Your browser doesn't support javascript.
Show: 20 | 50 | 100
Results 1 - 20 de 39
Filter
1.
Front Immunol ; 14: 1152186, 2023.
Article in English | MEDLINE | ID: covidwho-20238642

ABSTRACT

Background Severe coronavirus disease 2019 (COVID -19) has led to severe pneumonia or acute respiratory distress syndrome (ARDS) worldwide. we have noted that many critically ill patients with COVID-19 present with typical sepsis-related clinical manifestations, including multiple organ dysfunction syndrome, coagulopathy, and septic shock. The molecular mechanisms that underlie COVID-19, ARDS and sepsis are not well understood. The objectives of this study were to analyze potential molecular mechanisms and identify potential drugs for the treatment of COVID-19, ARDS and sepsis using bioinformatics and a systems biology approach. Methods Three RNA-seq datasets (GSE171110, GSE76293 and GSE137342) from Gene Expression Omnibus (GEO) were employed to detect mutual differentially expressed genes (DEGs) for the patients with the COVID-19, ARDS and sepsis for functional enrichment, pathway analysis, and candidate drugs analysis. Results We obtained 110 common DEGs among COVID-19, ARDS and sepsis. ARG1, FCGR1A, MPO, and TLR5 are the most influential hub genes. The infection and immune-related pathways and functions are the main pathways and molecular functions of these three diseases. FOXC1, YY1, GATA2, FOXL, STAT1 and STAT3 are important TFs for COVID-19. mir-335-5p, miR-335-5p and hsa-mir-26a-5p were associated with COVID-19. Finally, the hub genes retrieved from the DSigDB database indicate multiple drug molecules and drug-targets interaction. Conclusion We performed a functional analysis under ontology terms and pathway analysis and found some common associations among COVID-19, ARDS and sepsis. Transcription factors-genes interaction, protein-drug interactions, and DEGs-miRNAs coregulatory network with common DEGs were also identified on the datasets. We believe that the candidate drugs obtained in this study may contribute to the effective treatment of COVID-19.


Subject(s)
COVID-19 , MicroRNAs , Respiratory Distress Syndrome , Sepsis , Humans , Gene Expression Profiling/methods , COVID-19/genetics , MicroRNAs/genetics , Computational Biology/methods , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/genetics , Sepsis/complications , Sepsis/drug therapy , Sepsis/genetics
2.
J Clin Lab Anal ; 37(5): e24846, 2023 Mar.
Article in English | MEDLINE | ID: covidwho-2273569

ABSTRACT

BACKGROUND: Angiotensin I converting enzyme 2 (ACE-2) is the most important receptor and has important role in the entry of corona virus to the host cells. The present study aimed to investigate the different mechanisms involved in the expression regulation of this gene among the COVID-19 patients. METHODS: A total of 140 patients with COVID-19 (n = 70 mild COVID-19, n = 70 ARDS) and 120 controls were recruited. The expression of ACE-2 and miRNAs was evaluated by quantitative real-time PCR (QRT-PCR), and methylation of CpG dinucleotides in the ACE2 promoter was quantified using bisulfite pyro-sequencing. Finally, different polymorphisms of the ACE-2 gene were studied by Sanger sequencing. RESULTS: Our results showed a significant high expression of the ACE-2 gene in the blood samples of acute respiratory distress syndrome (ARDS) patients (3.8 ± 0.77) in comparison with controls (0.88 ± 0.12; p < 0.03). The methylation rate of the ACE-2 gene in ARDS patients was 14.07 ± 6.1 compared with controls (72.3 ± 5.1; p < 0.0001). Among the four studied miRNAs, only miR200c-3p showed significant downregulation in ARDS patients (0.14 ± 0.1) in comparison with controls (0.32 ± 0.17; p < 0.001). We did not see a substantial difference in the frequency of rs182366225 C>T and rs2097723 T>C polymorphisms between patients and controls (p > 0.05). There was a significant correlation between B12 (R = 0.32, p < 0.001), folate (R = 0.37, p < 0.001) deficiency, and hypo-methylation of the ACE-2 gene. CONCLUSION: These results for the first time indicated that among the different mechanisms of ACE-2 expression regulation, its promoter methylation is very crucial and can be affected by factors involved in one-carbon metabolisms such as B9 and B12 vitamins deficiency.


Subject(s)
COVID-19 , MicroRNAs , Respiratory Distress Syndrome , Humans , Peptidyl-Dipeptidase A/genetics , COVID-19/genetics , Respiratory Distress Syndrome/genetics , Folic Acid , Severity of Illness Index
3.
Crit Care ; 27(1): 112, 2023 03 16.
Article in English | MEDLINE | ID: covidwho-2264356

ABSTRACT

BACKGROUND: The use of glucocorticoids has given contradictory results for treating acute respiratory distress syndrome (ARDS). The use of intravenous Interferon beta (IFN ß) for the treatment of ARDS was recently tested in a phase III ARDS trial (INTEREST), in which more than half of the patients simultaneously received glucocorticoids. Trial results showed deleterious effects of glucocorticoids when administered together with IFN ß, and therefore, we aimed at finding the reason behind this. METHODS: We first sequenced the genes encoding the IFN α/ß receptor of the patients, who participated in the INTEREST study (ClinicalTrials.gov Identifier:  NCT02622724 , November 24, 2015) in which the patients were randomized to receive an intravenous injection of IFN ß-1a (144 patients) or placebo (152 patients). Genetic background was analyzed against clinical outcome, concomitant medication, and pro-inflammatory cytokine levels. Thereafter, we tested the influence of the genetic background on IFN α/ß receptor expression in lung organ cultures and whether, it has any effect on transcription factors STAT1 and STAT2 involved in IFN signaling. RESULTS: We found a novel disease association of a SNP rs9984273, which is situated in the interferon α/ß receptor subunit 2 (IFNAR2) gene in an area corresponding to a binding motif of the glucocorticoid receptor (GR). The minor allele of SNP rs9984273 associates with higher IFNAR expression, more rapid decrease of IFN γ and interleukin-6 (IL-6) levels and better outcome in IFN ß treated patients with ARDS, while the major allele associates with a poor outcome especially under concomitant IFN ß and glucocorticoid treatment. Moreover, the minor allele of rs9984273 associates with a less severe form of coronavirus diseases (COVID-19) according to the COVID-19 Host Genetics Initiative database. CONCLUSIONS: The distribution of this SNP within clinical study arms may explain the contradictory results of multiple ARDS studies and outcomes in COVID-19 concerning type I IFN signaling and glucocorticoids.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Humans , Glucocorticoids/pharmacology , Glucocorticoids/therapeutic use , COVID-19/genetics , Interferon-beta/pharmacology , Interferon-beta/therapeutic use , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/genetics , Interferon-alpha
4.
Arch Razi Inst ; 77(6): 2291-2298, 2022 12.
Article in English | MEDLINE | ID: covidwho-2226513

ABSTRACT

Elevated Interleukin-13 (IL-13) may play an important role in the pathophysiology of COVID-19, yet, the attenuated response did not notice across all severe cases. Susceptibility to asthma in specific populations is associated with several SNPs of multifunctional cytokines, such as IL-13, IL-31 and IL-33. This prospective case-control study is designed to investigate the extent of genetic susceptibility in subsets of Iraqi patients with COVID-19 by targeting the variants of interleukin IL-13rs20541 polymorphism in relation to disease susceptibility and severity of clinical presentation. One hundred samples were obtained from the throat, nasopharyngeal and nasal swabs enrolled in this study. Eighty samples of the throat, nasopharyngeal and nasal localization swabs were obtained from patients with acute respiratory distress syndrome (ARDS) (both COVID-19 and non-COVID19 patients), while other 20 nasopharyngeal swabs were included as a healthy control group (AHC). Detection of IL-13rs20541 polymorphism was done by ARMS technique. The frequencies of GG- genotype in ARDS- patients with COVID-19, non-COVID19-, and AHC groups were respectively 14%, 12% and 3%, where, and as compared to the control group, showed a significant increase in COVID-19 patients. The AA- genotype in patients with COVID-19 group, non- COVID-19 group and healthy control group documented the frequency of 9%, 7%, and 14%, respectively, where the frequency decreased in the patient's groups as compared to the AHC group. Finally, and among the studied groups, an increase of AG- genotype (as rate OR=1.89) was documented compared to genotype GG and A-. Genetic polymorphisms in the IL-13rs20541 gene might influence its functions in patients with SARS-associated respiratory tract infection and thus might involve the pathogenicity of patients with COVID-19.


Subject(s)
COVID-19 , Interleukin-13 , Respiratory Distress Syndrome , Humans , Case-Control Studies , COVID-19/genetics , Gene Frequency , Genomics , Interleukin-13/genetics , Polymorphism, Single Nucleotide , Respiratory Distress Syndrome/genetics
6.
Respir Res ; 23(1): 318, 2022 Nov 19.
Article in English | MEDLINE | ID: covidwho-2157439

ABSTRACT

In the last decade, research on acute respiratory distress syndrome (ARDS) has made considerable progress. However, ARDS remains a leading cause of mortality in the intensive care unit. ARDS presents distinct subphenotypes with different clinical and biological features. The pathophysiologic mechanisms of ARDS may contribute to the biological variability and partially explain why some pharmacologic therapies for ARDS have failed to improve patient outcomes. Therefore, identifying ARDS variability and heterogeneity might be a key strategy for finding effective treatments. Research involving studies on biomarkers and genomic, metabolomic, and proteomic technologies is increasing. These new approaches, which are dedicated to the identification and quantitative analysis of components from biological matrixes, may help differentiate between different types of damage and predict clinical outcome and risk. Omics technologies offer a new opportunity for the development of diagnostic tools and personalized therapy in ARDS. This narrative review assesses recent evidence regarding genomics, proteomics, and metabolomics in ARDS research.


Subject(s)
Precision Medicine , Respiratory Distress Syndrome , Humans , Proteomics , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/genetics , Phenotype , Biomarkers
7.
Biochem Pharmacol ; 208: 115382, 2023 02.
Article in English | MEDLINE | ID: covidwho-2158476

ABSTRACT

Acute respiratory distress syndrome (ARDS) is characterized by noncardiogenic pulmonary edema. It has a high mortality rate and lacks effective pharmacotherapy. With the outbreak of COVID-19 worldwide, the mortality of ARDS has increased correspondingly, which makes it urgent to find effective targets and strategies for the treatment of ARDS. Recent clinical trials of Janus kinase (JAK) inhibitors in treating COVID-19-induced ARDS have shown a positive outcome, which makes the Janus kinase/signal transducer and activator of transcription (JAK/STAT) pathway a potential therapeutic target for treating ARDS. Here, we review the complex cause of ARDS, the molecular JAK/STAT pathway involved in ARDS pathology, and the progress that has been made in strategies targeting JAK/STAT to treat ARDS. Specifically, JAK/STAT signaling directly participates in the progression of ARDS or colludes with other pathways to aggravate ARDS. We summarize JAK and STAT inhibitors with ARDS treatment benefits, including inhibitors in clinical trials and preclinical studies and natural products, and discuss the side effects of the current JAK inhibitors to reveal future trends in the design of JAK inhibitors, which will help to develop effective treatment strategies for ARDS in the future.


Subject(s)
COVID-19 , Janus Kinases , Respiratory Distress Syndrome , STAT Transcription Factors , Humans , COVID-19/genetics , Janus Kinase Inhibitors/pharmacology , Janus Kinases/genetics , Janus Kinases/metabolism , Respiratory Distress Syndrome/drug therapy , Respiratory Distress Syndrome/genetics , Signal Transduction , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism
8.
J Transl Med ; 20(1): 526, 2022 Nov 12.
Article in English | MEDLINE | ID: covidwho-2115628

ABSTRACT

BACKGROUND: COVID-19 infections could be complicated by acute respiratory distress syndrome (ARDS), increasing mortality risk. We sought to assess the methylome of peripheral blood mononuclear cells in COVID-19 with ARDS. METHODS: We recruited 100 COVID-19 patients with ARDS under mechanical ventilation and 33 non-COVID-19 controls between April and July 2020. COVID-19 patients were followed at four time points for 60 days. DNA methylation and immune cell populations were measured at each time point. A multivariate cox proportional risk regression analysis was conducted to identify predictive signatures according to survival. RESULTS: The comparison of COVID-19 to controls at inclusion revealed the presence of a 14.4% difference in promoter-associated CpGs in genes that control immune-related pathways such as interferon-gamma and interferon-alpha responses. On day 60, 24% of patients died. The inter-comparison of baseline DNA methylation to the last recorded time point in both COVID-19 groups or the intra-comparison between inclusion and the end of follow-up in every group showed that most changes occurred as the disease progressed, mainly in the AIM gene, which is associated with an intensified immune response in those who recovered. The multivariate Cox proportional risk regression analysis showed that higher methylation of the "Apoptotic execution Pathway" genes (ROC1, ZNF789, and H1F0) at inclusion increases mortality risk by over twofold. CONCLUSION: We observed an epigenetic signature of immune-related genes in COVID-19 patients with ARDS. Further, Hypermethylation of the apoptotic execution pathway genes predicts the outcome. TRIAL REGISTRATION: IMRPOVIE study, NCT04473131.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Humans , COVID-19/complications , COVID-19/genetics , DNA Methylation/genetics , Leukocytes, Mononuclear , Respiration, Artificial , Respiratory Distress Syndrome/complications , Respiratory Distress Syndrome/genetics , SARS-CoV-2
9.
J Clin Lab Anal ; 36(11): e24672, 2022 Nov.
Article in English | MEDLINE | ID: covidwho-2047648

ABSTRACT

BACKGROUND: The pandemic COVID-19 has caused a high mortality rate and poses a significant threat to the population of the entire world. Due to the novelty of this disease, the pathogenic mechanism of the disease and the host cell's response are not yet fully known, so lack of evidence prevents a definitive conclusion about treatment strategies. The current study employed a small RNA deep-sequencing approach for screening differentially expressed microRNA (miRNA) in blood and bronchoalveolar fluid (BALF) samples of acute respiratory distress syndrome (ARDS) patients. METHODS: In this study, BALF and blood samples were taken from patients with ARDS (n = 5). Control samples were those with suspected lung cancer candidates for lung biopsy (n = 3). Illumina high-throughput (HiSeq 2000) sequencing was performed to identify known and novel miRNAs differentially expressed in the blood and BALFs of ARDS patients compared with controls. RESULTS: Results showed 2234 and 8324 miRNAs were differentially expressed in blood and BALF samples, respectively. In BALF samples, miR-282, miR-15-5p, miR-4485-3p, miR-483-3p, miR-6891-5p, miR-200c, miR-4463, miR-483-5p, and miR-98-5p were upregulated and miR-15a-5p, miR-548c-5p, miR-548d-3p, miR-365a-3p, miR-3939, miR-514-b-5p, miR-513a-3p, miR-513a-5p, miR-664a-3p, and miR-766-3p were downregulated. On the contrary, in blood samples miR-15b-5p, miR-18a-3p, miR-486-3p, miR-486-5p, miR-146a-5p, miR-16-2-3p, miR-6501-5p, miR-365-3p, miR-618, and miR-623 were top upregulated miRNAs and miR-21-5p, miR-142a-3p, miR-181-a, miR-31-5p, miR-99-5p, miR-342-5p, miR-183-5p, miR-627-5p, and miR-144-3p were downregulated miRNAs. Network functional analysis for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG), in ARDS patients' blood and BALF samples, showed that the target genes were more involved in activating inflammatory and apoptosis process. CONCLUSION: Based on our results, the transcriptome profile of ARDS patients would be a valuable source for understanding molecular mechanisms of host response and developing clinical guidance on anti-inflammatory medication.


Subject(s)
COVID-19 , MicroRNAs , Respiratory Distress Syndrome , Humans , COVID-19/genetics , Gene Expression Profiling , High-Throughput Nucleotide Sequencing/methods , MicroRNAs/genetics , Respiratory Distress Syndrome/genetics , Sequence Analysis, RNA/methods
10.
Biomed Pharmacother ; 154: 113617, 2022 Oct.
Article in English | MEDLINE | ID: covidwho-2003885

ABSTRACT

BACKGROUND: Up to 80% of patients surviving acute respiratory distress syndrome (ARDS) secondary to SARS-CoV-2 infection present persistent anomalies in pulmonary function after hospital discharge. There is a limited understanding of the mechanistic pathways linked to post-acute pulmonary sequelae. AIM: To identify the molecular underpinnings associated with severe lung diffusion involvement in survivors of SARS-CoV-2-induced ARDS. METHODS: Survivors attended to a complete pulmonary evaluation 3 months after hospital discharge. RNA sequencing (RNA-seq) was performed using Illumina technology in whole-blood samples from 50 patients with moderate to severe diffusion impairment (DLCO<60%) and age- and sex-matched individuals with mild-normal lung function (DLCO≥60%). A transcriptomic signature for optimal classification was constructed using random forest. Transcriptomic data were analyzed for biological pathway enrichment, cellular deconvolution, cell/tissue-specific gene expression and candidate drugs. RESULTS: RNA-seq identified 1357 differentially expressed transcripts. A model composed of 14 mRNAs allowed the optimal discrimination of survivors with severe diffusion impairment (AUC=0.979). Hallmarks of lung sequelae involved cell death signaling, cytoskeleton reorganization, cell growth and differentiation and the immune response. Resting natural killer (NK) cells were the most important immune cell subtype for the prediction of severe diffusion impairment. Components of the signature correlated with neutrophil, lymphocyte and monocyte counts. A variable expression profile of the transcripts was observed in lung cell subtypes and bodily tissues. One upregulated gene, TUBB4A, constitutes a target for FDA-approved drugs. CONCLUSIONS: This work defines the transcriptional programme associated with post-acute pulmonary sequelae and provides novel insights for targeted interventions and biomarker development.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , COVID-19/complications , COVID-19/genetics , Humans , Lung , Respiratory Distress Syndrome/genetics , SARS-CoV-2 , Survivors , Tubulin
11.
BMC Infect Dis ; 21(1): 852, 2021 Aug 21.
Article in English | MEDLINE | ID: covidwho-1363547

ABSTRACT

BACKGROUND AND AIMS: Acute respiratory distress syndrome (ARDS) or acute lung injury (ALI) is one of the most common acute thoracopathy with complicated pathogenesis in ICU. The study is to explore the differentially expressed genes (DEGs) in the lung tissue and underlying altering mechanisms in ARDS. METHODS: Gene expression profiles of GSE2411 and GSE130936 were available from GEO database, both of them included in GPL339. Then, an integrated analysis of these genes was performed, including gene ontology (GO) and KEGG pathway enrichment analysis in DAVID database, protein-protein interaction (PPI) network construction evaluated by the online database STRING, Transcription Factors (TFs) forecasting based on the Cytoscape plugin iRegulon, and their expression in varied organs in The Human Protein Atlas. RESULTS: A total of 39 differential expressed genes were screened from the two datasets, including 39 up-regulated genes and 0 down-regulated genes. The up-regulated genes were mainly enriched in the biological process, such as immune system process, innate immune response, inflammatory response, and also involved in some signal pathways, including cytokine-cytokine receptor interaction, Salmonella infection, Legionellosis, Chemokine, and Toll-like receptor signal pathway with an integrated analysis. GBP2, IFIT2 and IFIT3 were identified as hub genes in the lung by PPI network analysis with MCODE plug-in, as well as GO and KEGG re-enrichment. All of the three hub genes were regulated by the predictive common TFs, including STAT1, E2F1, IRF1, IRF2, and IRF9. CONCLUSIONS: This study implied that hub gene GBP2, IFIT2 and IFIT3, which might be regulated by STAT1, E2F1, IRF1, IRF2, or IRF9, played significant roles in ARDS. They could be potential diagnostic or therapeutic targets for ARDS patients.


Subject(s)
Lipopolysaccharides , Respiratory Distress Syndrome , Computational Biology , Gene Expression Profiling , Humans , Protein Interaction Maps , Respiratory Distress Syndrome/genetics
12.
Am J Physiol Lung Cell Mol Physiol ; 323(1): L84-L92, 2022 07 01.
Article in English | MEDLINE | ID: covidwho-1891936

ABSTRACT

Increased plasma mitochondrial DNA concentrations are associated with poor outcomes in multiple critical illnesses, including COVID-19. However, current methods of cell-free mitochondrial DNA quantification in plasma are time-consuming and lack reproducibility. Here, we used next-generation sequencing to characterize the size and genome location of circulating mitochondrial DNA in critically ill subjects with COVID-19 to develop a facile and optimal method of quantification by droplet digital PCR. Sequencing revealed a large percentage of small mitochondrial DNA fragments in plasma with wide variability in coverage by genome location. We identified probes for the mitochondrial DNA genes, cytochrome B and NADH dehydrogenase 1, in regions of relatively high coverage that target small sequences potentially missed by other methods. Serial assessments of absolute mitochondrial DNA concentrations were then determined in plasma from 20 critically ill subjects with COVID-19 without a DNA isolation step. Mitochondrial DNA concentrations on the day of enrollment were increased significantly in patients with moderate or severe acute respiratory distress syndrome (ARDS) compared with those with no or mild ARDS. Comparisons of mitochondrial DNA concentrations over time between patients with no/mild ARDS who survived, patients with moderate/severe ARDS who survived, and nonsurvivors showed the highest concentrations in patients with more severe disease. Absolute mitochondrial DNA quantification by droplet digital PCR is time-efficient and reproducible; thus, we provide a valuable tool and rationale for future studies evaluating mitochondrial DNA as a real-time biomarker to guide clinical decision-making in critically ill subjects with COVID-19.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , COVID-19/diagnosis , COVID-19/genetics , Critical Illness , DNA, Mitochondrial/genetics , Humans , Intensive Care Units , Polymerase Chain Reaction , Reproducibility of Results , Respiratory Distress Syndrome/diagnosis , Respiratory Distress Syndrome/genetics
13.
Int J Mol Sci ; 23(10)2022 May 16.
Article in English | MEDLINE | ID: covidwho-1875642

ABSTRACT

Acute lung injury (ALI)/acute respiratory distress syndrome (ARDS) is an overactivated inflammatory response caused by direct or indirect injuries that destroy lung parenchymal cells and dramatically reduce lung function. Although some research progress has been made in recent years, the pathogenesis of ALI/ARDS remains unclear due to its heterogeneity and etiology. MicroRNAs (miRNAs), a type of small noncoding RNA, play a vital role in various diseases. In ALI/ARDS, miRNAs can regulate inflammatory and immune responses by targeting specific molecules. Regulation of miRNA expression can reduce damage and promote the recovery of ALI/ARDS. Consequently, miRNAs are considered as potential diagnostic indicators and therapeutic targets of ALI/ARDS. Given that inflammation plays an important role in the pathogenesis of ALI/ARDS, we review the miRNAs involved in the inflammatory process of ALI/ARDS to provide new ideas for the pathogenesis, clinical diagnosis, and treatment of ALI/ARDS.


Subject(s)
Acute Lung Injury , MicroRNAs , Respiratory Distress Syndrome , Acute Lung Injury/metabolism , Humans , Inflammation/genetics , Lung/metabolism , MicroRNAs/genetics , Respiratory Distress Syndrome/genetics
14.
Sci Rep ; 12(1): 6929, 2022 04 28.
Article in English | MEDLINE | ID: covidwho-1815599

ABSTRACT

One of the hallmarks of SARS-CoV-2 infection is an induced immune dysregulation, in some cases resulting in cytokine storm syndrome and acute respiratory distress syndrome (ARDS). Several physiological parameters are altered as a result of infection and cytokine storm. Among them, microRNAs (miRNAs) might reflect this poor condition since they play a significant role in immune cellular performance including inflammatory responses. Circulating miRNAs in patients who underwent ARDS and needed mechanical ventilation (MV+; n = 15) were analyzed by next generation sequencing in comparison with patients who had COVID-19 poor symptoms but without intensive care unit requirement (MV-; n = 13). A comprehensive in silico analysis by integration with public gene expression dataset and pathway enrichment was performed. Whole miRNA sequencing identified 170 differentially expressed miRNAs between patient groups. After the validation step by qPCR in an independent sample set (MV+ = 10 vs. MV- = 10), the miR-369-3p was found significantly decreased in MV+ patients (Fold change - 2.7). After integrating with gene expression results from COVID-19 patients, the most significant GO enriched pathways were acute inflammatory response, regulation of transmembrane receptor protein Ser/Thr, fat cell differentiation, and regulation of biomineralization and ossification. In conclusion, miR-369-3p was altered in patients with mechanical ventilation requirement in comparison with COVID-19 patients without this requirement. This miRNA is involved in inflammatory response which it can be considered as a prognosis factor for ARDS in COVID-19 patients.


Subject(s)
COVID-19 , Circulating MicroRNA , MicroRNAs , Respiratory Distress Syndrome , COVID-19/complications , COVID-19/genetics , Circulating MicroRNA/genetics , Cytokine Release Syndrome , Humans , MicroRNAs/genetics , Respiratory Distress Syndrome/genetics , SARS-CoV-2
15.
Cell Death Dis ; 13(3): 269, 2022 03 25.
Article in English | MEDLINE | ID: covidwho-1764162

ABSTRACT

Stimulator of interferon genes (STING) contributes to immune responses against tumors and may control viral infection including SARS-CoV-2 infection. However, activation of the STING pathway by airway silica or smoke exposure leads to cell death, self-dsDNA release, and STING/type I IFN dependent acute lung inflammation/ARDS. The inflammatory response induced by a synthetic non-nucleotide-based diABZI STING agonist, in comparison to the natural cyclic dinucleotide cGAMP, is unknown. A low dose of diABZI (1 µg by endotracheal route for 3 consecutive days) triggered an acute neutrophilic inflammation, disruption of the respiratory barrier, DNA release with NET formation, PANoptosis cell death, and inflammatory cytokines with type I IFN dependent acute lung inflammation. Downstream upregulation of DNA sensors including cGAS, DDX41, IFI204, as well as NLRP3 and AIM2 inflammasomes, suggested a secondary inflammatory response to dsDNA as a danger signal. DNase I treatment, inhibition of NET formation together with an investigation in gene-deficient mice highlighted extracellular DNA and TLR9, but not cGAS, as central to diABZI-induced neutrophilic response. Therefore, activation of acute cell death with DNA release may lead to ARDS which may be modeled by diABZI. These results show that airway targeting by STING activator as a therapeutic strategy for infection may enhance lung inflammation with severe ARDS. STING agonist diABZI induces neutrophilic lung inflammation and PANoptosis A, Airway STING priming induce a neutrophilic lung inflammation with epithelial barrier damage, double-stranded DNA release in the bronchoalvelolar space, cell death, NETosis and type I interferon release. B, 1. The diamidobenzimidazole (diABZI), a STING agonist is internalized into the cytoplasm through unknown receptor and induce the activation and dimerization of STING followed by TBK1/IRF3 phosporylation leading to type I IFN response. STING activation also leads to NF-kB activation and the production of pro-inflammatory cytokines TNFα and IL-6. 2. The activation of TNFR1 and IFNAR1 signaling pathway results in ZBP1 and RIPK3/ASC/CASP8 activation leading to MLKL phosphorylation and necroptosis induction. 3. This can also leads to Caspase-3 cleavage and apoptosis induction. 4. Self-dsDNA or mtDNA sensing by NLRP3 or AIM2 induces inflammsome formation leading to Gasdermin D cleavage enabling Gasdermin D pore formation and the release mature IL-1ß and pyroptosis. NLRP3 inflammasome formation can be enhanced by the ZBP1/RIPK3/CASP8 complex. 5. A second signal of STING activation with diABZI induces cell death and the release of self-DNA which is sensed by cGAS and form 2'3'-cGAMP leading to STING hyper activation, the amplification of TBK1/IRF3 and NF-kB pathway and the subsequent production of IFN-I and inflammatory TNFα and IL-6. This also leads to IFI204 and DDX41 upregulation thus, amplifying the inflammatory loop. The upregulation of apoptosis, pyroptosis and necroptosis is indicative of STING-dependent PANoptosis.


Subject(s)
COVID-19 , Respiratory Distress Syndrome , Animals , Cytokines/metabolism , DNA , Inflammasomes/metabolism , Interleukin-6/metabolism , Mice , NF-kappa B/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Nucleotidyltransferases/genetics , Nucleotidyltransferases/metabolism , RNA-Binding Proteins , Respiratory Distress Syndrome/genetics , SARS-CoV-2 , Tumor Necrosis Factor-alpha/metabolism
16.
Int J Mol Sci ; 23(3)2022 Feb 08.
Article in English | MEDLINE | ID: covidwho-1674674

ABSTRACT

Preventing the cytokine storm observed in COVID-19 is a crucial goal for reducing the occurrence of severe acute respiratory failure and improving outcomes. Here, we identify Aldo-Keto Reductase 1B10 (AKR1B10) as a key enzyme involved in the expression of pro-inflammatory cytokines. The analysis of transcriptomic data from lung samples of patients who died from COVID-19 demonstrates an increased expression of the gene encoding AKR1B10. Measurements of the AKR1B10 protein in sera from hospitalised COVID-19 patients suggests a significant link between AKR1B10 levels and the severity of the disease. In macrophages and lung cells, the over-expression of AKR1B10 induces the expression of the pro-inflammatory cytokines Interleukin-6 (IL-6), Interleukin-1ß (IL-1ß) and Tumor Necrosis Factor a (TNFα), supporting the biological plausibility of an AKR1B10 involvement in the COVID-19-related cytokine storm. When macrophages were stressed by lipopolysaccharides (LPS) exposure and treated by Zopolrestat, an AKR1B10 inhibitor, the LPS-induced production of IL-6, IL-1ß, and TNFα is significantly reduced, reinforcing the hypothesis that the pro-inflammatory expression of cytokines is AKR1B10-dependant. Finally, we also show that AKR1B10 can be secreted and transferred via extracellular vesicles between different cell types, suggesting that this protein may also contribute to the multi-organ systemic impact of COVID-19. These experiments highlight a relationship between AKR1B10 production and severe forms of COVID-19. Our data indicate that AKR1B10 participates in the activation of cytokines production and suggest that modulation of AKR1B10 activity might be an actionable pharmacological target in COVID-19 management.


Subject(s)
Aldo-Keto Reductases/physiology , COVID-19/genetics , Cytokine Release Syndrome/genetics , Respiratory Distress Syndrome/genetics , Aldo-Keto Reductases/antagonists & inhibitors , Aldo-Keto Reductases/genetics , Animals , COVID-19/complications , COVID-19/metabolism , COVID-19/pathology , Case-Control Studies , Cells, Cultured , Cytokine Release Syndrome/metabolism , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/virology , Cytokines/metabolism , Enzyme Inhibitors/pharmacology , Humans , Macrophages/drug effects , Macrophages/metabolism , Mice , Patient Acuity , RAW 264.7 Cells , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , SARS-CoV-2/physiology , Transcriptome
17.
Cell Rep Med ; 3(2): 100522, 2022 02 15.
Article in English | MEDLINE | ID: covidwho-1650891

ABSTRACT

The molecular mechanisms underlying the clinical manifestations of coronavirus disease 2019 (COVID-19), and what distinguishes them from common seasonal influenza virus and other lung injury states such as acute respiratory distress syndrome, remain poorly understood. To address these challenges, we combine transcriptional profiling of 646 clinical nasopharyngeal swabs and 39 patient autopsy tissues to define body-wide transcriptome changes in response to COVID-19. We then match these data with spatial protein and expression profiling across 357 tissue sections from 16 representative patient lung samples and identify tissue-compartment-specific damage wrought by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, evident as a function of varying viral loads during the clinical course of infection and tissue-type-specific expression states. Overall, our findings reveal a systemic disruption of canonical cellular and transcriptional pathways across all tissues, which can inform subsequent studies to combat the mortality of COVID-19 and to better understand the molecular dynamics of lethal SARS-CoV-2 and other respiratory infections.


Subject(s)
COVID-19/genetics , COVID-19/pathology , Lung/pathology , SARS-CoV-2 , Transcriptome/genetics , Adult , Aged , Aged, 80 and over , COVID-19/metabolism , COVID-19/virology , Case-Control Studies , Cohort Studies , Female , Gene Expression Regulation , Humans , Influenza, Human/genetics , Influenza, Human/pathology , Influenza, Human/virology , Lung/metabolism , Male , Middle Aged , Orthomyxoviridae , RNA-Seq/methods , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/microbiology , Respiratory Distress Syndrome/pathology , Viral Load
18.
Int J Mol Sci ; 23(3)2022 Jan 24.
Article in English | MEDLINE | ID: covidwho-1650418

ABSTRACT

Acute Respiratory Distress Syndrome is the most common cause of respiratory failure among critically ill patients, and its importance has been heightened during the COVID-19 pandemic. Even with the best supportive care, the mortality rate in the most severe cases is 40-50%, and the only pharmacological agent shown to be of possible benefit has been steroids. Mesenchymal stromal cells (MSCs) have been tested in several pre-clinical models of lung injury and been found to have significant therapeutic benefit related to: (a) potent immunomodulation; (b) secretion of epithelial and endothelial growth factors; and (c) augmentation of host defense to infection. Initial translational efforts have shown signs of promise, but the results have not yielded the anticipated outcomes. One potential reason is the relatively low survival of MSCs in inflammatory conditions as shown in several studies. Therefore, strategies to boost the survival of MSCs are needed to enhance their therapeutic effect. Protease-activated receptors (PARs) may represent one such possibility as they are G-protein coupled receptors expressed by MSCs and control several facets of cell behavior. This review summarizes some of the existing literature about PARs and MSCs and presents possible future areas of investigation in order to develop potential, PAR-modified MSCs with enhanced therapeutic efficiency.


Subject(s)
Graft Survival/genetics , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/metabolism , Receptors, Proteinase-Activated/physiology , Respiratory Distress Syndrome/therapy , Animals , COVID-19/genetics , COVID-19/pathology , COVID-19/therapy , Cell Survival/genetics , Critical Illness/therapy , Humans , Mesenchymal Stem Cells/physiology , Receptors, Proteinase-Activated/genetics , Receptors, Proteinase-Activated/metabolism , Respiratory Distress Syndrome/genetics , Respiratory Distress Syndrome/metabolism , Respiratory Distress Syndrome/virology , SARS-CoV-2/physiology , Signal Transduction/physiology , Transfection , Treatment Outcome
19.
EBioMedicine ; 74: 103695, 2021 Dec.
Article in English | MEDLINE | ID: covidwho-1596202

ABSTRACT

BACKGROUND: The heterogeneity in symptomatology and phenotypic profile attributable to COVID-19 is widely unknown. The objective of this manuscript is to conduct a trans-ancestry genome wide association study (GWAS) meta-analysis of COVID-19 severity to improve the understanding of potentially causal targets for SARS-CoV-2. METHODS: This cross-sectional study recruited 646 participants in the UAE that were divided into two phenotypic groups based on the severity of COVID-19 phenotypes, hospitalized (n=482) and non-hospitalized (n=164) participants. Hospitalized participants were COVID-19 patients that developed acute respiratory distress syndrome (ARDS), pneumonia or progression to respiratory failure that required supplemental oxygen therapy or mechanical ventilation support or had severe complications such as septic shock or multi-organ failure. We conducted a trans-ancestry meta-analysis GWAS of European (n=302), American (n=102), South Asian (n=99), and East Asian (n=107) ancestry populations. We also carried out comprehensive post-GWAS analysis, including enrichment of SNP associations in tissues and cell-types, expression quantitative trait loci and differential expression analysis. FINDINGS: Eight genes demonstrated a strong association signal: VWA8 gene in locus 13p14·11 (SNP rs10507497; p=9·54 x10-7), PDE8B gene in locus 5q13·3 (SNP rs7715119; p=2·19 x10-6), CTSC gene in locus 11q14·2 (rs72953026; p=2·38 x10-6), THSD7B gene in locus 2q22·1 (rs7605851; p=3·07x10-6), STK39 gene in locus 2q24·3 (rs7595310; p=4·55 x10-6), FBXO34 gene in locus 14q22·3 (rs10140801; p=8·26 x10-6), RPL6P27 gene in locus 18p11·31 (rs11659676; p=8·88 x10-6), and METTL21C gene in locus 13q33·1 (rs599976; p=8·95 x10-6). The genes are expressed in the lung, associated to tumour progression, emphysema, airway obstruction, and surface tension within the lung, as well as an association to T-cell-mediated inflammation and the production of inflammatory cytokines. INTERPRETATION: We have discovered eight highly plausible genetic association with hospitalized cases in COVID-19. Further studies must be conducted on worldwide population genetics to facilitate the development of population specific therapeutics to mitigate this worldwide challenge. FUNDING: This review was commissioned as part of a project to study the host cell receptors of coronaviruses funded by Khalifa University's CPRA grant (Reference number 2020-004).


Subject(s)
Genetic Predisposition to Disease/genetics , Quantitative Trait Loci/genetics , Quantitative Trait, Heritable , Respiratory Distress Syndrome/genetics , Severity of Illness Index , Adolescent , Adult , Aged , COVID-19/mortality , COVID-19/pathology , Cross-Sectional Studies , Female , Genome-Wide Association Study , Hospitalization/statistics & numerical data , Humans , Inflammation/genetics , Lung/pathology , Male , Middle Aged , Polymorphism, Single Nucleotide/genetics , Population Groups/genetics , Respiratory Distress Syndrome/pathology , SARS-CoV-2 , T-Lymphocytes/immunology , Treatment Outcome , United Arab Emirates , Young Adult
20.
Signal Transduct Target Ther ; 6(1): 439, 2021 12 24.
Article in English | MEDLINE | ID: covidwho-1585883

ABSTRACT

The development of animal models for COVID-19 is essential for basic research and drug/vaccine screening. Previously reported COVID-19 animal models need to be established under a high biosafety level condition for the utilization of live SARS-CoV-2, which greatly limits its application in routine research. Here, we generate a mouse model of COVID-19 under a general laboratory condition that captures multiple characteristics of SARS-CoV-2-induced acute respiratory distress syndrome (ARDS) observed in humans. Briefly, human ACE2-transgenic (hACE2) mice were intratracheally instilled with the formaldehyde-inactivated SARS-CoV-2, resulting in a rapid weight loss and detrimental changes in lung structure and function. The pulmonary pathologic changes were characterized by diffuse alveolar damage with pulmonary consolidation, hemorrhage, necrotic debris, and hyaline membrane formation. The production of fatal cytokines (IL-1ß, TNF-α, and IL-6) and the infiltration of activated neutrophils, inflammatory monocyte-macrophages, and T cells in the lung were also determined, suggesting the activation of an adaptive immune response. Therapeutic strategies, such as dexamethasone or passive antibody therapy, could effectively ameliorate the disease progression in this model. Therefore, the established mouse model for SARS-CoV-2-induced ARDS in the current study may provide a robust tool for researchers in the standard open laboratory to investigate the pathological mechanisms or develop new therapeutic strategies for COVID-19 and ARDS.


Subject(s)
Angiotensin-Converting Enzyme 2/immunology , COVID-19/immunology , Lung/immunology , Respiratory Distress Syndrome/immunology , SARS-CoV-2/immunology , Angiotensin-Converting Enzyme 2/genetics , Animals , COVID-19/genetics , Disease Models, Animal , Humans , Mice , Mice, Transgenic , Respiratory Distress Syndrome/genetics
SELECTION OF CITATIONS
SEARCH DETAIL