Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Front Immunol ; 14: 1185233, 2023.
Article in English | MEDLINE | ID: covidwho-20244458

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is a contagious respiratory virus that is the cause of the coronavirus disease 2019 (COVID-19) pandemic which has posed a serious threat to public health. COVID-19 is characterized by a wide spectrum of clinical manifestations, ranging from asymptomatic infection to mild cold-like symptoms, severe pneumonia or even death. Inflammasomes are supramolecular signaling platforms that assemble in response to danger or microbial signals. Upon activation, inflammasomes mediate innate immune defense by favoring the release of proinflammatory cytokines and triggering pyroptotic cell death. Nevertheless, abnormalities in inflammasome functioning can result in a variety of human diseases such as autoimmune disorders and cancer. A growing body of evidence has showed that SARS-CoV-2 infection can induce inflammasome assembly. Dysregulated inflammasome activation and consequent cytokine burst have been associated with COVID-19 severity, alluding to the implication of inflammasomes in COVID-19 pathophysiology. Accordingly, an improved understanding of inflammasome-mediated inflammatory cascades in COVID-19 is essential to uncover the immunological mechanisms of COVID-19 pathology and identify effective therapeutic approaches for this devastating disease. In this review, we summarize the most recent findings on the interplay between SARS-CoV-2 and inflammasomes and the contribution of activated inflammasomes to COVID-19 progression. We dissect the mechanisms involving the inflammasome machinery in COVID-19 immunopathogenesis. In addition, we provide an overview of inflammasome-targeted therapies or antagonists that have potential clinical utility in COVID-19 treatment.


Subject(s)
COVID-19 , Humans , Inflammasomes/metabolism , SARS-CoV-2/physiology , COVID-19 Drug Treatment , Cytokines
2.
Cureus ; 14(9): e29760, 2022 Sep.
Article in English | MEDLINE | ID: covidwho-20231825

ABSTRACT

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly contagious and has taken an enormous toll on the worldwide quality of life and the global economy, in addition to the lives lost due to coronavirus disease 2019 (COVID-19). Precautionary measures and timely identification of the infected cases are essential to minimize the spread of SARS-CoV-2. Infection with this virus causes a spike in the proinflammatory cytokines, resulting in immune system-mediated host tissue damage, thus leading to mortality. Therefore, identifying mild, moderate, and severe cases is crucial to rendering appropriate care. Recent research has focused on identifying laboratory techniques to predict the case severity and outcome of COVID-19 cases. Low serum lymphocyte levels, low lymphocyte-to-C-reactive protein ratio, low platelet-to-lymphocyte ratio, thrombocytopenia, and high neutrophil-lymphocyte ratio (NLR) have been observed in critical infections. NLR might be a prognostic marker for disease severity. Severe cases can be triaged at hospital admission for proper treatment planning and to reduce mortality. This review highlights the potential role of NLR hematological assay in SARS-CoV-2 infection and the mechanism of neutrophilic-induced host tissue damage.

3.
J Interferon Cytokine Res ; 43(6): 257-268, 2023 Jun.
Article in English | MEDLINE | ID: covidwho-20242330

ABSTRACT

Despite extensive research to decipher the immunological basis of coronavirus disease (COVID-19), limited evidence on immunological correlates of COVID-19 severity from MENA region and Egypt was reported. In a single-center cross-sectional study, we have analyzed 25 cytokines that are related to immunopathologic lung injury, cytokine storm, and coagulopathy in plasma samples from 78 hospitalized Egyptian COVID-19 patients in Tanta University Quarantine Hospital and 21 healthy control volunteers between April 2020 and September 2020. The enrolled patients were divided into 4 categories based on disease severity, namely mild, moderate, severe, and critically ill. Interestingly, interleukin (IL)-1-α, IL-2Rα, IL-6, IL-8, IL-18, tumor necrosis factor-alpha (TNF-α), FGF1, CCL2, and CXC10 levels were significantly altered in severe and/or critically ill patients. Moreover, principal component analysis (PCA) demonstrated that severe and critically ill COVID-19 patients cluster based on specific cytokine signatures that distinguish them from mild and moderate COVID-19 patients. Specifically, levels of IL-2Rα, IL-6, IL-10, IL-18, TNF-α, FGF1, and CXCL10 largely contribute to the observed differences between early and late stages of COVID-19 disease. Our PCA showed that the described immunological markers positively correlate with high D-dimer and C-reactive protein levels and inversely correlate with lymphocyte counts in severe and critically ill patients. These data suggest a disordered immune regulation, particularly in severe and critically ill Egyptian COVID-19 patients, manifested as overactivated innate immune and dysregulated T-helper1 responses. Additionally, our study emphasizes the importance of cytokine profiling to identify potentially predictive immunological signatures of COVID-19 disease severity.


Subject(s)
COVID-19 , Cytokines , Humans , Interleukin-18 , Cross-Sectional Studies , Egypt , Interleukin-6 , Tumor Necrosis Factor-alpha , Critical Illness , Interleukin-2 Receptor alpha Subunit , Fibroblast Growth Factor 1 , Patient Acuity
4.
Immunobiology ; 228(3): 152384, 2023 05.
Article in English | MEDLINE | ID: covidwho-2303646

ABSTRACT

INTRODUCTION: COVID-19 Associated Mucormycosis (CAM), an opportunistic fungal infection, surged during the second wave of SARS Cov-2 pandemic. Since immune responses play an important role in controlling this infection in immunocompetent hosts, it is required to understand immune perturbations associated with this condition for devising immunotherapeutic strategies for its control. We conducted a study to determine different immune parameters altered in CAM cases as compared to COVID-19 patients without CAM. METHODOLOGY: Cytokine levels in serum samples of CAM cases (n = 29) and COVID-19 patients without CAM (n = 20) were determined using luminex assay. Flow cytometric assays were carried out in 20 CAM cases and 10 controls for determination of frequency of NK cells, DCs, phagocytes, T cells and their functionalities. The cytokine levels were analyzed for their association with each other as well as with T cell functionality. The immune parameters were also analyzed with respect to the known risk factors such as diabetes mellitus and steroid treatment. RESULTS: Significant reduction in frequencies of total and CD56 + CD16 + NK cells (cytotoxic subset) was noted in CAM cases. Degranulation responses indicative of cytotoxicity of T cell were significantly hampered in CAM cases as compared to the controls. Conversely, phagocytic functions showed no difference in CAM cases versus their controls except for migratory potential which was found to be enhanced in CAM cases. Levels of proinflammatory cytokines such as IFN-γ, IL-2, TNF-α, IL-17, IL-1ß, IL-18 and MCP-1 were significantly elevated in cases as compared to the control with IFN-γ and IL-18 levels correlating negatively with CD4 T cell cytotoxicity. Steroid administration was associated with higher frequency of CD56 + CD16- NK cells (cytokine producing subset) and higher MCP-1 levels. Whereas diabetic participants had higher phagocytic and chemotactic potential and had higher levels of IL-6, IL-17 and MCP-1. CONCLUSION: CAM cases differed from the controls in terms of higher titers of proinflammatory cytokines, reduced frequency of total and cytotoxic CD56 + CD16 + NK cell. They also had reduced T cell cytotoxicity correlating inversely with IFN-γ and IL-18 levels, possibly indicating induction of negative feedback mechanisms while diabetes mellitus or steroid administration did not affect the responses negatively.


Subject(s)
COVID-19 , Mucormycosis , Humans , Interleukin-18 , Interleukin-17 , Cytokines , Steroids
5.
Viral Immunol ; 36(4): 282-289, 2023 05.
Article in English | MEDLINE | ID: covidwho-2248975

ABSTRACT

Middle East respiratory syndrome coronavirus (MERS-CoV) is associated with significant morbidity and mortality. This study was performed to assess the proinflammatory cytokines profile among MERS-CoV patients. A total of 46 MERS-CoV-infected patients (27 symptomatic and 19 asymptomatic) were assessed and compared with 52 normal healthy controls for plasma levels of interleukin (IL)-1ß, tumor necrosis factor (TNF)-α, IL-17, IL-7, IL-6, interferon (IFN)-α, and IL-15 using a customized luminex kit. Whereas asymptomatic MERS-CoV patients and controls were no different; the mean plasma levels among MERS-CoV symptomatic patients were significantly higher than the normal controls: IL-1ß (16.89 ± 1.23 vs. 12.80 ± 0.59 pg/mL; p < 0.001), TNF-α (14.04 ± 0.93 vs. 10.35 ± 0.29 pg/mL; p < 0.0001), IL-17 (14.3 ± 0.89 vs. 11.47 ± 0.61 pg/mL; p < 0.001), IL-7 (21.56 ± 1.00 vs. 16.31 ± 0.30 pg/mL; p < 0.0001), IL-6 (156.5 ± 37.90 vs. 18.60 ± 1.59 pg/mL; p < 0.0001), and IFN-α (68.73 ± 13.06 vs. 23.57 ± 1.05 pg/mL; p < 0.0001). The mean plasma levels of IL-7 (24.81 ± 1.63 vs. 19.79 ± 0.94 pg/mL; p < 0.01), IL-6 (312.7 ± 94.67 vs. 101.2 ± 25.67 pg/mL; p < 0.01), and IFN-α (89.00 ± 18.97 vs. 51.05 ± 8.68 pg/mL; p < 0.05) were significantly elevated among MERS-CoV symptomatic patients with fatal outcome compared with MERS-CoV symptomatic patients who survived. Only IL-7 was found to have a higher risk ratio of mortality (4.76, 95% confidence interval: 1.5-14.94; p < 0.01). No differences were observed in IL-15 levels among the groups. Significantly elevated proinflammatory cytokines among symptomatic MERS-CoV-infected patients may contribute to manifestations of cytokine storm frequently observed among critically ill MERS-CoV patients and IL-7 may serve as a marker for disease activity.


Subject(s)
Coronavirus Infections , Middle East Respiratory Syndrome Coronavirus , Humans , Cytokines , Interleukin-15 , Interleukin-17 , Interleukin-6 , Interleukin-7 , Interferon-alpha
6.
Arch Razi Inst ; 78(1): 305-313, 2023 02.
Article in English | MEDLINE | ID: covidwho-2248109

ABSTRACT

Due to the pandemic of COVID -19 disease and the fact that the effective variables in the severity and control of the disease have not been established, numerous factors have been investigated, including the study of inflammatory factors. A cross-sectional study was carried out to investigate the proinflammatory cytokines in patients with COVID -19, conducted in Baghdad, Iraq. The age of the patients was above > (15) years old, with confirmed infection documented by polymerase chain reaction (PCR). The subjects were 132 patients, 69 (52.3%) males, and 63 (47.7%) females. Patients were divided into three pathological groups: mild patients (45), moderate patients (34), and severe patients (53), each group was divided into four weeks according to symptoms onset date. The most common clinical symptoms were cough, fever, and headache, while sore throat, gastrointestinal symptoms, chest pain, and loss of taste and smell were less common in COVID -19 patients. Sandwich-Enzyme-Linked Immunosorbent Assay kits were used to evaluate levels of proinflammatory cytokines, including IL-1ß, IL-6, IL-8, and TNF-α. The results IL-6 and TNF-α were significantly elevated in mild during the four weeks with (P=0.0071) and (0.0266) respectively, levels of IL-1ß were increased with highly significant differences (P=0.0001) while levels of IL-8 were decreased with highly significant differences (P=0.0001) during the four weeks. In moderate patients, levels of (IL-1ß, IL-6, and IL-8) increased without significance (P=0.661, 0.074, 0.0651), respectively; in contrast, the levels of TNF-α increased with significant (P=0.0452) across four weeks. Severe COVID-19 patients showed significantly increased differences in levels of (IL-6, IL-8, and TNFα) (P=0.0438, 0.0348, 0.0447), respectively, while no significant differences in the level of IL-1ß (P=0.0774). This study showed that investigating inflammatory factors in the COVID-19 pandemic is crucial in controlling and treating.


Subject(s)
COVID-19 , Cytokines , Female , Humans , Male , Cross-Sectional Studies , Interleukin-6 , Interleukin-8 , Iraq/epidemiology , Pandemics , Tumor Necrosis Factor-alpha
7.
Inflamm Regen ; 43(1): 7, 2023 Jan 26.
Article in English | MEDLINE | ID: covidwho-2237199

ABSTRACT

Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) is the causative virus of pandemic acute respiratory disease called coronavirus disease 2019 (COVID-19). Most of the infected individuals have asymptomatic or mild symptoms, but some patients show severe and critical systemic inflammation including tissue damage and multi-organ failures. Immune responses to the pathogen determine clinical course. In general, the activation of innate immune responses is mediated by host pattern-recognition receptors (PRRs) that recognize pathogen-associated molecular patterns (PAMPs) as well as host damage-associated molecular patterns (DAMPs), which results in the activation of the downstream gene induction programs of types I and III interferons (IFNs) and proinflammatory cytokines for inducing antiviral activity. However, the excessive activation of these responses may lead to deleterious inflammation. Here, we review the recent advances in our understanding of innate immune responses to SARS-CoV-2 infection, particularly in terms of innate recognition and the subsequent inflammation underlying COVID-19 immunopathology.

8.
Coronavirus Drug Discovery: Volume 1: SARS-CoV-2 (COVID-19) Prevention, Diagnosis, and Treatment ; : 169-179, 2022.
Article in English | Scopus | ID: covidwho-2048785

ABSTRACT

The corticosteroid drug “dexamethasone” has been in use since 1960s for reducing inflammation in a variety of conditions such as certain cancers and other inflammatory disorders. It is an affordable agent and currently off-patent in most countries and listed in multiple formulations since 1977 in the World Health Organization model list of essential medicines. The cytokines production and damaging effect has been limited through the use of dexamethasone and this will also block B cells from antibodies production and inhibit the T cell's protective function potential leading to elevated viral load in the plasma that persists for longer time after a patient survives SARS. In addition, dexamethasone would chunk the macrophages from clearing the resultant nosocomial infections. Thus, dexamethasone may be valuable for the immediate relief in severe cases of COVID-19, but could be dangerous on the long run as the body will be barred from producing protective antibodies in addition to the persistence of the virus. © 2022 Elsevier Inc. All rights reserved.

9.
Int J Mol Sci ; 23(16)2022 Aug 22.
Article in English | MEDLINE | ID: covidwho-1997648

ABSTRACT

The aims of our study are to: (i) investigate the ability of nicotine to modulate the expression level of inflammatory cytokines in A549 cells infected with SARS-CoV-2; (ii) elucidate the ultrastructural features caused by the combination nicotine+SARS-CoV-2; and (iii) demonstrate the mechanism of action. In this study, A549 cells pretreated with nicotine were either exposed to LPS or poly(I:C), or infected with SARS-CoV-2. Treated and untreated cells were analyzed for cytokine production, cytotoxicity, and ultrastructural modifications. Vero E6 cells were used as a positive reference. Cells pretreated with nicotine showed a decrease of IL6 and TNFα in A549 cells induced by LPS or poly(I:C). In contrast, cells exposed to SARS-CoV-2 showed a high increase of IL6, IL8, IL10 and TNFα, high cytopathic effects that were dose- and time-dependent, and profound ultrastructural modifications. These modifications were characterized by membrane ruptures and fragmentation, the swelling of cytosol and mitochondria, the release of cytoplasmic content in extracellular spaces (including osmiophilic granules), the fragmentation of endoplasmic reticulum, and chromatin disorganization. Nicotine increased SARS-CoV-2 cytopathic effects, elevating the levels of inflammatory cytokines, and inducing severe cellular damage, with features resembling pyroptosis and necroptosis. The protective role of nicotine in COVID-19 is definitively ruled out.


Subject(s)
Nicotine , SARS-CoV-2 , A549 Cells , COVID-19 , Cell Survival/drug effects , Cytokines/metabolism , Humans , Interleukin-6 , Lipopolysaccharides , Nicotine/adverse effects , Nicotine/pharmacology , Tumor Necrosis Factor-alpha
10.
1st International Conference on Technologies for Smart Green Connected Society 2021, ICTSGS 2021 ; 107:17797-17804, 2022.
Article in English | Scopus | ID: covidwho-1950332

ABSTRACT

INTRODUCTION: The ultra-modern new coronavirus disease (COVID-19) outbreak, that's resulting from the excessive acute breathing syndrome coronavirus 2 (SARS-CoV2), is seeing a dramatic upward thrust in inflamed people all around the world. In the aetiology and scientific signs of SARS-CoV2, the host immune reaction seems to be crucial. In sufferers with excessive COVID-19, SARS-CoV2 now no longer handiest turns on antiviral immune responses, however it could additionally cause out of control inflammatory responses characterized with the aid of using excessive ranges of pro-inflammatory cytokines, ensuing in lymphopenia, lymphocyte dysfunction, and granulocyte and monocyte abnormalities. These immunological abnormalities resulting from SARS-CoV2 may want to cause microbial infections, septic shock, and excessive a couple of organ failure. As a result, the approaches underlying immunological abnormalities in COVID-19 sufferers want to be explored if you want to manual scientific care of the disease. Furthermore, sensible law of SARS-CoV2 immune responses, which incorporates growing antiviral immunity at the same time as restricting systemic inflammation, will be important to a hit treatment. CONCLUSION: SARS-CoV2 has produced a global public health emergency, which promotes greater efforts to assess viral transmission, pathogenesis, and immune responses, as well as improve disease prevention and treatment strategies. Data from these scientific endeavors have shown that managing the interaction between the virus and the immune system is important in reducing morbidity and mortality, in addition to opening up the development of vaccines and vaccination processes. © The Electrochemical Society

11.
Biomedical and Biotechnology Research Journal ; 6(2):170-174, 2022.
Article in English | Scopus | ID: covidwho-1924404

ABSTRACT

Background: The cytokine storm and specific biochemical results were both observed during Coronavirus disease 2019 (COVID-19) infection. The aim of this study was to see if there was any correlation between biochemical findings and cytokine levels. Methods: A total of 65 COVID-19 patients, including 32 males and 33 females with ages between (16 and 90 years) were enrolled in this study. Between December 2020 and February 2021, these patients were admitted to Al-Amal Specialized Hospital or ward of COVID-19 care in Al-Sader Hospital. These patients were classified into severe/critical (43 cases), mild/moderate (22 cases) according to the guidelines released by National Health World depending on SpO 2 percentage. The biochemical indices were measured using FUJI DRI-CHEM NX500 automated clinical chemistry analyzer (FUJIFILM, Japan). The proinflammatory cytokine (interleukin-6 [IL-6], IL-1α, and IL-1β) were measured using ELISA technique. Results: IL-6 serum levels were negatively correlated with SpO2 (P = 0.002, R = - 0.372) and serum albumin (P = 0.034, R = - 0.301). IL-1α serum levels showed significant negative correlation with serum albumin levels (P = 0.039, R = - 0.259). Furthermore, a positive correlation found between IL-1β serum with serum levels of both AST and LDH (P = 0.049;R = 0.255;P = 0.054;R = 0.320 respectively). Discussion: Increased IL-6 serum levels have a direct impact on SpO2 percentage and serum albumin. Whereas the elevated levels of both IL-1α and IL-1β are a possible cause of acute inflammation and liver damage in COVID-19. Conclusions: This study further confirms the growing evidence on the direct role of proinflammatory cytokines in the biochemical changes shown in COVID-19. © 2022 EDP Sciences. All rights reserved.

12.
Front Immunol ; 13: 876555, 2022.
Article in English | MEDLINE | ID: covidwho-1809408

ABSTRACT

SARS-CoV-2 infects cells via binding to ACE2 and TMPRSS2, which allows the virus to fuse with host cells. The viral RNA is detected in the placenta of SARS-CoV-2-infected pregnant women and infection is associated with adverse pregnancy complications. Therefore, we hypothesize that SARS-CoV-2 infection of placental cells induces pro-inflammatory cytokine release to contribute to placental dysfunction and impaired pregnancy outcomes. First, expression of ACE2 and TMPRSS2 was measured by qPCR in human primary cultured term cytotrophoblasts (CTBs), syncytiotrophoblast (STBs), term and first trimester decidual cells (TDCs and FTDCs, respectively), endometrial stromal cells (HESCs) as well as trophoblast cell lines HTR8, JEG3, placental microvascular endothelial cells (PMVECs) and endometrial endothelial cells (HEECs). Later, cultured HTR8, JEG3, PMVECs and HEECs were treated with 10, 100, 1000 ng/ml of recombinant (rh-) SARS-CoV-2 S-protein ± 10 ng/ml rh-IFNγ. Pro-inflammatory cytokines IL-1ß, 6 and 8, chemokines CCL2, CCL5, CXCL9 and CXCL10 as well as tissue factor (F3), the primary initiator of the extrinsic coagulation cascade, were measured by qPCR as well as secreted IL-6 and IL-8 levels were measured by ELISA. Immunohistochemical staining for SARS-CoV-2 spike protein was performed in placental specimens from SARS-CoV-2-positive and normal pregnancies. ACE2 levels were significantly higher in CTBs and STBs vs. TDCs, FTDCs and HESCs, while TMPRSS2 levels were not detected in TDCs, FTDCs and HESCs. HTR8 and JEG3 express ACE2 and TMPRSS2, while PMVECs and HEECs express only ACE2, but not TMPRSS2. rh-S-protein increased proinflammatory cytokines and chemokines levels in both trophoblast and endothelial cells, whereas rh-S-protein only elevated F3 levels in endothelial cells. rh-IFNγ ± rh-S-protein augments expression of cytokines and chemokines in trophoblast and endothelial cells. Elevated F3 expression by rh-IFNγ ± S-protein was observed only in PMVECs. In placental specimens from SARS-CoV-2-infected mothers, endothelial cells displayed higher immunoreactivity against spike protein. These findings indicated that SARS-CoV-2 infection in placental cells: 1) induces pro-inflammatory cytokine and chemokine release, which may contribute to the cytokine storm observed in severely infected pregnant women and related placental dysfunction; and 2) elevates F3 expression that may trigger systemic or placental thrombosis.


Subject(s)
COVID-19 , Placenta Diseases , Pregnancy Complications, Infectious , Angiotensin-Converting Enzyme 2 , Cell Line, Tumor , Cytokines/metabolism , Endothelial Cells/pathology , Female , Humans , Placenta/metabolism , Placenta Diseases/pathology , Pregnancy , Pregnant Women , SARS-CoV-2 , Spike Glycoprotein, Coronavirus/metabolism , Thromboplastin/metabolism
13.
Microbiology Spectrum ; 10(1):16, 2022.
Article in English | Web of Science | ID: covidwho-1790428

ABSTRACT

The vascular endothelial injury occurs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, but the mechanisms are poorly understood. We sought to determine the frequency and type of cytokine elevations and their relationship to endothelial injury induced by plasma from patients with SARS-CoV-2 versus controls. Plasma from eight consecutively enrolled patients hospitalized with acute SARS-CoV-2 infection was compared to controls. Endothelial cell (EC) barrier integrity was evaluated using ECIS (electric cell-substrate impedance sensing) on human lung microvascular EC. Plasma from all SARS-CoV-2 but none from controls decreased transendothelial resistance to a greater degree than that produced by tumor necrosis factor-alpha (TNF-alpha), the positive control for the assay. Thrombin, angiopoietin 2 (Ang2), and vascular endothelial growth factor (VEGF), complement factor C3a and C5a, and spike protein increased endothelial permeability, but to a lesser extent and a shorter duration when compared to SARS-CoV-2 plasma. Analysis of Ang2, VEGF, and 15 cytokines measured in plasma revealed striking patient-to-patient variability within the SARS-CoV-2 patients. Pretreatment with thrombin inhibitors, single, or combinations of neutralizing antibodies against cytokines, Ca3 and C5a receptor antagonists, or with ACE2 antibody failed to lessen the SARS-CoV-2 plasma-induced EC permeability. The EC barrier destructive effects of plasma from patients with SARS-CoV-2 were susceptible to heat inactivation. Plasma from patients hospitalized with acute SARS-CoV-2 infection uniformly disrupts lung microvascular integrity. No predicted single, or set of, cytokine(s) accounted for the enhanced vascular permeability, although the factor(s) were heat-labile. A still unidentified but potent circulating factor(s) appears to cause the EC disruption in SARS-CoV-2 infected patients. IMPORTANCE Lung vascular endothelial injury in SARS-CoV-2 patients is one of the most important causes of morbidity and mortality and has been linked to more severe complications including acute respiratory distress syndrome (ARDS) and subsequent death due to multiorgan failure. We have demonstrated that in eight consecutive patients with SARS-CoV-2, who were not selected for evidence of endothelial injury, the diluted plasma-induced intense lung microvascular damage, in vitro. Known endothelial barrier-disruptive agents and proposed mediators of increased endothelial permeability in SARS-CoV-2, induced changes in permeability that were smaller in magnitude and shorter in duration than plasma from patients with SARSCoV-2. The effect on endothelial cell permeability of plasma from patients with SARS-CoV-2 was heat-labile. The main plasma factor that causes the increased endothelial permeability remains to be identified. Our study provides a possible approach for future studies to understand the underlying mechanisms leading to vascular injury in SARS-CoV-2 infections.

14.
J Virol ; 96(5): e0208621, 2022 03 09.
Article in English | MEDLINE | ID: covidwho-1736026

ABSTRACT

Coronavirus infections induce the expression of multiple proinflammatory cytokines and chemokines. We have previously shown that in cells infected with gammacoronavirus infectious bronchitis virus (IBV), interleukin 6 (IL-6), and IL-8 were drastically upregulated, and the MAP kinase p38 and the integrated stress response pathways were implicated in this process. In this study, we report that coronavirus infection activates a negative regulatory loop that restricts the upregulation of a number of proinflammatory genes. As revealed by the initial transcriptomic and subsequent validation analyses, the anti-inflammatory adenine-uridine (AU)-rich element (ARE)-binding protein, zinc finger protein 36 (ZFP36), and its related family members were upregulated in cells infected with IBV and three other coronaviruses, alphacoronaviruses porcine epidemic diarrhea virus (PEDV), human coronavirus 229E (HCoV-229E), and betacoronavirus HCoV-OC43, respectively. Characterization of the functional roles of ZFP36 during IBV infection demonstrated that ZFP36 promoted the degradation of transcripts coding for IL-6, IL-8, dual-specificity phosphatase 1 (DUSP1), prostaglandin-endoperoxide synthase 2 (PTGS2) and TNF-α-induced protein 3 (TNFAIP3), through binding to AREs in these transcripts. Consistently, knockdown and inhibition of JNK and p38 kinase activities reduced the expression of ZFP36, as well as the expression of IL-6 and IL-8. On the contrary, overexpression of mitogen-activated protein kinase kinase 3 (MKK3) and MAPKAP kinase-2 (MK2), the upstream and downstream kinases of p38, respectively, increased the expression of ZFP36 and decreased the expression of IL-8. Taken together, this study reveals an important regulatory role of the MKK3-p38-MK2-ZFP36 axis in coronavirus infection-induced proinflammatory response. IMPORTANCE Excessive and uncontrolled induction and release of proinflammatory cytokines and chemokines, the so-called cytokine release syndrome (CRS), would cause life-threatening complications and multiple organ failure in severe coronavirus infections, including severe acute respiratory syndrome (SARS), Middle East respiratory syndrome (MERS) and COVID-19. This study reveals that coronavirus infection also induces the expression of ZFP36, an anti-inflammatory ARE-binding protein, promoting the degradation of ARE-containing transcripts coding for IL-6 and IL-8 as well as a number of other proteins related to inflammatory response. Furthermore, the p38 MAP kinase, its upstream kinase MKK3 and downstream kinase MK2 were shown to play a regulatory role in upregulation of ZFP36 during coronavirus infection cycles. This MKK3-p38-MK2-ZFP36 axis would constitute a potential therapeutic target for severe coronavirus infections.


Subject(s)
Coronavirus Infections/metabolism , Interleukin-6/metabolism , Interleukin-8/metabolism , Tristetraprolin/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Adenine/metabolism , Animals , Cell Line , Chlorocebus aethiops , Coronavirus Infections/genetics , Gene Expression Regulation , Humans , Infectious bronchitis virus/metabolism , Infectious bronchitis virus/pathogenicity , Interleukin-6/genetics , Interleukin-8/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Transcriptional Activation , Up-Regulation , Uridine/metabolism , Vero Cells
15.
J Genet Genomics ; 48(9): 792-802, 2021 09 20.
Article in English | MEDLINE | ID: covidwho-1720311

ABSTRACT

Gut microbial dysbiosis has been linked to many noncommunicable diseases. However, little is known about specific gut microbiota composition and its correlated metabolites associated with molecular signatures underlying host response to infection. Here, we describe the construction of a proteomic risk score based on 20 blood proteomic biomarkers, which have recently been identified as molecular signatures predicting the progression of the COVID-19. We demonstrate that in our cohort of 990 healthy individuals without infection, this proteomic risk score is positively associated with proinflammatory cytokines mainly among older, but not younger, individuals. We further discover that a core set of gut microbiota can accurately predict the above proteomic biomarkers among 301 individuals using a machine learning model and that these gut microbiota features are highly correlated with proinflammatory cytokines in another independent set of 366 individuals. Fecal metabolomics analysis suggests potential amino acid-related pathways linking gut microbiota to host metabolism and inflammation. Overall, our multi-omics analyses suggest that gut microbiota composition and function are closely related to inflammation and molecular signatures of host response to infection among healthy individuals. These results may provide novel insights into the cross-talk between gut microbiota and host immune system.


Subject(s)
Gastrointestinal Microbiome/physiology , Inflammation/metabolism , COVID-19/microbiology , Dysbiosis/microbiology , Gastrointestinal Microbiome/genetics , Humans , Inflammation/genetics , Proteomics/methods
16.
Microbiol Spectr ; 10(1): e0167121, 2022 02 23.
Article in English | MEDLINE | ID: covidwho-1691404

ABSTRACT

The vascular endothelial injury occurs in severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infections, but the mechanisms are poorly understood. We sought to determine the frequency and type of cytokine elevations and their relationship to endothelial injury induced by plasma from patients with SARS-CoV-2 versus controls. Plasma from eight consecutively enrolled patients hospitalized with acute SARS-CoV-2 infection was compared to controls. Endothelial cell (EC) barrier integrity was evaluated using ECIS (electric cell-substrate impedance sensing) on human lung microvascular EC. Plasma from all SARS-CoV-2 but none from controls decreased transendothelial resistance to a greater degree than that produced by tumor necrosis factor-alpha (TNF-α), the positive control for the assay. Thrombin, angiopoietin 2 (Ang2), and vascular endothelial growth factor (VEGF), complement factor C3a and C5a, and spike protein increased endothelial permeability, but to a lesser extent and a shorter duration when compared to SARS-CoV-2 plasma. Analysis of Ang2, VEGF, and 15 cytokines measured in plasma revealed striking patient-to-patient variability within the SARS-CoV-2 patients. Pretreatment with thrombin inhibitors, single, or combinations of neutralizing antibodies against cytokines, Ca3 and C5a receptor antagonists, or with ACE2 antibody failed to lessen the SARS-CoV-2 plasma-induced EC permeability. The EC barrier destructive effects of plasma from patients with SARS-CoV-2 were susceptible to heat inactivation. Plasma from patients hospitalized with acute SARS-CoV-2 infection uniformly disrupts lung microvascular integrity. No predicted single, or set of, cytokine(s) accounted for the enhanced vascular permeability, although the factor(s) were heat-labile. A still unidentified but potent circulating factor(s) appears to cause the EC disruption in SARS-CoV-2 infected patients. IMPORTANCE Lung vascular endothelial injury in SARS-CoV-2 patients is one of the most important causes of morbidity and mortality and has been linked to more severe complications including acute respiratory distress syndrome (ARDS) and subsequent death due to multiorgan failure. We have demonstrated that in eight consecutive patients with SARS-CoV-2, who were not selected for evidence of endothelial injury, the diluted plasma-induced intense lung microvascular damage, in vitro. Known endothelial barrier-disruptive agents and proposed mediators of increased endothelial permeability in SARS-CoV-2, induced changes in permeability that were smaller in magnitude and shorter in duration than plasma from patients with SARS-CoV-2. The effect on endothelial cell permeability of plasma from patients with SARS-CoV-2 was heat-labile. The main plasma factor that causes the increased endothelial permeability remains to be identified. Our study provides a possible approach for future studies to understand the underlying mechanisms leading to vascular injury in SARS-CoV-2 infections.


Subject(s)
COVID-19/blood , Capillary Permeability , Cytokines/blood , Lung/blood supply , SARS-CoV-2/physiology , Adult , Aged , COVID-19/physiopathology , COVID-19/virology , Endothelial Cells/virology , Female , Humans , Lung/virology , Male , Middle Aged , SARS-CoV-2/genetics , Tumor Necrosis Factor-alpha/blood , Vascular Endothelial Growth Factor A , Young Adult
17.
Front Immunol ; 12: 797390, 2021.
Article in English | MEDLINE | ID: covidwho-1686476

ABSTRACT

Phosphodiesterase 4 (PDE4) inhibitors are immunomodulatory drugs approved to treat diseases associated with chronic inflammatory conditions, such as COPD, psoriasis and atopic dermatitis. Tanimilast (international non-proprietary name of CHF6001) is a novel, potent and selective inhaled PDE4 inhibitor in advanced clinical development for the treatment of COPD. To begin testing its potential in limiting hyperinflammation and immune dysregulation associated to SARS-CoV-2 infection, we took advantage of an in vitro model of dendritic cell (DC) activation by SARS-CoV-2 genomic ssRNA (SCV2-RNA). In this context, Tanimilast decreased the release of pro-inflammatory cytokines (TNF-α and IL-6), chemokines (CCL3, CXCL9, and CXCL10) and of Th1-polarizing cytokines (IL-12, type I IFNs). In contrast to ß-methasone, a reference steroid anti-inflammatory drug, Tanimilast did not impair the acquisition of the maturation markers CD83, CD86 and MHC-II, nor that of the lymph node homing receptor CCR7. Consistent with this, Tanimilast did not reduce the capability of SCV2-RNA-stimulated DCs to activate CD4+ T cells but skewed their polarization towards a Th2 phenotype. Both Tanimilast and ß-methasone blocked the increase of MHC-I molecules in SCV2-RNA-activated DCs and restrained the proliferation and activation of cytotoxic CD8+ T cells. Our results indicate that Tanimilast can modulate the SCV2-RNA-induced pro-inflammatory and Th1-polarizing potential of DCs, crucial regulators of both the inflammatory and immune response. Given also the remarkable safety demonstrated by Tanimilast, up to now, in clinical studies, we propose this inhaled PDE4 inhibitor as a promising immunomodulatory drug in the scenario of COVID-19.


Subject(s)
COVID-19/immunology , Dendritic Cells , Phosphodiesterase 4 Inhibitors/pharmacology , RNA/pharmacology , SARS-CoV-2/physiology , Virus Activation/drug effects , CD8-Positive T-Lymphocytes/immunology , Cytokines/immunology , Dendritic Cells/immunology , Dendritic Cells/virology , Humans , Th1 Cells/immunology , Th2 Cells/immunology , Virus Activation/immunology , COVID-19 Drug Treatment
18.
Viral Immunol ; 34(10): 722-725, 2021 12.
Article in English | MEDLINE | ID: covidwho-1591011

ABSTRACT

Coronavirus disease 2019 (COVID-19) a global infectious disease caused by severe acute respiratory coronavirus 2 (SARS-CoV-2) affects various organs, primarily the respiratory system, and presented with pulmonary manifestations such as acute lung injury (ALI) and acute respiratory distress syndrome. Levamisole (LVM) is an anthelminthic drug; it has immune-modulating effects through induction of type 1 immune response. Based on these findings several recent studies highlighted that LVM might be effective in preventing and treating SARS-CoV-2 infections. The aim of this report is to illustrate the potential role of LVM in SARS-CoV-2 infection and in the management of COVID-19. Different studies proposed that LVM may inhibit proliferation of SARS-CoV-2 through inhibition of papain-like protease. LVM may prevent ALI and acute kidney injury through activation of glucocorticoid receptors. In general, LVM has strong immune stimulant effects by modulating cellular and humoral immune responses. This effect is beneficial in the early phase of COVID-19 and harmful in the late phase. In the early phase, immune stimulation facilitates SARS-CoV-2 clearance and tissue repair, however, in the late phase, immune stimulation in COVID-19 may increase propagation risk of cytokine storm. In conclusion, LVM therapy in COVID-19 has bidirectional effects, beneficial in the early phase and harmful effects in the late phase of COVID-19. Clinical trial and prospective studies are warranted in this regard to confirm the efficacy and timing administration of LVM in the management of COVID-19.


Subject(s)
COVID-19 Drug Treatment , COVID-19/immunology , Levamisole/administration & dosage , Levamisole/therapeutic use , Cytokine Release Syndrome/drug therapy , Cytokine Release Syndrome/immunology , Humans , Lung/immunology , Lung/pathology , SARS-CoV-2/immunology , SARS-CoV-2/pathogenicity
19.
Clin Microbiol Rev ; 34(3)2021 06 16.
Article in English | MEDLINE | ID: covidwho-1501524

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), a rapidly evolving pandemic worldwide with at least 68 million COVID-19-positive cases and a mortality rate of about 2.2%, as of 10 December 2020. About 20% of COVID-19 patients exhibit moderate to severe symptoms. Severe COVID-19 manifests as acute respiratory distress syndrome (ARDS) with elevated plasma proinflammatory cytokines, including interleukin 1ß (IL-1ß), IL-6, tumor necrosis factor α (TNF-α), C-X-C motif chemokine ligand 10 (CXCL10/IP10), macrophage inflammatory protein 1 alpha (MIP-1α), and chemokine (C-C motif) ligand 2 (CCL2), with low levels of interferon type I (IFN-I) in the early stage and elevated levels of IFN-I during the advanced stage of COVID-19. Most of the severe and critically ill COVID-19 patients have had preexisting comorbidities, including hypertension, diabetes, cardiovascular diseases, and respiratory diseases. These conditions are known to perturb the levels of cytokines, chemokines, and angiotensin-converting enzyme 2 (ACE2), an essential receptor involved in SARS-CoV-2 entry into the host cells. ACE2 downregulation during SARS-CoV-2 infection activates the angiotensin II/angiotensin receptor (AT1R)-mediated hypercytokinemia and hyperinflammatory syndrome. However, several SARS-CoV-2 proteins, including open reading frame 3b (ORF3b), ORF6, ORF7, ORF8, and the nucleocapsid (N) protein, can inhibit IFN type I and II (IFN-I and -II) production. Thus, hyperinflammation, in combination with the lack of IFN responses against SARS-CoV-2 early on during infection, makes the patients succumb rapidly to COVID-19. Therefore, therapeutic approaches involving anti-cytokine/anti-cytokine-signaling and IFN therapy would favor the disease prognosis in COVID-19. This review describes critical host and viral factors underpinning the inflammatory "cytokine storm" induction and IFN antagonism during COVID-19 pathogenesis. Therapeutic approaches to reduce hyperinflammation and their limitations are also discussed.


Subject(s)
COVID-19/pathology , Cytokine Release Syndrome/blood , Cytokine Release Syndrome/pathology , Interferon Type I/blood , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/metabolism , COVID-19/blood , COVID-19/therapy , Comorbidity , Humans , Immunity, Innate/immunology , Immunization, Passive/methods , Interleukin-6/antagonists & inhibitors , Interleukin-6/blood , Spike Glycoprotein, Coronavirus/metabolism , COVID-19 Serotherapy
20.
Cytokine ; 148: 155719, 2021 12.
Article in English | MEDLINE | ID: covidwho-1433128

ABSTRACT

Effective therapies for coronavirus disease 2019 (COVID-19) are urgently needed. Maladaptive hyperinflammation and excessive cytokine release underlie the disease severity, with antiinflammatory and cytokine inhibiting agents expected to exert therapeutic effects. A major present challenge is identification of appropriate phase of the illness for a given intervention to yield optimum outcomes. Considering its established disease biomarker and drug discovery potential, a compendious analysis of existing transcriptomic data is presented here toward addressing this gap. The analysis is based on COVID-19 data related to intensive care unit (ICU) and non-ICU admissions, discharged and deceased patients, ventilation and non-ventilation phases, and high oxygen supplementation. It integrates transcriptomic data related to the effects of, in various cellular treatment models, the COVID-19 randomized clinical trial (RCT) successful drug dexamethasone, and the failed drug, with a potential to harm, hydroxychloroquine/chloroquine. Similarly, effects of various COVID-19 candidate drugs/anticytokines as well as proinflammatory cytokines implicated in the illness are also examined. The underlying assumption was that compared to COVID-19, an effective drug/anticytokine and a disease aggravating agent would affect gene regulation in opposite and same direction, in that order. Remarkably, the assumption was supported with respect to both the RCT drugs. With this control validation, etanercept, followed by tofacitinib and adalimumab, showed transcriptomic effects predictive of benefits in both ventilation and non-ventilation ICU stages as well as in non-ICU phase. On the other hand, canakinumab showed potential for effectiveness in high oxygen supplementation phase. These findings may inform experimental and clinical studies toward drug repurposing in COVID-19.


Subject(s)
COVID-19 Drug Treatment , COVID-19/genetics , Gene Expression Profiling , COVID-19/physiopathology , Humans , Intensive Care Units , Oxygen/therapeutic use , Patient Discharge , Proof of Concept Study , Respiration, Artificial
SELECTION OF CITATIONS
SEARCH DETAIL