This Key Event Relationship is licensed under the Creative Commons BY-SA license. This license allows reusers to distribute, remix, adapt, and build upon the material in any medium or format, so long as attribution is given to the creator. The license allows for commercial use. If you remix, adapt, or build upon the material, you must license the modified material under identical terms.
Relationship: 1703
Title
Increased proinflammatory mediators leads to Recruitment of inflammatory cells
Upstream event
Downstream event
Key Event Relationship Overview
AOPs Referencing Relationship
AOP Name | Adjacency | Weight of Evidence | Quantitative Understanding | Point of Contact | Author Status | OECD Status |
---|---|---|---|---|---|---|
Substance interaction with the pulmonary resident cell membrane components leading to pulmonary fibrosis | adjacent | Moderate | Low | Sabina Halappanavar (send email) | Under development: Not open for comment. Do not cite | WPHA/WNT Endorsed |
Decreased fibrinolysis and activated bradykinin system leading to hyperinflammation | adjacent | Penny Nymark (send email) | Under development: Not open for comment. Do not cite | Under Development | ||
Frustrated phagocytosis leads to malignant mesothelioma | adjacent | High | Not Specified | Penny Nymark (send email) | Under development: Not open for comment. Do not cite | |
Interaction with lung resident cell membrane components leads to lung cancer | adjacent | Moderate | Low | Penny Nymark (send email) | Under development: Not open for comment. Do not cite | |
Binding of SARS-CoV-2 to ACE2 leads to hyperinflammation (via cell death) | adjacent | High | High | Laure-Alix Clerbaux (send email) | Under development: Not open for comment. Do not cite |
Taxonomic Applicability
Sex Applicability
Life Stage Applicability
Key Event Relationship Description
Pro-inflammatory mediators are the chemical and biological molecules that initiate and regulate inflammatory reactions. They are secreted following inflammation or exposure to an inflammogen. Commonly measured pro-inflammatory mediators include Interleukin (IL)-1 family cytokines, IL-4, IL-5, IL-6, Tumor necrosis factor alpha (TNF-α), Interferon gamma (IFN-γ) (KE1496)
Proinflammatory mediator increase is caused when there’s increased inflammation. This can be found in many ways, including bradykinin system activation or hypofibrinolysis (Hofman et al., 2016). With more proinflammatory mediators, this causes increased signaling from proinflammatory cytokines, which promotes leukocyte recruitment, which will differentiate into proinflammatory cells (Villeneuve et al., 2018). Increased proinflammatory mediators means this process happens more, which means increase recruitment of inflammatory cells.
Evidence Collection Strategy
Evidence Supporting this KER
Biological Plausibility
The biological plausibility of this KER is high. There are very well established functional relationships between the secreted signalling molecules and the chemotactic effects on pro-inflammatory cells (Harris, 1954; Petri and Sanz 2018).
Increased proinflammatory mediators means more pro-inflammatory cytokines, chemokines, vasoactive amines, and lipid mediators (Villeneuve et al., 2018). Increased signaling from these Cytokines and Chemokines promote leukocyte recruitment to areas of infection, including monocytes and neutrophils (Khatri et al., 2017; Leick et al., 2014; Marchini et al., 2016). The leukocytes will differentiate into mature pro-inflammatory cells, in response to mediators they encounter in the local tissue microenvironment (Villeneuve et al., 2018). With higher levels of leukocytes from increased pro-inflammatory mediators, it causes an increase in pro-inflammatory cells (Libby, 2015).
Empirical Evidence
The empirical support for this KER is moderate. There are many studies which show temporal and dose-dependent recruitment of immune cells following increases in pro-inflammatory mediators. However, these mediators exhibit pleiotropy, and knockdown or knockout of a single pathway or mediator can result in compensation and recruitment of immune cells at a later time, as is seen in Nikota et al.,. 2017. (Chen et al., 2016; Nikota et al., 2017; Schremmer et al., 2016) (Additional studies available in Table 1).
Dose-Response Evidence:
Many studies provide dose-response evidence of this KER. For example, in vitro and in vivo studies testing stressors at different doses/concentrations have demonstrated a dose-response relationship; at the higher dose of the stressor, the pro-inflammatory mediators increased, leading to an increase of pro-inflammatory cell recruitment.
Ma et al. (2016) studied inflammatory responses in male BALB/c mice exposed to multi-walled carbon nanotubes (MWCNTs) administered intravenously at different doses (0.5-4 mg/kg) for 2 days. A dose-dependent relationship was found between the levels of the inflammatory mediators IL-6 and TNF-α and the MWCNT dose. At the highest dose, 4 mg/Kg, white blood cells, lymphocytes, and neutrophils levels increased.
Porter et al. (2020) have demonstrated that MWCNTs caused dose-dependent and time-dependent pulmonary inflammation in male C57BL/6J mice. Animals received a single dose of 2.5, 10, or 40 µg/mouse. At 40 µg/mouse, IL-1β and IL-18 increased at one day post-exposure. Moreover, polymorphonuclear leukocytes increased on day 1, and after 7 days the number of inflammatory cells was higher.
Zinc oxide (ZnO) nanoparticles (NPs) can induce metal fume fever and acute inflammation. Female C57BL/6J mice were intratracheally instilled once at 11, 33, and 100 µg/kg with coated ZnO NPs. Inflammatory responses were evaluated after 1, 3, and 28 days of exposure. An increase in Serum Amyloid A3 (Saa3) mRNA in lung tissue was observed at 33 and 100 µg/kg. Neutrophils accumulated in bronchoalveolar lavage fluid (BALF) after 28 days of exposure in a dose-dependent manner (Hadrup et al., 2019).
Polyhexamethylene guanidine phosphate (PHMG-P) is used as a disinfectant. PHMG-P at 0.3, 0.9, and 1.5 mg/kg was instilled into the lungs of mice. At 7- and 14-days post-exposure an increase in the levels of pro-inflammatory markers (IL-1β, IL-6, and C-X-C motif chemokine ligand [CXCL]1) and an increase in mRNA levels of Monocyte chemoattractant protein (MCP)1, Matrix metalloproteinase (MMP)2, and MMP12 was seen. Moreover, on day 7, neutrophils were recruited to the inflamed site. These changes were observed in a dose-response manner (Song et al., 2014).
Bourdon et al. 2012 evaluated the toxicity of carbon black nanoparticles (CBNPs) in mouse lung and liver. C57BL/6 mice were exposed to Printex 90 CBNPs with 0.018, 0.054, or 0.162 mg, and after 1, 3, and 28 days of the single instillation, BALF was analyzed. Polymorphonuclear cell counts in BALF increased in a dose-dependent manner with the strongest recruitment 1- and 3-days post-exposure and remained elevated at day 28. CBNPs also increased the expression of Saa3 mRNA levels in lung tissue on days 1, 3, and 28 in a dose-dependent manner. Although this response decreased over time, the expression of Saa3 mRNA increased at all time points, which indicates a persistent acute phase response.
A study evaluated the mechanisms of toxicity after exposure to particulate matter (PM2.5) in a tri-culture system: A549 cells (alveolar epithelial cells) and THP-1 differentiated macrophages in the apical chamber; meanwhile, EA.hy926 endothelial cells were cultured in the basolateral chamber. The system was exposed to PM2.5 at three different concentrations 20, 60, and 180 µg/ml for 24 h. An increase in the pro-inflammatory mediators IL-6, IL-8, and TNF-α was observed, as well an increase in mRNA expression of MMP9, Intercellular adhesion molecule 1 (ICAM-1), and caveolin 1 (CAV-1). These genes are involved in the movement and recruitment of leukocytes in sites of inflammation. Changes were observed in a concentration-dependent manner (Wang et al., 2019).
In another study, female C57BL/6 mice were exposed to 18, 54, or 162 µg of MWCNT/mouse via single intratracheal instillation. An increased gene expression of Cxcl1, IL-6, Metallothionein-2 (Mt2), Saa1, and Saa2 was observed in a dose-dependent manner at 24 h post-exposure. Moreover, an increase in the recruitment of pro-inflammatory cells was observed in a dose-dependent manner (Poulsen et al., 2013).
Temporal Evidence:
There is significant evidence of the temporal relationship between the two KES. In vitro and in vivo studies have demonstrated that pro-inflammatory mediators (Event 1496) increased prior to the recruitment of pro-inflammatory cells (Event 1497).
Female C57BL/6J mice were exposed to carbon NPs at 20 µg/mouse via intratracheal instillation. An increase in the levels of cytokines CXCL1, CXCL2, and CXCL5 at 3 h post-exposure was observed, with peaks after 12 and 18 h post-exposure. These pro-inflammatory mediators preceded neutrophil recruitment (12 and 24 h post-exposure) (Chen et al., 2016). Alveolar macrophages (AM) were isolated from lungs 3 to 12 h after instillation, but they did not show a pro-inflammatory response. The authors suggest that AM are not involved in the initiation of the inflammatory response. Meanwhile, alveolar epithelial type II cells induced the highest CXCL levels and acute neutrophilic inflammation.
Nickel oxide (NiO) NPs intratracheally instilled at one single dose 200 cm2/rat into female Wistar rats induced an increase of pro-inflammatory cytokines in BALF, at 24 and 74 h for Cytokine-Induced neutrophil chemoattractant 3 (CINC-3) and eotaxin, respectively. At 24 h and 48 h, neutrophils were observed, and after 72 h, the levels of neutrophils, eosinophils, and macrophages increased (Lee et al., 2016).
Porter et al. (2002) have shown pulmonary inflammation in rats exposed to crystalline silica aerosol at a concentration of 15 mg/m3 (6h/day, 5 days/week) for 116 days. Lung disease was linked to TNF-α and IL-10 production in a timely response (10-116 days). The number of polymorphonuclear cells in the BALF increased progressively from day 41 - 116.
One study has demonstrated a dose-response and temporal relationship for these two KEs (Patowary et al., 2020). Female Wistar rats were exposed to oleoresin capsicum sprays at 2, 6, and 10%, and after 1, 3, and 24 h post-exposure, blood cell and BALF cytokines were evaluated. The pro-inflammatory cytokine TNF-α increased in a dose-dependent manner, and polymorphonuclear cells increased in a time-dependent manner.
Schremmer et al. (2016) have reported the time course of chemotaxis in vitro in response to the challenge of biopersistent particles and their relation to inflammatory mediators. NR8383 rat alveolar macrophages were challenged with different types of particles for 1, 4, and 16 h. The cell supernatants obtained from different time points were used to evaluate the chemotaxis of unexposed NR8383 macrophages. They found that nanosized silica at 16 µg/cm2 induced an elevated transcription of C-C motif chemokine ligand (CCL)4, CXCL1, CXCL3, and TNF-α in a time-dependent manner. The pro-inflammatory cytokines present in the supernatants induced chemotaxis of unexposed macrophages at 4 and 16 h post-exposure.
Husain et al. (2015) found increased expression of genes related to chemotactic recruitment of pro-inflammatory cells at 3 h and 1 day after exposure to 162 µg/mouse CBNPs in female C57BL/6 mice. They observed an increase in the gene expression of pro-inflammatory mediators at day 1 (CXCL2, Ccl2), day 3 (IL-17, IL-33), day 14 (Cd2), and day 42 (Cxcl) post-exposure. The KE2 (Event 1497) increased over time with the maximum levels of neutrophils, macrophages, eosinophils, and lymphocytes at 4- and 5-days post-exposure. This response suggests chronic inflammation occurs because of an incomplete resolution of acute inflammation.
Rahman et al. (2017) evaluated whether different titanium dioxide (TiO2) NPs induce lung inflammation. C57BL/6 mice were exposed to 18, 54, 162, or 486 µg/mouse of TiO2 NPs via single intratracheal instillation. At 1-day post-exposure, gene expression analysis showed more changes in genes associated with inflammation and fibrosis. Moreover, after 1- and 28 days post-exposure, an increase in cell counts in BALF was observed in a dose-dependent manner.
Ho et al. (2013) evaluated the inflammatory response in mice exposed to coated quantum dots, cadmium-based NPs, (QD705-poly(ethylene glycol[PEG], QD705-COOH) at 12 or 60 µg/mouse. At 2-, 17- and 90-days post-exposure, an increase in the level of TNF-α, IL-1b, IL-6, CXCL1, CCL2, CCL1, CCL17, and CXCL13 mRNA levels in lungs was observed and the amount of polymorphonuclear cells in BALF increased in a dose-dependent manner at day 7 post-exposure. The inflammatory response increased on days 2 and 17, but on day 90 decreased. QD705-COOH induced granulomas persistently presented from 2 to 90 days.
Morimoto et al. (2010) examined the different kinds of cytokines related to lung inflammation by NiO exposure. Rats were intratracheally exposed to 0.33 mg/Kg and 0.66 mg/kg NiO NPs and were sacrificed at day 3, after 1 week, 1, 3, and 6 months post-exposure. Infiltration of alveolar macrophages in lung tissue and BALF was observed from day 3 to 3 months post-exposure, with higher levels after 1 and 3 months. Before the recruitment of inflammatory cells, an increase in the level of pro-inflammatory cells such as MCP-1 and IL-1β in BALF was observed. NiO NPs induced a persistent inflammatory effect.
Kamata et al. (2011) studied the impact of CBNPs on susceptible subjects with predisposing lung disease and the effects of nanoparticles on inflammation and fibrotic changes. To achieve this goal, female C57BL/6J mice were intratracheally administered with bleomycin 20 µg/mouse and CBNPs 10 µg/mouse. Evaluations were performed post-exposure at different time points. An increase of IL-6 and CCL2 in BALF was observed at days 2 and 7. After 7- and 14 days, a recruitment of pro-inflammatory cells was observed. Oxidant injury (evaluated as nitrotyrosine expression) was observed after 7 days and 14 days. The levels of Transforming growth factor beta (TGF-β) increased over time with the highest level at day 14. Finally, they observed an increase in lung collagen deposition and a decrease in lung compliance at day 21.
Uncertainties and Inconsistencies
Attenuation or complete abrogation of KE1 (KE1496) and KE2 (KE1497) following inflammogenic stimuli is observed in rodents lacking functional Interleukin 1 receptor type 1 (IL-1R1) or other cell surface receptors that engage innate immune response upon stimulation (Gasse et al., 2007; Halappanavar et al., 2013). However, following exposure to MWCNTs, it has been shown that absence of IL-1R1 signalling is compensated for eventually and neutrophil influx is observed at a later post-exposure time point (Nikota et al., 2017). In another study, acute neutrophilic inflammation induced by MWCNTs was suppressed at 24 h in mice deficient in IL-1R1 signalling; however, these mice showed exacerbated neutrophilic influx and fibrotic response at 28 days post-exposure (Girtsman et al., 2014). The early defence mechanisms involving damage-associated molecular patterns is fundamental for survival, which may necessitate activation of compensatory signalling pathways. As a result, inhibition of a single biological pathway mediated by an individual cell surface receptor may not be sufficient to completely abrogate the lung inflammatory response. Forced suppression of pro-inflammatory and immune responses early after exposure to substances that cannot be effectively cleared from lungs, may enhance the injury and initiate other pathways leading to exacerbated response.
Most of the studies evaluate one dose at different time points or one-time point at different concentrations. Moreover, some studies have demonstrated that a stressor can lead to the recruitment of pro-inflammatory cells, but the presence of pro-inflammatory mediators was not determined (Westphal et al., 2015).
Recruitment of pro-inflammatory cells is a key event that is complicated to replicate in vitro conditions as cell migration is induced by cooperative chemotactic mediators (Gouwy et al., 2015) which are produced and released from different cells. Therefore, more kinetics studies in co-culture techniques are needed to fill this gap.
Known modulating factors
Modulating Factor (MF) | MF Specification | Effect(s) on the KER | Reference(s) | |
---|---|---|---|---|
Air pollution |
Air pollution primes immunity; increases the levels of circulating IL-1β, IL-6 and TNF-α; impairs the normal functions of macrophages and alveolar cells. Exposure to particulate air pollution, such as PM2.5, is associated with pulmonary inflammation [1,2]. Both short term and chronic exposures to fine particulate matter (PM) have been shown to increase levels of circulating IL-1β, IL-6 and TNF-α [3-5]. Air pollution works as a priming factor that exacerbates the inflammatory phenotype of COVID-19 and further dysregulates immune cell activity. Dysregulation of the immune cell functions, on the other hand, plays a role in tissue damage and the ability of the immune system to fight pathogens, which increases the susceptibility to concomitant bacterial superinfection, for instance [6-9]. |
[1] Zhao et al., 2013 [2] Jia et al., 2021 [3] Tsai et al., 2012 [4] Ljungman et al., 2009 [5] Kido et al., 2011 [6] Knoll et al., 2021 [7] Glencross et al., 2020 [8] Yamasaki and Eeden, 2018 9) Signorini et al., 2018 |
||
Chemicals (weak evidence) |
Per- and polyfluoroalkyl substances (PFAS) (Perfluorooctane sulfonate [PFOS], perfluorooctanoic acid [PFOA], perfluorobutane sulfonic acid [PFBS], perfluorooctane sulfonamide [PFOSA], and perfluorodecanoic acid [PFDA]) |
Several in vitro studies in human-derived cells have shown that PFAS can modify the secretion of pro-inflammatory mediators in a dose-dependent manner [1]. PFOS exposure significantly induced IL-1 IL-4, IL-6, and IL-8 in human lymphocytes and reduced chemokines CXCL8 and CXCL10 secretion in human bronchial epithelial cells while increasing of IL-1α release [2]; both PFOS and PFOA enhanced IL-1β release in response to Poly I:C [3]; PFOS, PFOA, PFBS, PFOSA, and PFDA exposure decreased PHA-induced release of IL-4, IL-10, and IL-6 and PFOS, PFOSA, and PFDA decreased IFN-γ release in human leukocytes with PFOS as a more potent inhibitor of cytokine production than other PFAS, and leukocytes obtained from female donors appeared to be more sensitive to the in vitro immunomodulating effects of PFAS, compared to leukocytes from male donors [4]. In a rat study exposed to PFOS, increased serum levels of TNF-α and IL-6 were observed. Kupffer cells exposed to PFOS showed cell activation, which was mostly inhibited by anti-TNF-α or anti-IL-6 treatment. Moreover, NF-κB inhibitor and JNK inhibitor significantly inhibited the production of IL-6 [5,6]. |
[1] Tian et al., 2021 [2] Li et al., 2020 [3] Sørli et al., 2020 [4] Corsini et al., 2012 [5] Han et al., 2018 [6] EFSA CONTAM Panel, 2020 |
|
Sex | Female sex (XX chromosomes) |
Females produce higher amounts of the antiviral infection cytokine IFN-α than men [1]. Estrogens are critical regulators of gene expression and functions in innate immune cells, including monocytes, macrophages, and dendritic cells, as well as lymphocytes such as T helper 1/2 (TH1/2) cells, regulatory T-cells (Treg cells), and B cells. One of the major forms of estrogen, estradiol, has been shown to dampen the production of excessive innate inflammatory cytokines by monocytes and macrophages [2]. In the presence of progesterone, CD4+ Th cells skew from Th-1 to Th-2 in the production of anti-inflammatory cytokines, specifically IL-4 and IL-10 [3]. The cellular types involved in male and female immune responses to SARS-CoV-2 are distinct and immune response in females is enriched with activated T-cells [1]. In lactating women, higher SARS-CoV-2 reactive memory B-cells and antibody titers have been associated with the hormone prolactin [4]. Poor T-cell response to SARS-CoV-2 correlates with worse disease progression in female patients. |
[1] Takahashi et al., 2020 [2] Scully et al., 2020 [3] Mauvais-Jarvis et al., 2020 [4] Gonçalves et al., 2021 |
|
Male sex (XY chromosomes) |
Males display a higher innate immune response to SARS-CoV-2 than females, which conditions their cytokine profile. Men have higher levels of the innate immune cytokines IL-8 and IL-18 in circulation [1]. Moreover, elderly men in particular display autoantibodies against IFN-α more frequently [5]. The cellular types involved in male and female immune responses to SARS-CoV-2 are distinct. Men display higher circulating levels of non-classical monocytes [1]. Higher innate immune activation in men leads to higher plasma levels of the inflammatory cytokines IFN-α [6], IL-8 and IL-18 [1], driving hyperinflammation and more pronounced lymphopenia in males. |
[5] Bastard et al., 2020 [6] Agrawal et al., 2021 |
||
Age |
Old people | During aging, a subclinical chronic inflammatory response develops leading to an immune senescent state, where pathogen protective immune responses are impaired, but the production of inflammatory cytokines, such as IL-6, is increased. This process is called inflammaging. The persistent IL-6 elevation can induce lung tissue inflammation and mortality. The rate of inflammaging is higher in men and accelerated inflammaging is believed to worsen COVID-19 outcomes [1]. The chronic inflammatory status is associated with a dramatic depletion of B lymphocyte-driven acquired immunity. Aging also attenuates the upregulation of co-stimulatory molecules critical for T-cell priming and reduces antiviral IFN production by alveolar macrophages and dendritic cells in response to infection with the influenza virus [2]. |
[1] Bonafè et al., 2020 [2] Kovacs et al., 2017 |
|
Lipids |
Atherogenic dyslipidemia |
Lipids impact innate and adaptive immune responses [1,2]. In COVID-19. The atherogenic dyslipidemia associated with COVID-19 severity (high tryglycerides and low total, low density lipoprotein and high density lipoprotein cholesterol) was inversely correlated with inflammatory biomarkers such as increased levels of serum C-reactive protein (CRP), IL-6, IL-8, and IL-10 [3,4]. |
[1] Hubler and Kennedy, 2016 [2] Bernardi et al., 2018 [3] Henry et al., 2021 [4] Caterino et al., 2021 [5] Hubler and Kennedy, 2016 [6] Winer et al., 2009 [7] Im et al., 2011 [8] Muscogiuri et al., 2020 |
|
Obesity |
In obesity, immune cells interact with various classes of lipids, which can control the plasticity of macrophages and T lymphocytes. In COVID-19. Altered lipid homeostasis is associated with severe COVID-19 outcomes and, at the same time, with chronic inflammation and inflammatory polarization of macrophages and T lymphocytes [5]. Th1 lymphocytes are more prevalent in adipose tissue of obese patients [6]. In the same way, Th1 lymphocytes are elevated in visceral fat [6]. Both macrophages and T lymphocytes interact with lipids that influence their proliferation, differentiation, polarization [7] and transcriptional regulation, which is tightly controlled by Sterol regulatory element-binding protein (SREBP) and Liver X receptors (LXRs), expressed in macrophages and known regulators of cytokine release. Adipose tissue produces many pro-inflammatory adipokines and cytokines, which lead to low-grade inflammation and the recruitment of immune cells which may clarify the connection between obesity and COVID-19 severity [8]. |
|||
Gut microbiota | Gut dysbiosis (alteration of gut microbiota) |
The gut microbiota is increasingly acknowledged to play a central role in human health and disease, notably by shaping the immune response. Notably some bacteria living in the gut produce short-chain fatty acids (SCFA), recognized as mediators of the intestinal inflammatory response [1]. SCFAs modulate inflammation by regulating immune cell cytokine production such as TNF-α, IL-12, IL-6 [2]. For example, butyrate decreased the lipopolysaccharide (LPS)-induced TNF-α expression in monocytes [4] and activated Treg cells, blocking an excessive inflammatory response [1,3]. In COVID-19. In a COVID-19 cohort, the depletion of several bacterial species (B. adolescentis, E. rectale and F. prausnitzii, known to play immunomodulatory roles in the human gastrointestinal system) was linked to increased plasma concentrations of TNF-α, CXCL10, CCL2 and IL-10 [4]. Conversely, two species enriched in the COVID-19 cohort, B. dorei and Akkermansia muciniphila, were positively correlated with IL-1β, IL-6 and CXCL8. Using a machine learning model [5], it was reported that the disruption of gut microbiota significantly correlated with pro-inflammatory cytokines and may predispose normal individuals to severe COVID-19. Decreases in the abundance of butyrate-producing bacteria and a decline in SCFA were observed in severe COVID-19 [4,6,7,8]. Reduced relative proportion of bacteria producing SCFA was observed in Syrian hamsters infected with SARS-CoV-2, compared to non-infected controls, with a transient decrease in systemic SCFA amounts [9]. However, SCFA supplementation in hamsters during infection had no effect on inflammatory parameters. Targeted analysis of fecal metabolites showed significantly lower fecal concentrations of SCFAs in COVID-19 patients, which correlated with disease severity and increased plasma concentrations of CXCL-10 and CRP [10]. |
[1] Yoo et al., 2020 [2] Vinolo et al., 2011 [3] Atarashi et al., 2013 [4] Yeo et al., 2021 [5] Gou et al., 2021 [6] Zuo et al., 2020 [7] Gu et al., 2020 [8] Grenga et al., 2022 [9] Sencio et al., 2022 [10] Zhang et al., 2022 |
|
Vitamin D (low evidence) | Vitamin D deficiency |
There is a complex interplay between vitamin D and the immune response to viral infections. Low vitamin D status is proposed to induce upregulation of the TNF-α and downstream of Nuclear Factor Kappa B Subunit 1 (NF–κB1) signaling pathway, which regulates inflammatory reactions toward viral infection in macrophages [1,2]. Vitamin D was shown as a potent suppressor of IFN-γ mediated macrophages response, preventing the release of inflammatory cytokines and chemokines [3]. Thus, release of pro-inflammatory cytokines might be exacerbated in COVID-19 patients with vitamin D deficiency [4]. |
[1] Hassan et al., 2022 [2] Książek et al., 2021 [3] Helming et al., 2005 [4] Munshi et al., 2021 |
|
Genetic factors |
The inflammatory response manifested by increased cytokine levels results in inhibition of heme oxygenase (HO-1), with a subsequent loss of cytoprotection. In the 50-non-coding regions of the HO-1 gene, there are two polymorphic sites, namely the (GT)n dinucleotide and T (-413) A sites, which regulate the transcriptional activity of HO-1. These polymorphisms have been shown to be associated with the occurrence and progression of numerous diseases, including COVID-19 [1]. The timing of the IFN response to SARS-CoV-2 infection can vary with viral load and genetic differences in host response. When the viral load is low, IFN responses are engaged and contribute to viral clearance, resulting in mild infection. When viral load is high and/or genetic factors slow antiviral responses, virus replication can delay the IFN response and cytokine storm can occur before adaptive responses clear the virus, resulting in severe disease including multisystem inflammatory syndrome in Children (MIS-C) [2]. |
[1] Singh et al., 2020 [2] Rowley, 2020 |
||
Therapeutic intervention against COVID-19 |
Tocilizumab and Sarilumab |
Are anti-IL-6 receptor monoclonal antibodies, which reduce inflammation [1] by attaching to the IL-6 receptor (as IL-6 receptor inhibitors) [2]. Tocilizumab, a biological drug approved for rheumatoid arthritis, is currently being evaluated for its efficacy against the effects of systemic IL-6 elevation (ClinicalTrial.gov accessed on March 2022, NCT04317092, NCT04320615, NCT04306705) [3]. |
[1] WHO, 2021. [2] European Medicines Agency, 2021 [3] Bonafè et al., 2020 |
|
Baricitinib |
Is an immunosuppressant that blocks the action of enzymes known as Janus kinases (JK), which play an important role in inflammatory processes (JAK inhibitor) [1–4]. |
[1] Jorgensen et al., 2020 [2] Bekerman et al., 2017 [3] Neveu et al., 2015 [4] Richardson et al., 2020 |
||
Low molecular weight heparins (LMWHs) |
Have anti-inflammatory effects by blocking pro-inflammatory mediators (TNF-α, IL-6 and Leukotriene [LTB4]) [1]. |
[1] Buijsers et al., 2020 |
||
Pre-existing heart failure |
Dysregulation of renin angiotensin system due to pre-existing heart failure can have detrimental inflammatory effects both locally (in the heart) and systematically. The Angiotensin converting enzyme 2 (ACE2)/Angiotensin (Ang) (1-7) pathway is associated with the attenuation of a wide range of pro-inflammatory cytokines and chemokines, such as IL-1, IL-5, IL-6, IL-12, CCL2, TNF-α and MCP-1 [1]. |
[1] Rodrigues Prestes et al., 2017. |
||
Diet |
Dietary elements linked to pro-inflammatorymediators |
High-fat diets have been linked—in multiple studies—to promote an “inflammatory status” in the gut and subsequently other organs [1]. Compounds found in many plant foods may affect COVID-19 prognosis by blocking inflammatory mediators and pathways. Bousquet et al. [2,3] identified bioactive compounds contained in spices and fermented vegetables, including capsaicin, cinnamaldehyde, curcumin, genistein, gingerol, mustard oil, piperine, wasabi, and sulforaphane, that upregulate the signaling of nuclear factor (erythroid-derived 2)-like 2 (Nrf2), a potent endogenous antioxidant which blocks oxidative stress from the Angiotensin type I receptor (AT1R) axis, inhibits overproduction of proinflammatory cytokines and chemokines (including IL-6), and limits the activation of NF-κB. There is some in vitro evidence that Lactobacillus, found in many fermented foods, works through the same mechanism [4]. Finally, naringin, a compound found in citrus fruits, reduced LPS-induced IL-6 expression levels in vitro [5]. |
[1] Duan et al., 2018 [2] Bousquet et al., 2021a [3] Bousquet et al., 2020 [4] Bousquet et al., 2021b [5] Liu et al., 2022 |
Quantitative Understanding of the Linkage
A majority of the in vivo studies are conducted with only one dose and thus, it is difficult to derive quantitative dose-response relationships based on the existing data. However, it is clear from the studies referenced above that greater concentrations or doses of pro-fibrotic substances results in higher release of alarmins, and consequently, higher pro-inflammatory signalling. The above studies also demonstrate strong temporal relationships between the individual KEs.
Response-response Relationship
Time-scale
Known Feedforward/Feedback loops influencing this KER
Activated pro-inflammatory cells secrete pro-inflammatory mediators, and those mediators' goal is to cause signalling and response, which can lead to chronic inflammation (KE1497). Chronic inflammation means proinflammatory mediators increase and increased recruitment of inflammatory cells acts in a positive feedback loop, which continues a pro-inflammatory environment.
Domain of Applicability
References
-
Agrawal S, Salazar J, Tran TM, Agrawal A. Sex-Related Differences in Innate and Adaptive Immune Responses to SARS-CoV-2. Front Immunol. 2021 Oct 20;12:739757. doi: 10.3389/fimmu.2021.739757.
-
Atarashi K, Tanoue T, Oshima K, Suda W, Nagano Y, Nishikawa H, Fukuda S, Saito T, Narushima S, Hase K, Kim S, Fritz JV, Wilmes P, Ueha S, Matsushima K, Ohno H, Olle B, Sakaguchi S, Taniguchi T, Morita H, Hattori M, Honda K. Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota. Nature. 2013 Aug 8;500(7461):232-6. doi: 10.1038/nature12331.
-
Bastard P, Rosen LB, Zhang Q, Michailidis E, Hoffmann HH, Zhang Y, Dorgham K, Philippot Q, Rosain J, Béziat V, Manry J, Shaw E, Haljasmägi L, Peterson P, Lorenzo L, Bizien L, Trouillet-Assant S, Dobbs K, de Jesus AA, Belot A, Kallaste A, Catherinot E, Tandjaoui-Lambiotte Y, Le Pen J, Kerner G, Bigio B, Seeleuthner Y, Yang R, Bolze A, Spaan AN, Delmonte OM, Abers MS, Aiuti A, Casari G, Lampasona V, Piemonti L, Ciceri F, Bilguvar K, Lifton RP, Vasse M, Smadja DM, Migaud M, Hadjadj J, Terrier B, Duffy D, Quintana-Murci L, van de Beek D, Roussel L, Vinh DC, Tangye SG, Haerynck F, Dalmau D, Martinez-Picado J, Brodin P, Nussenzweig MC, Boisson-Dupuis S, Rodríguez-Gallego C, Vogt G, Mogensen TH, Oler AJ, Gu J, Burbelo PD, Cohen JI, Biondi A, Bettini LR, D'Angio M, Bonfanti P, Rossignol P, Mayaux J, Rieux-Laucat F, Husebye ES, Fusco F, Ursini MV, Imberti L, Sottini A, Paghera S, Quiros-Roldan E, Rossi C, Castagnoli R, Montagna D, Licari A, Marseglia GL, Duval X, Ghosn J; HGID Lab; NIAID-USUHS Immune Response to COVID Group; COVID Clinicians; COVID-STORM Clinicians; Imagine COVID Group; French COVID Cohort Study Group; Milieu Intérieur Consortium; CoV-Contact Cohort; Amsterdam UMC Covid-19 Biobank; COVID Human Genetic Effort; Tsang JS, Goldbach-Mansky R, Kisand K, Lionakis MS, Puel A, Zhang SY, Holland SM, Gorochov G, Jouanguy E, Rice CM, Cobat A, Notarangelo LD, Abel L, Su HC, Casanova JL. Autoantibodies against type I IFNs in patients with life-threatening COVID-19. Science. 2020 Oct 23;370(6515):eabd4585. doi: 10.1126/science.abd4585.
-
Bekerman E, Neveu G, Shulla A, Brannan J, Pu SY, Wang S, Xiao F, Barouch-Bentov R, Bakken RR, Mateo R, Govero J, Nagamine CM, Diamond MS, De Jonghe S, Herdewijn P, Dye JM, Randall G, Einav S. Anticancer kinase inhibitors impair intracellular viral trafficking and exert broad-spectrum antiviral effects. J Clin Invest. 2017 Apr 3;127(4):1338-1352. doi: 10.1172/JCI89857.
-
Bernardi S, Marcuzzi A, Piscianz E, Tommasini A, Fabris B. The Complex Interplay between Lipids, Immune System and Interleukins in Cardio-Metabolic Diseases. Int J Mol Sci. 2018 Dec 14;19(12):4058. doi: 10.3390/ijms19124058.
-
Bonafè M, Prattichizzo F, Giuliani A, Storci G, Sabbatinelli J, Olivieri F. Inflamm-aging: Why older men are the most susceptible to SARS-CoV-2 complicated outcomes. Cytokine Growth Factor Rev. 2020 Jun;53:33-37. doi: 10.1016/j.cytogfr.2020.04.005.
-
Bourdon JA, Saber AT, Jacobsen NR, Jensen KA, Madsen AM, Lamson JS, Wallin H, Møller P, Loft S, Yauk CL, Vogel UB. Carbon black nanoparticle instillation induces sustained inflammation and genotoxicity in mouse lung and liver. Part Fibre Toxicol. 2012 Feb 2;9:5. doi: 10.1186/1743-8977-9-5.
-
Bousquet J, Cristol JP, Czarlewski W, Anto JM, Martineau A, Haahtela T, Fonseca SC, Iaccarino G, Blain H, Fiocchi A, Canonica GW, Fonseca JA, Vidal A, Choi HJ, Kim HJ, Le Moing V, Reynes J, Sheikh A, Akdis CA, Zuberbier T; ARIA group. Nrf2-interacting nutrients and COVID-19: time for research to develop adaptation strategies. Clin Transl Allergy. 2020 Dec 3;10(1):58. doi: 10.1186/s13601-020-00362-7
-
Bousquet J, Czarlewski W, Zuberbier T, Mullol J, Blain H, Cristol JP, De La Torre R, Le Moing V, Pizarro Lozano N, Bedbrook A, Agache I, Akdis CA, Canonica GW, Cruz AA, Fiocchi A, Fonseca JA, Fonseca S, Gemicioğlu B, Haahtela T, Iaccarino G, Ivancevich JC, Jutel M, Klimek L, Kuna P, Larenas-Linnemann DE, Melén E, Okamoto Y, Papadopoulos NG, Pfaar O, Reynes J, Rolland Y, Rouadi PW, Samolinski B, Sheikh A, Toppila-Salmi S, Valiulis A, Choi HJ, Kim HJ, Anto JM. Spices to Control COVID-19 Symptoms: Yes, but Not Only…. Int Arch Allergy Immunol. 2021a;182(6):489-495. doi: 10.1159/000513538.
-
Bousquet J, Anto JM, Czarlewski W, Haahtela T, Fonseca SC, Iaccarino G, Blain H, Vidal A, Sheikh A, Akdis CA, Zuberbier T; ARIA group. Cabbage and fermented vegetables: From death rate heterogeneity in countries to candidates for mitigation strategies of severe COVID-19. Allergy. 2021b Mar;76(3):735-750. doi: 10.1111/all.14549.
-
Buijsers B, Yanginlar C, Maciej-Hulme ML, de Mast Q, van der Vlag J. Beneficial non-anticoagulant mechanisms underlying heparin treatment of COVID-19 patients. EBioMedicine. 2020 Sep;59:102969. doi: 10.1016/j.ebiom.2020.102969.
-
Caterino M, Gelzo M, Sol S, Fedele R, Annunziata A, Calabrese C, Fiorentino G, D'Abbraccio M, Dell'Isola C, Fusco FM, Parrella R, Fabbrocini G, Gentile I, Andolfo I, Capasso M, Costanzo M, Daniele A, Marchese E, Polito R, Russo R, Missero C, Ruoppolo M, Castaldo G. Dysregulation of lipid metabolism and pathological inflammation in patients with COVID-19. Sci Rep. 2021 Feb 3;11(1):2941. doi: 10.1038/s41598-021-82426-7.
-
Chen S, Yin R, Mutze K, Yu Y, Takenaka S, Königshoff M, Stoeger T. No involvement of alveolar macrophages in the initiation of carbon nanoparticle induced acute lung inflammation in mice. Part Fibre Toxicol. 2016 Jun 21;13(1):33. doi: 10.1186/s12989-016-0144-6.
-
Corsini E, Sangiovanni E, Avogadro A, Galbiati V, Viviani B, Marinovich M, Galli CL, Dell'Agli M, Germolec DR. In vitro characterization of the immunotoxic potential of several perfluorinated compounds (PFCs). Toxicol Appl Pharmacol. 2012 Jan 15;258(2):248-55. doi: 10.1016/j.taap.2011.11.004.
-
Duan Y, Zeng L, Zheng C, Song B, Li F, Kong X, Xu K. Inflammatory Links Between High Fat Diets and Diseases. Front Immunol. 2018 Nov 13;9:2649. doi: 10.3389/fimmu.2018.02649
-
EFSA Panel on Contaminants in the Food Chain (EFSA CONTAM Panel); Schrenk D, Bignami M, Bodin L, Chipman JK, Del Mazo J, Grasl-Kraupp B, Hogstrand C, Hoogenboom LR, Leblanc JC, Nebbia CS, Nielsen E, Ntzani E, Petersen A, Sand S, Vleminckx C, Wallace H, Barregård L, Ceccatelli S, Cravedi JP, Halldorsson TI, Haug LS, Johansson N, Knutsen HK, Rose M, Roudot AC, Van Loveren H, Vollmer G, Mackay K, Riolo F, Schwerdtle T. Risk to human health related to the presence of perfluoroalkyl substances in food. EFSA J. 2020 Sep 17;18(9):e06223. doi: 10.2903/j.efsa.2020.6223.
-
European Medicines Agency. RoActemra. 2021. Available online: https://www.ema.europa.eu/en/medicines/human/EPAR/roactemra (accessed 12 May 2022).
-
Gasse P, Mary C, Guenon I, Noulin N, Charron S, Schnyder-Candrian S, Schnyder B, Akira S, Quesniaux VF, Lagente V, Ryffel B, Couillin I. IL-1R1/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice. J Clin Invest. 2007 Dec;117(12):3786-99. doi: 10.1172/JCI32285.
-
Girtsman TA, Beamer CA, Wu N, Buford M, Holian A. IL-1R signalling is critical for regulation of multi-walled carbon nanotubes-induced acute lung inflammation in C57Bl/6 mice. Nanotoxicology. 2014 Feb;8(1):17-27. doi: 10.3109/17435390.2012.744110.
-
Glencross DA, Ho TR, Camiña N, Hawrylowicz CM, Pfeffer PE. Air pollution and its effects on the immune system. Free Radic Biol Med. 2020 May 1;151:56-68. doi: 10.1016/j.freeradbiomed.2020.01.179.
-
Gonçalves J, Juliano AM, Charepe N, Alenquer M, Athayde D, Ferreira F, Archer M, Amorim MJ, Serrano F, Soares H. Secretory IgA and T cells targeting SARS-CoV-2 spike protein are transferred to the breastmilk upon mRNA vaccination. Cell Rep Med. 2021 Dec 21;2(12):100468. doi: 10.1016/j.xcrm.2021.100468.
-
Gou W, Fu Y, Yue L, Chen GD, Cai X, Shuai M, Xu F, Yi X, Chen H, Zhu Y, Xiao ML, Jiang Z, Miao Z, Xiao C, Shen B, Wu X, Zhao H, Ling W, Wang J, Chen YM, Guo T, Zheng JS. Gut microbiota, inflammation, and molecular signatures of host response to infection. J Genet Genomics. 2021 Sep 20;48(9):792-802. doi: 10.1016/j.jgg.2021.04.002.
-
Gouwy M, De Buck M, Pörtner N, Opdenakker G, Proost P, Struyf S, Van Damme J. Serum amyloid A chemoattracts immature dendritic cells and indirectly provokes monocyte chemotaxis by induction of cooperating CC and CXC chemokines. Eur J Immunol. 2015 Jan;45(1):101-12. doi: 10.1002/eji.201444818.
-
Grenga L, Pible O, Miotello G, Culotta K, Ruat S, Roncato MA, Gas F, Bellanger L, Claret PG, Dunyach-Remy C, Laureillard D, Sotto A, Lavigne JP, Armengaud J. Taxonomical and functional changes in COVID-19 faecal microbiome could be related to SARS-CoV-2 faecal load. Environ Microbiol. 2022 Sep;24(9):4299-4316. doi: 10.1111/1462-2920.16028.
-
Gu S, Chen Y, Wu Z, Chen Y, Gao H, Lv L, Guo F, Zhang X, Luo R, Huang C, Lu H, Zheng B, Zhang J, Yan R, Zhang H, Jiang H, Xu Q, Guo J, Gong Y, Tang L, Li L. Alterations of the Gut Microbiota in Patients With Coronavirus Disease 2019 or H1N1 Influenza. Clin Infect Dis. 2020 Dec 17;71(10):2669-2678. doi: 10.1093/cid/ciaa709.
-
Hadrup N, Rahmani F, Jacobsen NR, Saber AT, Jackson P, Bengtson S, Williams A, Wallin H, Halappanavar S, Vogel U. Acute phase response and inflammation following pulmonary exposure to low doses of zinc oxide nanoparticles in mice. Nanotoxicology. 2019 Nov;13(9):1275-1292. doi: 10.1080/17435390.2019.1654004.
-
Halappanavar S, Nikota J, Wu D, Williams A, Yauk CL, Stampfli M. IL-1 receptor regulates microRNA-135b expression in a negative feedback mechanism during cigarette smoke-induced inflammation. J Immunol. 2013 Apr 1;190(7):3679-86. doi: 10.4049/jimmunol.1202456.
-
Han R, Hu M, Zhong Q, Wan C, Liu L, Li F, Zhang F, Ding W. Perfluorooctane sulphonate induces oxidative hepatic damage via mitochondria-dependent and NF-κB/TNF-α-mediated pathway. Chemosphere. 2018 Jan;191:1056-1064. doi: 10.1016/j.chemosphere.2017.08.070.
-
HARRIS H. Role of chemotaxis in inflammation. Physiol Rev. 1954 Jul;34(3):529-62. doi: 10.1152/physrev.1954.34.3.529.
-
Hassan MH, Elsadek AAM, Mahmoud MA, Elsadek BEM. Vitamin D Receptor Gene Polymorphisms and Risk of Knee Osteoarthritis: Possible Correlations with TNF-α, Macrophage Migration Inhibitory Factor, and 25-Hydroxycholecalciferol Status. Biochem Genet. 2022 Apr;60(2):611-628. doi: 10.1007/s10528-021-10116-0.
-
Helming L, Böse J, Ehrchen J, Schiebe S, Frahm T, Geffers R, Probst-Kepper M, Balling R, Lengeling A. 1alpha,25-Dihydroxyvitamin D3 is a potent suppressor of interferon gamma-mediated macrophage activation. Blood. 2005 Dec 15;106(13):4351-8. doi: 10.1182/blood-2005-03-1029.
-
Henry BM, Szergyuk I, de Oliveira MHS, Abosamak MF, Benoit SW, Benoit JL, Lippi G. Alterations in the lipid profile associate with a dysregulated inflammatory, prothrombotic, anti-fibrinolytic state and development of severe acute kidney injury in coronavirus disease 2019 (COVID-19): A study from Cincinnati, USA. Diabetes Metab Syndr. 2021 May-Jun;15(3):863-868. doi: 10.1016/j.dsx.2021.04.011.
-
Ho CC, Chang H, Tsai HT, Tsai MH, Yang CS, Ling YC, Lin P. Quantum dot 705, a cadmium-based nanoparticle, induces persistent inflammation and granuloma formation in the mouse lung. Nanotoxicology. 2013 Feb;7(1):105-15. doi: 10.3109/17435390.2011.635814.
-
Hofman Z, de Maat S, Hack CE, Maas C. Bradykinin: Inflammatory Product of the Coagulation System. Clin Rev Allergy Immunol. 2016 Oct;51(2):152-61. doi: 10.1007/s12016-016-8540-0.
-
Hubler MJ, Kennedy AJ. Role of lipids in the metabolism and activation of immune cells. J Nutr Biochem. 2016 Aug;34:1-7. doi: 10.1016/j.jnutbio.2015.11.002.
-
Husain M, Kyjovska ZO, Bourdon-Lacombe J, Saber AT, Jensen KA, Jacobsen NR, Williams A, Wallin H, Halappanavar S, Vogel U, Yauk CL. Carbon black nanoparticles induce biphasic gene expression changes associated with inflammatory responses in the lungs of C57BL/6 mice following a single intratracheal instillation. Toxicol Appl Pharmacol. 2015 Dec 15;289(3):573-88. doi: 10.1016/j.taap.2015.11.003.
-
Im SS, Yousef L, Blaschitz C, Liu JZ, Edwards RA, Young SG, Raffatellu M, Osborne TF. Linking lipid metabolism to the innate immune response in macrophages through sterol regulatory element binding protein-1a. Cell Metab. 2011 May 4;13(5):540-9. doi: 10.1016/j.cmet.2011.04.001
-
Jia H, Liu Y, Guo D, He W, Zhao L, Xia S. PM2.5-induced pulmonary inflammation via activating of the NLRP3/caspase-1 signaling pathway. Environmental Toxicology, 2021 Mar;36(3):298-307. doi: 10.1002/tox.23035.
-
Jorgensen SCJ, Tse CLY, Burry L, Dresser LD. Baricitinib: A Review of Pharmacology, Safety, and Emerging Clinical Experience in COVID-19. Pharmacotherapy. 2020 Aug;40(8):843-856. doi: 10.1002/phar.2438.
-
Kamata H, Tasaka S, Inoue K, Miyamoto K, Nakano Y, Shinoda H, Kimizuka Y, Fujiwara H, Ishii M, Hasegawa N, Takamiya R, Fujishima S, Takano H, Ishizaka A. Carbon black nanoparticles enhance bleomycin-induced lung inflammatory and fibrotic changes in mice. Exp Biol Med (Maywood). 2011 Mar;236(3):315-24. doi: 10.1258/ebm.2011.010180.
-
Khatri M, Bello D, Martin J, Bello A, Gore R, Demokritou P, et al. Chronic upper airway inflammation and systemic oxidative stress from nanoparticles in photocopier operators: Mechanistic insights. NanoImpact. 2017;5:133-45. doi: https://doi.org/10.1016/j.impact.2017.01.007.
-
Kido T, Tamagawa E, Bai N, Suda K, Yang HH, Li Y, Chiang G, Yatera K, Mukae H, Sin DD, Van Eeden SF. Particulate matter induces translocation of IL-6 from the lung to the systemic circulation. Am J Respir Cell Mol Biol. 2011 Feb;44(2):197-204. doi: 10.1165/rcmb.2009-0427OC.
-
Knoll R, Schultze JL, Schulte-Schrepping J. Monocytes and Macrophages in COVID-19. Front Immunol. 2021 Jul 21;12:720109. doi: 10.3389/fimmu.2021.720109.
-
Kovacs EJ, Boe DM, Boule LA, Curtis BJ. Inflammaging and the Lung. Clin Geriatr Med. 2017 Nov;33(4):459-471. doi: 10.1016/j.cger.2017.06.002.
-
Książek A, Zagrodna A, Bohdanowicz-Pawlak A, Lwow F, Słowińska-Lisowska M. Relationships between Vitamin D and Selected Cytokines and Hemogram Parameters in Professional Football Players-Pilot Study. Int J Environ Res Public Health. 2021 Jul 2;18(13):7124. doi: 10.3390/ijerph18137124.
-
Lee S, Hwang SH, Jeong J, Han Y, Kim SH, Lee DK, Lee HS, Chung ST, Jeong J, Roh C, Huh YS, Cho WS. Nickel oxide nanoparticles can recruit eosinophils in the lungs of rats by the direct release of intracellular eotaxin. Part Fibre Toxicol. 2016 Jun 9;13(1):30. doi: 10.1186/s12989-016-0142-8.
-
Leick M, Azcutia V, Newton G, Luscinskas FW. Leukocyte recruitment in inflammation: basic concepts and new mechanistic insights based on new models and microscopic imaging technologies. Cell Tissue Res. 2014 Mar;355(3):647-56. doi: 10.1007/s00441-014-1809-9.
-
Li R, Guo C, Lin X, Chan TF, Lai KP, Chen J. Integrative omics analyses uncover the mechanism underlying the immunotoxicity of perfluorooctanesulfonate in human lymphocytes. Chemosphere. 2020 Oct;256:127062. doi: 10.1016/j.chemosphere.2020.127062
-
Libby P. Fanning the flames: inflammation in cardiovascular diseases. Cardiovasc Res. 2015 Aug 1;107(3):307-9. doi: 10.1093/cvr/cvv188.
-
Liu W, Zheng W, Cheng L, Li M, Huang J, Bao S, Xu Q, Ma Z. Citrus fruits are rich in flavonoids for immunoregulation and potential targeting ACE2. Nat Prod Bioprospect. 2022 Feb 14;12(1):4. doi: 10.1007/s13659-022-00325-4.
-
Ljungman P, Bellander T, Schneider A, Breitner S, Forastiere F, Hampel R, Illig T, Jacquemin B, Katsouyanni K, von Klot S, Koenig W, Lanki T, Nyberg F, Pekkanen J, Pistelli R, Pitsavos C, Rosenqvist M, Sunyer J, Peters A. Modification of the interleukin-6 response to air pollution by interleukin-6 and fibrinogen polymorphisms. Environ Health Perspect. 2009 Sep;117(9):1373-9. doi: 10.1289/ehp.0800370.
-
Ma J, Li R, Qu G, Liu H, Yan B, Xia T, Liu Y, Liu S. Carbon nanotubes stimulate synovial inflammation by inducing systemic pro-inflammatory cytokines. Nanoscale. 2016 Oct 27;8(42):18070-18086. doi: 10.1039/c6nr06041b.
-
Marchini T, Wolf D, Michel NA, Mauler M, Dufner B, Hoppe N, Beckert J, Jäckel M, Magnani N, Duerschmied D, Tasat D, Alvarez S, Reinöhl J, von Zur Muhlen C, Idzko M, Bode C, Hilgendorf I, Evelson P, Zirlik A. Acute exposure to air pollution particulate matter aggravates experimental myocardial infarction in mice by potentiating cytokine secretion from lung macrophages. Basic Res Cardiol. 2016 Jul;111(4):44. doi: 10.1007/s00395-016-0562-5.
-
Mauvais-Jarvis F, Bairey Merz N, Barnes PJ, Brinton RD, Carrero JJ, DeMeo DL, De Vries GJ, Epperson CN, Govindan R, Klein SL, Lonardo A, Maki PM, McCullough LD, Regitz-Zagrosek V, Regensteiner JG, Rubin JB, Sandberg K, Suzuki A. Sex and gender: modifiers of health, disease, and medicine. Lancet. 2020 Aug 22;396(10250):565-582. doi: 10.1016/S0140-6736(20)31561-0.
-
Morimoto Y, Ogami A, Todoroki M, Yamamoto M, Murakami M, Hirohashi M, Oyabu T, Myojo T, Nishi K, Kadoya C, Yamasaki S, Nagatomo H, Fujita K, Endoh S, Uchida K, Yamamoto K, Kobayashi N, Nakanishi J, Tanaka I. Expression of inflammation-related cytokines following intratracheal instillation of nickel oxide nanoparticles. Nanotoxicology. 2010 Jun;4(2):161-76. doi: 10.3109/17435390903518479
-
Munshi R, Hussein MH, Toraih EA, Elshazli RM, Jardak C, Sultana N, Youssef MR, Omar M, Attia AS, Fawzy MS, Killackey M, Kandil E, Duchesne J. Vitamin D insufficiency as a potential culprit in critical COVID-19 patients. J Med Virol. 2021 Feb;93(2):733-740. doi: 10.1002/jmv.26360.
-
Muscogiuri G, Pugliese G, Barrea L, Savastano S, Colao A. Commentary: Obesity: The "Achilles heel" for COVID-19? Metabolism. 2020 Jul;108:154251. doi: 10.1016/j.metabol.2020.154251.
-
Neveu G, Ziv-Av A, Barouch-Bentov R, Berkerman E, Mulholland J, Einav S. AP-2-associated protein kinase 1 and cyclin G-associated kinase regulate hepatitis C virus entry and are potential drug targets. J Virol. 2015 Apr;89(8):4387-404. doi: 10.1128/JVI.02705-14.
-
Nikota J, Banville A, Goodwin LR, Wu D, Williams A, Yauk CL, Wallin H, Vogel U, Halappanavar S. Stat-6 signaling pathway and not Interleukin-1 mediates multi-walled carbon nanotube-induced lung fibrosis in mice: insights from an adverse outcome pathway framework. Part Fibre Toxicol. 2017 Sep 13;14(1):37. doi: 10.1186/s12989-017-0218-0.
-
Patowary P, Pathak MP, Zaman K, Dwivedi SK, Chattopadhyay P. Innate inflammatory response to acute inhalation exposure of riot control agent oleoresin capsicum in female rats: An interplay between neutrophil mobilization and inflammatory markers. Exp Lung Res. 2020 Apr-May;46(3-4):81-97. doi: 10.1080/01902148.2020.1733709.
-
Petri B, Sanz MJ. Neutrophil chemotaxis. Cell Tissue Res. 2018 Mar;371(3):425-436. doi: 10.1007/s00441-017-2776-8.
-
Porter DW, Ye J, Ma J, Barger M, Robinson VA, Ramsey D, McLaurin J, Khan A, Landsittel D, Teass A, Castranova V. Time course of pulmonary response of rats to inhalation of crystalline silica: NF-kappa B activation, inflammation, cytokine production, and damage. Inhal Toxicol. 2002 Apr;14(4):349-67. doi: 10.1080/08958370252870998.
-
Porter DW, Orandle M, Zheng P, Wu N, Hamilton RF Jr, Holian A, Chen BT, Andrew M, Wolfarth MG, Battelli L, Tsuruoka S, Terrones M, Castranova V. Mouse pulmonary dose- and time course-responses induced by exposure to nitrogen-doped multi-walled carbon nanotubes. Inhal Toxicol. 2020 Jan;32(1):24-38. doi: 10.1080/08958378.2020.1723746.
-
Poulsen SS, Jacobsen NR, Labib S, Wu D, Husain M, Williams A, Bøgelund JP, Andersen O, Købler C, Mølhave K, Kyjovska ZO, Saber AT, Wallin H, Yauk CL, Vogel U, Halappanavar S. Transcriptomic analysis reveals novel mechanistic insight into murine biological responses to multi-walled carbon nanotubes in lungs and cultured lung epithelial cells. PLoS One. 2013 Nov 19;8(11):e80452. doi: 10.1371/journal.pone.0080452.
-
Rahman L, Wu D, Johnston M, William A, Halappanavar S. Toxicogenomics analysis of mouse lung responses following exposure to titanium dioxide nanomaterials reveal their disease potential at high doses. Mutagenesis. 2017 Jan;32(1):59-76. doi: 10.1093/mutage/gew048.
-
Richardson P, Griffin I, Tucker C, Smith D, Oechsle O, Phelan A, Rawling M, Savory E, Stebbing J. Baricitinib as potential treatment for 2019-nCoV acute respiratory disease. Lancet. 2020 Feb 15;395(10223):e30-e31. doi: 10.1016/S0140-6736(20)30304-4.
-
Rodrigues Prestes TR, Rocha NP, Miranda AS, Teixeira AL, Simoes-E-Silva AC. The Anti-Inflammatory Potential of ACE2/Angiotensin-(1-7)/Mas Receptor Axis: Evidence from Basic and Clinical Research. Curr Drug Targets. 2017;18(11):1301-1313. doi: 10.2174/1389450117666160727142401
-
Rowley AH. Understanding SARS-CoV-2-related multisystem inflammatory syndrome in children. Nat Rev Immunol. 2020 Aug;20(8):453-454. doi: 10.1038/s41577-020-0367-5.
-
Schremmer I, Brik A, Weber DG, Rosenkranz N, Rostek A, Loza K, Brüning T, Johnen G, Epple M, Bünger J, Westphal GA. Kinetics of chemotaxis, cytokine, and chemokine release of NR8383 macrophages after exposure to inflammatory and inert granular insoluble particles. Toxicol Lett. 2016 Nov 30;263:68-75. doi: 10.1016/j.toxlet.2016.08.014.
-
Scully EP, Haverfield J, Ursin RL, Tannenbaum C, Klein SL. Considering how biological sex impacts immune responses and COVID-19 outcomes. Nat Rev Immunol. 2020 Jul;20(7):442-447. doi: 10.1038/s41577-020-0348-8.
-
Sencio V, Machelart A, Robil C, Benech N, Hoffmann E, Galbert C, Deryuter L, Heumel S, Hantute-Ghesquier A, Flourens A, Brodin P, Infanti F, Richard V, Dubuisson J, Grangette C, Sulpice T, Wolowczuk I, Pinet F, Prévot V, Belouzard S, Briand F, Duterque-Coquillaud M, Sokol H, Trottein F. Alteration of the gut microbiota following SARS-CoV-2 infection correlates with disease severity in hamsters. Gut Microbes. 2022 Jan-Dec;14(1):2018900. doi: 10.1080/19490976.2021.2018900.
-
Signorini C, Pignatti P, Coccini T. How Do Inflammatory Mediators, Immune Response and Air Pollution Contribute to COVID-19 Disease Severity? A Lesson to Learn. Life (Basel). 2021 Feb 25;11(3):182. doi: 10.3390/life11030182.
-
Singh D, Wasan H, Reeta KH. Heme oxygenase-1 modulation: A potential therapeutic target for COVID-19 and associated complications. Free Radic Biol Med. 2020 Dec;161:263-271. doi: 10.1016/j.freeradbiomed.2020.10.016.
-
Song JA, Park HJ, Yang MJ, Jung KJ, Yang HS, Song CW, Lee K. Polyhexamethyleneguanidine phosphate induces severe lung inflammation, fibrosis, and thymic atrophy. Food Chem Toxicol. 2014 Jul;69:267-75. doi: 10.1016/j.fct.2014.04.027.
-
Sørli JB, Låg M, Ekeren L, Perez-Gil J, Haug LS, Da Silva E, Matrod MN, Gützkow KB, Lindeman B. Per- and polyfluoroalkyl substances (PFASs) modify lung surfactant function and pro-inflammatory responses in human bronchial epithelial cells. Toxicol In Vitro. 2020 Feb;62:104656. doi: 10.1016/j.tiv.2019.104656.
-
Takahashi T, Ellingson MK, Wong P, Israelow B, Lucas C, Klein J, Silva J, Mao T, Oh JE, Tokuyama M, Lu P, Venkataraman A, Park A, Liu F, Meir A, Sun J, Wang EY, Casanovas-Massana A, Wyllie AL, Vogels CBF, Earnest R, Lapidus S, Ott IM, Moore AJ; Yale IMPACT Research Team; Shaw A, Fournier JB, Odio CD, Farhadian S, Dela Cruz C, Grubaugh ND, Schulz WL, Ring AM, Ko AI, Omer SB, Iwasaki A. Sex differences in immune responses that underlie COVID-19 disease outcomes. Nature. 2020 Dec;588(7837):315-320. doi: 10.1038/s41586-020-2700-3.
-
Tian J, Hong Y, Li Z, Yang Z, Lei B, Liu J, Cai Z. Immunometabolism-modulation and immunotoxicity evaluation of perfluorooctanoic acid in macrophage. Ecotoxicol Environ Saf. 2021 Jun 1;215:112128. doi: 10.1016/j.ecoenv.2021.112128.
-
Tsai DH, Amyai N, Marques-Vidal P, Wang JL, Riediker M, Mooser V, Paccaud F, Waeber G, Vollenweider P, Bochud M. Effects of particulate matter on inflammatory markers in the general adult population. Particle and Fibre Toxicolgy. 2012 Jul 6;9:24. doi: 10.1186/1743-8977-9-24
-
Villeneuve DL, Landesmann B, Allavena P, Ashley N, Bal-Price A, Corsini E, Halappanavar S, Hussell T, Laskin D, Lawrence T, Nikolic-Paterson D, Pallardy M, Paini A, Pieters R, Roth R, Tschudi-Monnet F. Representing the Process of Inflammation as Key Events in Adverse Outcome Pathways. Toxicol Sci. 2018 Jun 1;163(2):346-352. doi: 10.1093/toxsci/kfy047.
-
Vinolo MA, Rodrigues HG, Nachbar RT, Curi R. Regulation of inflammation by short chain fatty acids. Nutrients. 2011 Oct;3(10):858-76. doi: 10.3390/nu3100858.
-
Wang G, Zhang X, Liu X, Zheng J, Chen R, Kan H. Ambient fine particulate matter induce toxicity in lung epithelial-endothelial co-culture models. Toxicol Lett. 2019 Feb;301:133-145. doi: 10.1016/j.toxlet.2018.11.010.
-
Westphal GA, Schremmer I, Rostek A, Loza K, Rosenkranz N, Brüning T, Epple M, Bünger J. Particle-induced cell migration assay (PICMA): A new in vitro assay for inflammatory particle effects based on permanent cell lines. Toxicol In Vitro. 2015 Aug;29(5):997-1005. doi: 10.1016/j.tiv.2015.04.005.
-
World Health Organization, 2021. Therapeutics and COVID-19: Living Guideline. WHO/2019-NCoV/Therapeutics/2021.1'. IL-6 Receptor Blockers 2021. Available online: https://www.who.int/publications/i/item/WHO-2019-nCoV-therapeutics-2022.4 (accessed 12 May 2022).
-
Winer S, Chan Y, Paltser G, Truong D, Tsui H, Bahrami J, Dorfman R, Wang Y, Zielenski J, Mastronardi F, Maezawa Y, Drucker DJ, Engleman E, Winer D, Dosch HM. Normalization of obesity-associated insulin resistance through immunotherapy. Nat Med. 2009 Aug;15(8):921-9. doi: 10.1038/nm.2001
-
Yamasaki K, Eeden SFV. Lung Macrophage Phenotypes and Functional Responses: Role in the Pathogenesis of COPD. Int J Mol Sci. 2018 Feb 15;19(2):582. doi: 10.3390/ijms19020582.
-
Yeoh YK, Zuo T, Lui GC, Zhang F, Liu Q, Li AY, Chung AC, Cheung CP, Tso EY, Fung KS, Chan V, Ling L, Joynt G, Hui DS, Chow KM, Ng SSS, Li TC, Ng RW, Yip TC, Wong GL, Chan FK, Wong CK, Chan PK, Ng SC. Gut microbiota composition reflects disease severity and dysfunctional immune responses in patients with COVID-19. Gut. 2021 Apr;70(4):698-706. doi: 10.1136/gutjnl-2020-323020.
-
Yoo JY, Groer M, Dutra SVO, Sarkar A, McSkimming DI. Gut Microbiota and Immune System Interactions. Microorganisms. 2020 Oct 15;8(10):1587. doi: 10.3390/microorganisms8101587.
-
Zhang F, Wan Y, Zuo T, Yeoh YK, Liu Q, Zhang L, Zhan H, Lu W, Xu W, Lui GCY, Li AYL, Cheung CP, Wong CK, Chan PKS, Chan FKL, Ng SC. Prolonged Impairment of Short-Chain Fatty Acid and L-Isoleucine Biosynthesis in Gut Microbiome in Patients With COVID-19. Gastroenterology. 2022 Feb;162(2):548-561.e4. doi: 10.1053/j.gastro.2021.10.013.
-
Zhao J, Gao Z, Tian Z, Xie Y, Xin F, Jiang R, Kan H, Song W. The biological effects of individual-level PM(2.5) exposure on systemic immunity and inflammatory response in traffic policemen. Occup Environ Med. 2013 Jun;70(6):426-31. doi: 10.1136/oemed-2012-100864.
-
Zuo T, Zhang F, Lui GCY, Yeoh YK, Li AYL, Zhan H, Wan Y, Chung ACK, Cheung CP, Chen N, Lai CKC, Chen Z, Tso EYK, Fung KSC, Chan V, Ling L, Joynt G, Hui DSC, Chan FKL, Chan PKS, Ng SC. Alterations in Gut Microbiota of Patients With COVID-19 During Time of Hospitalization. Gastroenterology. 2020 Sep;159(3):944-955.e8. doi: 10.1053/j.gastro.2020.05.048.