Virus Genes 2014, 48, 252–259. [Google Scholar] [CrossRef] [PubMed]
  54. Tabatabai, J.; Prifert, C.; Pfeil, J.; Grulich-Henn, J.; Schnitzler, P. Novel respiratory syncytial virus (RSV) genotype ON1 predominates in Germany during winter season 2012–13. PLoS ONE 2014, 9, e109191. [Google Scholar] [CrossRef]
  55. Calderón, A.; Pozo, F.; Calvo, C.; García-García, M.; González-Esguevillas, M.; Molinero, M.; Casas, I. Genetic variability of respiratory syncytial virus A in hospitalized children in the last five consecutive winter seasons in Central Spain. J. Med. Virol. 2017, 89, 767–774. [Google Scholar] [CrossRef]
  56. Gaymard, A.; Bouscambert-Duchamp, M.; Pichon, M.; Frobert, E.; Vallee, J.; Lina, B.; Casalegno, J.-S.; Morfin, F. Genetic characterization of respiratory syncytial virus highlights a new BA genotype and emergence of the ON1 genotype in Lyon, France, between 2010 and 2014. J. Clin. Virol. 2018, 102, 12–18. [Google Scholar] [CrossRef] [PubMed]
  57. Madi, N.; Chehadeh, W.; Asadzadeh, M.; Al-Turab, M.; Al-Adwani, A. Analysis of genetic variability of respiratory syncytial virus groups A and B in Kuwait. Arch. Virol. 2018, 163, 2405–2413. [Google Scholar] [CrossRef]
  58. Abou-El-Hassan, H.; Massaad, E.; Soudani, N.; Assaf-Casals, A.; Shaker, R.; Lteif Khoury, M.; Ghanem, S.; Karam, M.; Andary, R.; Saito, R.; et al. Detection of ON1 and novel genotypes of human respiratory syncytial virus and emergence of palivizumab resistance in Lebanon. PLoS ONE 2019, 14, e0212687. [Google Scholar] [CrossRef]
  59. Russell, C.D.; Unger, S.A.; Walton, M.; Schwarze, J. The human immune response to respiratory syncytial virus infection. Clin. Microbiol. Rev. 2017, 30, 481–502. [Google Scholar] [CrossRef]
  60. Vázquez, Y.; González, L.; Noguera, L.; González, P.A.; Riedel, C.A.; Bertrand, P.; Bueno, S.M. Cytokines in the respiratory airway as biomarkers of severity and prognosis for respiratory syncytial virus infection: An update. Front. Immunol. 2019, 10, 1154. [Google Scholar] [CrossRef]
  61. Öner, D.; Drysdale, S.B.; McPherson, C.; Lin, G.-L.; Janet, S.; Broad, J.; Pollard, A.J.; Aerssens, J. Biomarkers for disease severity in children infected with respiratory syncytial virus: A systematic literature review. J. Infect. Dis. 2020, 222, S648–S657. [Google Scholar] [CrossRef] [PubMed]
  62. Sheeran, P.; Jafri, H.; Carubelli, C.; Saavedra, J.; Johnson, C.; Krisher, K.; Sanchez, P.J.; Ramilo, O. Elevated cytokine concentrations in the nasopharyngeal and tracheal secretions of children with respiratory syncytial virus disease. Pediatr. Infect. Dis. J. 1999, 18, 115–122. [Google Scholar] [CrossRef]
  63. Midulla, F.; Tromba, V.; Russo, L.L.; Mileto, F.; Sabatino, G.; Sgarrella, M.; Panuska, J.; Manganozzi, L.; Korn, D.; Moretti, C. Cytokines in the nasal washes of children with respiratory syncytial virus bronchiolitis. Int. J. Immunopathol. Pharmacol. 2006, 19, 205873920601900124. [Google Scholar] [CrossRef]
  64. Kristjansson, S.; Bjarnarson, S.P.; Wennergren, G.; Palsdottir, A.H.; Arnadottir, T.; Haraldsson, A.; Jonsdottir, I. Respiratory syncytial virus and other respiratory viruses during the first 3 months of life promote a local TH2-like response. J. Allergy Clin. Immunol. 2005, 116, 805–811. [Google Scholar] [CrossRef] [PubMed]
  65. Saravia, J.; You, D.; Shrestha, B.; Jaligama, S.; Siefker, D.; Lee, G.I.; Harding, J.N.; Jones, T.L.; Rovnaghi, C.; Bagga, B. Respiratory syncytial virus disease is mediated by age-variable IL-33. PLoS Pathog. 2015, 11, e1005217. [Google Scholar] [CrossRef] [PubMed]
  66. Kerrin, A.; Fitch, P.; Errington, C.; Kerr, D.; Waxman, L.; Riding, K.; McCormack, J.; Mehendele, F.; McSorley, H.; MacKenzie, K.; et al. Differential lower airway dendritic cell patterns may reveal distinct endotypes of RSV bronchiolitis. Thorax 2017, 72, 620–627. [Google Scholar] [CrossRef]
  67. Khalfaoui, S.; Eichhorn, V.; Karagiannidis, C.; Bayh, I.; Brockmann, M.; Pieper, M.; Windisch, W.; Schildgen, O.; Schildgen, V. Lung infection by human bocavirus induces the release of profibrotic mediator cytokines in vivo and in vitro. PLoS ONE 2016, 11, e0147010. [Google Scholar] [CrossRef]
  68. Franz, A.; Adams, O.; Willems, R.; Bonzel, L.; Neuhausen, N.; Schweizer-Krantz, S.; Ruggeberg, J.U.; Willers, R.; Henrich, B.; Schroten, H.; et al. Correlation of viral load of respiratory pathogens and co-infections with disease severity in children hospitalized for lower respiratory tract infection. J. Clin. Virol. 2010, 48, 239–245. [Google Scholar] [CrossRef]
  69. Mação, P.; Dias, A.; Azevedo, L.; Jorge, A.; Rodrigues, C. Acute bronchiolitis: A prospective study. Acta Med. Port. 2011, 24, 407–412. [Google Scholar]
  70. Jain, S.; Williams, D.J.; Arnold, S.R.; Ampofo, K.; Bramley, A.M.; Reed, C.; Stockmann, C.; Anderson, E.J.; Grijalva, C.G.; Self, W.H.; et al. Community-acquired pneumonia requiring hospitalization among US children. N. Engl. J. Med. 2015, 372, 835–845. [Google Scholar] [CrossRef]
  71. Sun, H.; Sun, J.; Ji, W.; Hao, C.; Yan, Y.; Chen, Z.; Wang, Y.I. Impact of RSV coinfection on human bocavirus in children with acute respiratory infections. J. Trop. Pediatr. 2019, 65, 342–351. [Google Scholar] [CrossRef] [PubMed]
  72. Tanaka, T.; Narazaki, M.; Kishimoto, T. IL-6 in inflammation, immunity, and disease. Cold Spring Harb. Perspect. Biol. 2014, 6, a016295. [Google Scholar] [CrossRef] [PubMed]
  73. Jones, S.A.; Jenkins, B.J. Recent insights into targeting the IL-6 cytokine family in inflammatory diseases and cancer. Nat. Rev. Immunol. 2018, 18, 773–789. [Google Scholar] [CrossRef] [PubMed]
  74. Tabarani, C.M.; Bonville, C.A.; Suryadevara, M.; Branigan, P.; Wang, D.; Huang, D.; Rosenberg, H.F.; Domachowske, J.B. Novel inflammatory markers, clinical risk factors, and virus type associated with severe respiratory syncytial virus infection. Pediatr. Infect. Dis. J. 2013, 32, e437. [Google Scholar] [CrossRef] [PubMed]
  75. Brown, P.M.; Schneeberger, D.L.; Piedimonte, G. Biomarkers of respiratory syncytial virus (RSV) infection: Specific neutrophil and cytokine levels provide increased accuracy in predicting disease severity. Paediatr. Respir. Rev. 2015, 16, 232–240. [Google Scholar] [CrossRef]
  76. Russo, R.C.; Garcia, C.C.; Teixeira, M.M.; Amaral, F.A. The CXCL8/IL-8 chemokine family and its receptors in inflammatory diseases. Expert Rev. Clin. Immunol. 2014, 10, 593–619. [Google Scholar] [CrossRef]
  77. Neto, E.A.; Durigon, T.S.; Oliveira-Junior, M.C.; dos Santos Dias, A.; MacKenzie, B.; Constancio, F.S.; El-Mafarjeh, E.; Rigonato-Oliveira, N.C.; de Brito, A.A.; Moraes, G.d.A.C.; et al. New serum and nasal immunological biomarkers in pediatric infection caused by respiratory syncytial virus. Eur. Respir. Soc. 2016, 48, PA1277. [Google Scholar]
  78. Diaz, P.V.; Gaggero, A.A.; Pinto, R.A.; Mamani, R.; Uasapud, P.A.; Bono, M.R. Levels of inflammatory cytokines and plasma cortisol in respiratory syncytial virus bronchiolitis. Rev. Med. Chile 2013, 141, 574–581. [Google Scholar]
  79. Brand, H.K.; Ferwerda, G.; Preijers, F.; de Groot, R.; Neeleman, C.; Staal, F.J.; Warris, A.; Hermans, P.W. CD4+ T-cell counts and interleukin-8 and CCL-5 plasma concentrations discriminate disease severity in children with RSV infection. Pediatr. Res. 2013, 73, 187–193. [Google Scholar] [CrossRef]
  80. Chan, B.C.; Lam, C.W.; Tam, L.-S.; Wong, C.K. IL33: Roles in allergic inflammation and therapeutic perspectives. Front. Immunol. 2019, 10, 364. [Google Scholar] [CrossRef]
  81. Donovan, C.; Bourke, J.E.; Vlahos, R. Targeting the IL-33/IL-13 axis for respiratory viral infections. Trends Pharmacol. Sci. 2016, 37, 252–261. [Google Scholar] [CrossRef] [PubMed]
  82. Jackson, D.J.; Makrinioti, H.; Rana, B.M.; Shamji, B.W.; Trujillo-Torralbo, M.-B.; Footitt, J.; Del-Rosario, J.; Telcian, A.G.; Nikonova, A.; Zhu, J. IL-33–dependent type 2 inflammation during rhinovirus-induced asthma exacerbations in vivo. Am. J. Respir. Crit. Care Med. 2014, 190, 1373–1382. [Google Scholar] [CrossRef] [PubMed]
  83. Mattes, J.; Yang, M.; Mahalingam, S.; Kuehr, J.; Webb, D.C.; Simson, L.; Hogan, S.P.; Koskinen, A.; McKenzie, A.N.; Dent, L.A. Intrinsic defect in T cell production of interleukin (IL)-13 in the absence of both IL-5 and eotaxin precludes the development of eosinophilia and airways hyperreactivity in experimental asthma. J. Exp. Med. 2002, 195, 1433–1444. [Google Scholar] [CrossRef] [PubMed]
  84. Kuchta, K.; Towpik, J.; Biernacka, A.; Kutner, J.; Kudlicki, A.; Ginalski, K.; Rowicka, M. Predicting proteome dynamics using gene expression data. Sci. Rep. 2018, 8, 13866. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Comparison of cytokines and chemokines markers in the NPAs among children with HRSV and/or HBoV and disease severity. Note: Data presented Median (25th–75th) percentile. * Denotes a significant difference between mild and HBoV of each virus respectively at 0.05 level.
Figure 1. Comparison of cytokines and chemokines markers in the NPAs among children with HRSV and/or HBoV and disease severity. Note: Data presented Median (25th–75th) percentile. * Denotes a significant difference between mild and HBoV of each virus respectively at 0.05 level.
Genes 14 01101 g001
Table 1. General patient characteristics.
Table 1. General patient characteristics.
ParametersChildren with HRSV
n (%)
Children with HBoV
n (%)
Children with HRSV Coinfection with HBoV
n (%)
Control
n (%)
Numbers36231625
Age in years
<116 (44.5)15 (65.2)8 (50.0)11 (44.0)
1–311 (30.5)5 (21.8)5 (31.2)9 (36.0)
3–59 (25.0)3 (13.0)3 (18.8)5 (20.0)
Gender
Male22 (61.1)14 (60.9)12 (75)14 (56.0)
Female14 (38.9)31 (39.1)4 (25)11 (44.0)
Type of virus
HRSV-A24 (66.7)
HRSV-B12 (33.3)
Severity
Mild20 (55.6)18 (78.3)10 (62.5)
Severe16 (44.4)5 (21.7)6 (37.5)
Note: Data presented as number (%).
Table 2. Comparison of cytokine and chemokine profile between 75 patients and 25 healthy controls.
Table 2. Comparison of cytokine and chemokine profile between 75 patients and 25 healthy controls.
Healthy Controls
(n = 25)
Patients
(n = 75)
p Value
INF-G2.36 (1.8–4.18)2.32 (0.81–4.17)0.922
TNF-a1.51 (0.38–0.80)1.03 (0.74–6.10)0.222
IL-1a2.77 (1.44–34.9)2.96 (0.92- 44.9)0.452
IL-219.5 (0.53–27.0)18.5 (0.53–41.0)0.626
IL-41.39 (0.33–0.54)1.56 (0.13–0.67)0.455
IL-61.50 (0.49–1.43)1.26 (0.49–4.32)0.015
IL-80.53 (0.27–1.50)1.42 (0.27–3.02)0.011
IL-100.11 (0.26–1.87)0.20 (0.13–2.87)0.007
IL-130.70 (0.50–1.56)3.28 (0.50–13.3)0.018
IL-170.80 (0.80–1.56)0.16 (0.05–1.56)0.336
IL-222.23 (1.38–3.56)2.55 (0.16–7.96)0.525
IL-335.68 (7.1–12.01)5.89 (3.73–37.01)0.021
G-CSF2.9 (2.64–60.03)3.15 (1.55–61.0)0.030
CCL-28.30 (0.35–13.7)10.77 (0.35–22.45)0.851
CCL-32.33 (0.35–0.12)2.38 (0.78–0.35)0.551
GM-CSF1.10 (0.68–1.93)1.18 (0.68–2.72)0.737
CCL-41.89 (0.41–2.22)2.10 (0.23–11.2)0.801
CCL-50.18 (0.23–0.53)0.11 (0.07–1.09)0.22
Note: Data presented Median (25th–75th) percentile. p-value significant at 0.05 and 0.01 level.
Table 3. Comparison of cytokine and chemokine profile between HRSV and HBoV infected patients.
Table 3. Comparison of cytokine and chemokine profile between HRSV and HBoV infected patients.
ParametersHRSV (n = 36)HBoV (n = 23)HRSV Coinfection with HBoV (n = 16)p-Value
INF-G2.32 (0.81–4.17)2.12 (1.16–3.18)3.70 (2.91–8.34)0.450
TNF-a1.94 (0.9–4.93)1.51 (0.38–0.80)1.87 (1.50–5.83) B0.015
IL-1a3.11 (1.45–10.16)3.91 (2.44–34.86)24.47 (4.88–44.90)0.161
IL-218.50 (3.39–41.0)21.0 (0.53–27.0)45.55 (24.06–64.25) B0.012
IL-41.56 (0.13–0.67)1.40 (0.33–0.54)1.90 (0.67–1.0) AB0.001
IL-61.26 (0.50–4.32)1.67 (0.49–1.43)2.31 (1.35–4.50) B0.008
IL-81.42 (0.70–3.02)1.80 (0.27–1.16)2.65 (1.45–4.75) B0.024
IL-101.20 (0.13–0.67)1.63 (0.26–2.87) A1.06 (0.51–1.51) A<0.001
IL-133.28 (0.82–13.34)1.90 (0.50–1.56)4.05 (2.93–14.25) B0.017
IL-171.16 (0.08–0.75)1.13 (0.05–0.13)1.16 (1.08–1.95) AB<0.001
IL-222.55 (0.16–7.96)2.15 (1.38–3.56)4.55 (2.26–11.46)0.196
IL-335.89 (3.73–12.34)25.58 (7.07–37.01) A30.75 (17.05–37.82) A<0.001
G-CSF3.15 (1.55–61.0)28.9 (2.64–63.03)28.53 (9.31–87.0)0.139
CCL-210.77 (1.96–22.45)6.69 (1.38–13.69)18.19 (8.32–37.38)0.198
CCL-32.38 (0.78–5.17)4.33 (0.35–24.02)14.96 (5.10–28.78) B0.002
GM-CSF1.18 (0.73–2.72)0.88 (0.68–1.93)3.81 (2.42–4.78) AB<0.001
CCL-41.37 (0.18–8.46)0.89 (0.41–2.22)8.28 (3.0–12.37) B0.119
CCL-51.11 (0.07–1.09)1.28 (0.23–0.53)2.71 (1.11–2.25) AB0.001
Note: Data presented Median (25th–75th) percentile. p-value significant at 0.05 and 0.01 level. Superscript A & B presented posthoc analysis between HRSV and HBoV respectively.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Alkubaisi, N.A.; Aziz, I.M.; Alsaleh, A.N.; Alhetheel, A.F.; Almajhdi, F.N. Molecular Profiling of Inflammatory Mediators in Human Respiratory Syncytial Virus and Human Bocavirus Infection. Genes 2023, 14, 1101. https://doi.org/10.3390/genes14051101

AMA Style

Alkubaisi NA, Aziz IM, Alsaleh AN, Alhetheel AF, Almajhdi FN. Molecular Profiling of Inflammatory Mediators in Human Respiratory Syncytial Virus and Human Bocavirus Infection. Genes. 2023; 14(5):1101. https://doi.org/10.3390/genes14051101

Chicago/Turabian Style

Alkubaisi, Noorah A., Ibrahim M. Aziz, Asma N. Alsaleh, Abdulkarim F. Alhetheel, and Fahad N. Almajhdi. 2023. "Molecular Profiling of Inflammatory Mediators in Human Respiratory Syncytial Virus and Human Bocavirus Infection" Genes 14, no. 5: 1101. https://doi.org/10.3390/genes14051101

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