Skip to main content
Erschienen in: BMC Pulmonary Medicine 1/2024

Open Access 01.12.2024 | Research

Identification of biomarkers and pathways for the SARS-CoV-2 infections in obstructive sleep apnea patients based on machine learning and proteomic analysis

verfasst von: Hong Luo, Jisong Yan, Rui Gong, Dingyu Zhang, Xia Zhou, Xianguang Wang

Erschienen in: BMC Pulmonary Medicine | Ausgabe 1/2024

Abstract

Background

The prevalence of obstructive sleep apnea (OSA) was found to be higher in individuals following COVID-19 infection. However, the intricate mechanisms that underscore this concomitance remain partially elucidated. The aim of this study was to delve deeper into the molecular mechanisms that underpin this comorbidity.

Methods

We acquired gene expression profiles for COVID-19 (GSE157103) and OSA (GSE75097) from the Gene Expression Omnibus (GEO) database. Upon identifying shared feature genes between OSA and COVID-19 utilizing LASSO, Random forest and Support vector machines algorithms, we advanced to functional annotation, analysis of protein–protein interaction networks, module construction, and identification of pivotal genes. Furthermore, we established regulatory networks encompassing transcription factor (TF)-gene and TF-miRNA interactions, and searched for promising drug targets. Subsequently, the expression levels of pivotal genes were validated through proteomics data from COVID-19 cases.

Results

Fourteen feature genes shared between OSA and COVID-19 were selected for further investigation. Through functional annotation, it was indicated that metabolic pathways play a role in the pathogenesis of both disorders. Subsequently, employing the cytoHubba plugin, ten hub genes were recognized, namely TP53, CCND1, MDM2, RB1, HIF1A, EP300, STAT3, CDK2, HSP90AA1, and PPARG. The finding of proteomics unveiled a substantial augmentation in the expression level of HSP90AA1 in COVID-19 patient samples, especially in severe conditions.

Conclusions

Our investigation illuminate a mutual pathogenic mechanism that underlies both OSA and COVID-19, which may provide novel perspectives for future investigations into the underlying mechanisms.
Begleitmaterial
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12890-024-02921-1.
Hong Luo and Jisong Yan contributed equally to this work.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Introduction

During the past three years, an extensive number of individuals have faced the challenging period of coronavirus disease outbreaks. The latest estimates from the World Health Organization indicate a staggering 765,222,932 confirmed cases of COVID-19 as of May 3rd, 2023, with 6,921,614 reported fatalities (https://​covid19.​who.​int/​). This has placed a significant burden on healthcare systems worldwide. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of COVID-19, primarily affects the upper respiratory tract and lung tissues. Previous studies have identified older age, diabetes, severe asthma, hypertension, obesity, and cardiovascular and cerebrovascular comorbidities as high-risk factors associated with COVID-19 infection and unfavorable prognosis [13]. Intriguingly, several of these comorbidities, such as hypertension, diabetes mellitus, obesity, and vascular disease, have been shown a strong association with OSA [46]. Hence, exploring shared pathogenic mechanisms between these two diseases has captured significant interest among researchers and clinicians.
OSA is a disorder characterized by recurring episodes of pharyngeal airway collapse, leading to intermittent partial or complete cessation of breathing during sleep. This condition has been strongly associated with various cardiovascular, cerebrovascular, and metabolic disorders. In recent years, extensive clinical observational studies have indicated that individuals with OSA may be more susceptible to COVID-19 infection and experience poorer outcomes [715]. Furthermore, two meta-analyses have revealed that OSA is associated with a 2.0 and 1.7 times increased risk of severe COVID-19, respectively [16, 17].
Nevertheless, the precise pathological mechanisms underlying the association between OSA and the susceptibility to COVID-19 infection as well as the development of COVID-19 complications remain inadequately elucidated. Several investigations propose an intricate interplay involving intermittent hypoxia, oxidative stress, sympathetic activation, inflammation, endothelial dysfunction, and associated complications [17, 18]. To delve further into these intricate mechanisms, a case–control study has unveiled a correlation between sleep-related hypoxemia and an augmented likelihood of adverse COVID-19 outcomes. This discovery implies a potential linkage between COVID-19-induced hypoxia and pre-existing sleep-related hypoxemia. Furthermore, given the concurrent involvement and impact on the respiratory system in both conditions, certain studies postulate that individuals with pre-existing OSA are at an elevated risk of morbidity and mortality upon exposure to SARS-CoV-2 due to an excessive inflammatory response [19, 20]. Moreover, a review conducted by Mashaqi et al. suggested that alterations in the gut microbiota, which occur in the context of COVID-19 and OSA, can exacerbate systemic inflammation by facilitating the translocation of microorganisms across a compromised intestinal barrier [8]. Overall, the current theories and hypotheses fail to provide a comprehensive elucidation of the intrinsic molecular mechanisms underlying the association between OSA and COVID-19. Comprehending the potential interconnections and plausible molecular pathways connecting these two conditions is imperative for the effective management of COVID-19 patients with concomitant OSA and for the identification of viable therapeutic interventions.
In recent times, the clinical utilization of bioinformatics has witnessed a surge, enabling the exploration of intricate disease mechanisms. Its remarkable advantages lie in its capacity to prognosticate the structure, prognosis, functionality, and evolution of uncharted genes and proteins, while also facilitating the screening of medications and vaccines from extensive repositories of sequence data. Consequently, the objectives of this investigation encompassed an examination of the plausible association between COVID-19 and OSA, along with an in-depth exploration of the molecular mechanisms that underpin this comorbidity, employing a bioinformatics framework.

Materials and methods

Datasets information

The dataset denoted as GSE157103 encompasses a COVID-19 cohort, encompassing gene expression profiles and accompanying clinical data obtained from a cohort comprising 100 COVID-19 patients, alongside 26 non-COVID-19 patients [21]. Additionally, GSE75097 has captured gene expression profiles from 28 individuals afflicted with OSA, in conjunction with 6 healthy controls [22].

Screening feature genes based on LASSO, random forest and support vector machine algorithms

In COVID-19, we conducted gene screening employing the LASSO, random forest, and support vector machine algorithms. The resultant intersection set was derived from these methodologies to ascertain the distinctive feature genes. Furthermore, similar gene screening procedures were applied to the realm of OSA utilizing the LASSO, random forest, and support vector machine algorithms. Following this, the R package "venn" was employed to discern the overlapping feature genes that are shared between the datasets of COVID-19 and OSA.

Functional annotation of feature genes

Enrichr is an internet-based application designed for conducting gene set enrichment analysis, enabling users to investigate gene lists derived from high-throughput experiments and gain a comprehensive understanding of the biological implications and underlying mechanisms. This versatile tool incorporates over 100 gene set libraries, encompassing diverse categories such as biological pathways, Gene Ontology, transcription factor targets, protein domains, and more [23]. In this study, we employed the Enrichr tool to carry out functional annotation of the shared feature genes, encompassing Gene Ontology (GO) analysis and KEGG pathway analysis. Statistical significance was defined as a p-value less than 0.05.

Network construction for protein–protein interactions, hub gene identification and module analysis

In this investigation, we utilized the STRING online platform to investigate the interconnectedness among the shared feature genes [24]. Furthermore, the Cytoscape software was employed to illustrate and visually represent the interaction relationships among these genes [25]. To identify core nodes and modules within the network, we utilized the CytoHubba plugin, which also facilitated the identification of hub genes [26]. More specifically, the MCC algorithm of the CytoHubba plugin was employed to ascertain the hub genes, while the MCODE plugin for Cytoscape aided in the identification of core functional modules within the protein–protein interaction network [27].

Identification of TF-gene interactions and construction of TF-miRNA interaction network

To unravel the complex interplay between transcription factors (TFs), genes, and miRNAs, we leveraged the extensive resources provided by the ENCODE database and RegNetwork database, which were accessible through the NetworkAnalyst platform. This enabled us to discover TF-gene interactions and construct a TF-miRNA interaction network [28, 29].

Screening potential drugs

By utilizing the DSigDB database, we predicted plausible potential drugs that may be associated with the hub genes. DSigDB serves as a comprehensive repository of drug-gene expression profile data sourced from public databases. It provides researchers with data analysis tools and visualization interfaces to explore the relationship between drugs and gene expression, facilitating the identification of promising therapeutic targets and drugs [30]. Leveraging this database, we predicted potential target drugs linked to the hub genes, holding promise for the treatment of OSA and COVID-19.

Validation of hub genes based on proteomics and immune infiltration analysis

Drawing upon the findings elucidated in our previously disseminated proteomic investigations, conducted under the aegis of our research team, we embarked upon the validation of the expression gradients exhibited by the corresponding proteins linked with the hub genes within both the COVID-19 tissue samples and serum samples [31]. Subsequently, we engaged the CIBERSORT algorithm to quantitatively measure the extent of infiltration demonstrated by 22 distinct immune cell populations across the panorama of COVID-19 samples and OSA samples [32]. To further delineate the intricate interplay between the central genes and the immunological attributes, a comprehensive correlation analysis was conducted. To discern the diagnostic efficacy of the central genes in both COVID-19 and OSA, Receiver Operating Characteristic (ROC) curves were meticulously constructed and the Area Under the Curve (AUC) was computed individually for evaluation purposes.

Results

Identifying feature genes

The methodology employed in this investigation is delineated in Fig. 1, elucidating the process of discerning feature genes within the GSE157103 and GSE75097 datasets. A collective sum of 25 feature genes and 24 feature genes were respectively identified within these datasets (Figure S1; Figure S2). Through the implementation of intersection analysis, a set of fourteen genes commonly differentially expressed was obtained.

Functional enrichment analysis

In this study, we conducted an enrichment analysis utilizing the web-based tool Enrichr, which facilitated the identification of multiple enriched GO categories. The outcomes of the enrichment analysis revealed numerous significant biological processes, cellular components, and molecular functions that potentially hold key roles in the studied system. Noteworthy enriched biological processes encompassed gas homeostasis, caveola assembly, and Schwann cell differentiation, suggesting their relevance in the system. Moreover, various molecular functions such as ferric iron binding, oxidoreductase activity, and phosphatidylethanolamine binding were enriched, highlighting their potential significance in the system. Furthermore, the analysis yielded several significant cellular components, including the condensed chromosome, centromeric region, and autolysosome, implicating their involvement in the studied system. Notably, the enrichment analysis also highlighted the significance of the SWI/SNF complex and caveola. (Fig. 2; Table 1).
Table 1
GO terms, GO pathways and their corresponding P-values and genes for common differentially expressed genes
Category
GO ID
GO pathways
P-value
Genes
GO Biological Process
GO:0033483
gas homeostasis
9.54E-06
CAV1;HIF1A
GO:0032077
positive regulation of deoxyribonuclease activity
0.003495399
PCNA
GO:0070836
caveola assembly
0.003495399
CAV1
GO:0072584
caveolin-mediated endocytosis
0.003495399
CAV1
GO:1,903,377
negative regulation of oxidative stress-induced neuron intrinsic apoptotic signaling pathway
0.003495399
HIF1A
GO:0048550
negative regulation of pinocytosis
0.003495399
CAV1
GO:0014037
Schwann cell differentiation
0.003495399
NF1
GO:0006880
intracellular sequestering of iron ion
0.003495399
FTH1
GO:1,901,979
regulation of inward rectifier potassium channel activity
0.003495399
CAV1
GO:0061419
positive regulation of transcription from RNA polymerase II promoter in response to hypoxia
0.003495399
HIF1A
GO Cellular Component
GO:0000779
condensed chromosome, centromeric region
0.006979474
NCAPD2
GO:0044754
autolysosome
0.006979474
FTH1
GO:0005767
secondary lysosome
0.01114544
FTH1
GO:0031092
platelet alpha granule membrane
0.011838189
CD109
GO:0016514
SWI/SNF complex
0.013222334
RB1
GO:0042383
sarcolemma
0.03580243
CAV1
GO:0000793
condensed chromosome
0.03715538
NCAPD2
GO:1,904,724
tertiary granule lumen
0.037831194
FTH1
GO:0005901
caveola
0.041203662
CAV1
GO:0005776
autophagosome
0.047246461
FTH1
GO Molecular Function
GO:0032135
DNA insertion or deletion binding
0.003495399
PCNA
GO:0035035
histone acetyltransferase binding
9.48E-05
PCNA;HIF1A
GO:0005113
patched binding
0.004890388
CAV1
GO:0032405
MutLalpha complex binding
0.004890388
PCNA
GO:0008199
ferric iron binding
0.004890388
FTH1
GO:0004322
ferroxidase activity
0.004890388
FTH1
GO:0016724
oxidoreductase activity, acting on metal ions, oxygen as acceptor
0.004890388
FTH1
GO:0050998
nitric-oxide synthase binding
0.004890388
CAV1
GO:0019870
potassium channel inhibitor activity
0.006283564
CAV1
GO:0008429
phosphatidylethanolamine binding
0.006283564
NF1
The enrichment analysis revealed noteworthy pathways derived from four comprehensive databases: KEGG, WikiPathways, Reactome, and BioCarta. Among these databases, the prominent pathways identified encompassed DNA repair and replication pathways, regulation of the cell cycle, as well as pathways associated with cancer, such as bladder cancer, Fanconi anemia pathway, and Kaposi sarcoma-associated herpesvirus infection. Additionally, several pathways pertaining to metabolism, aging, and gene regulation were also identified. Collectively, these findings provide valuable insights into the intricate molecular mechanisms that underlie diverse biological processes (Fig. 3; Table 2).
Table 2
Top pathways from KEGG, WikiPathways, Reactome and BioCarta databases and their corresponding P-values and genes for common differentially expressed genes
Databases
Pathways
P-value
Genes
KEGG
Mismatch repair
0.015985227
PCNA
Base excision repair
0.022861069
PCNA
DNA replication
0.024915106
PCNA
Cell cycle
0.003304756
RB1;PCNA
Bladder cancer
0.028329595
RB1
Ferroptosis
0.028329595
FTH1
Nucleotide excision repair
0.032412327
PCNA
Hepatitis B
0.005566263
RB1;PCNA
Fanconi anemia pathway
0.03715538
UBE2T
Kaposi sarcoma-associated herpesvirus infection
0.007811021
RB1;HIF1A
WikiPathways
LncRNA-mediated mechanisms of therapeutic resistance WP3672
0.00419312
HIF1A
Pilocytic astrocytoma WP2253
0.00419312
NF1
MicroRNA for Targeting Cancer Growth and Vascularization in Glioblastoma WP3593
0.004890388
HIF1A
HIF1A and PPARG regulation of glycolysis WP2456
0.005587202
HIF1A
Glycolysis in senescence WP5049
0.007674931
RB1
NAD metabolism, sirtuins and aging WP3630
0.007674931
HIF1A
Iron metabolism in placenta WP2007
0.008369936
FTH1
Aryl Hydrocarbon Receptor Netpath WP2586
0.000462719
RB1;NF1
H19 action Rb-E2F1 signaling and CDK-Beta-catenin activity WP3969
0.009758591
RB1
ID signaling pathway WP53
0.01114544
RB1
Reactome
PTK6 Expression R-HSA-8849473
0.003495399
HIF1A
SARS-CoV-1 Genome Replication And Transcription R-HSA-9679514
0.003495399
RB1
SARS-CoV-2 Genome Replication And Transcription R-HSA-9694682
0.003495399
RB1
NOSTRIN Mediated eNOS Trafficking R-HSA-203641
0.003495399
CAV1
PTK6 Promotes HIF1A Stabilization R-HSA-8857538
0.00419312
HIF1A
RAS Signaling Downstream Of NF1 Loss-Of-Function Variants R-HSA-6802953
0.00419312
NF1
CD163 Mediating An Anti-Inflammatory Response R-HSA-9662834
0.006283564
PLK2
Condensation Of Prometaphase Chromosomes R-HSA-2514853
0.006979474
NCAPD2
STAT3 Nuclear Events Downstream Of ALK Signaling R-HSA-9701898
0.006979474
HIF1A
eNOS Activation R-HSA-203615
0.007674931
CAV1
BioCarta
p53 Signaling Pathway Homo sapiens h p53Pathway
3.53E-05
RB1;PCNA
Overview of telomerase RNA component gene hTerc Transcriptional Regulation Homo sapiens h tercPathway
0.004890388
RB1
VEGF, Hypoxia, and Angiogenesis Homo sapiens h vegfPathway
0.000195723
CAV1;HIF1A
BTG family proteins and cell cycle regulation Homo sapiens h btg2Pathway
0.006283564
RB1
Cyclin E Destruction Pathway Homo sapiens h fbw7Pathway
0.006283564
RB1
E2F1 Destruction Pathway Homo sapiens h skp2e2fPathway
0.006979474
RB1
Regulation of transcriptional activity by PML Homo sapiens h pmlPathway
0.007674931
RB1
Regulation of p27 Phosphorylation during Cell Cycle Progression Homo sapiens h p27Pathway
0.009064489
RB1
RB Tumor Suppressor/Checkpoint Signaling in response to DNA damage Homo sapiens h rbPathway
0.009064489
RB1
Erythropoietin mediated neuroprotection through NF-kB Homo sapiens h eponfkbPathway
0.009064489
HIF1A

Protein–protein interaction network

The protein–protein interaction network visually illustrates the tightly interconnected relationships among the common feature genes (Fig. 4A). Utilizing the cytoHubba plugin, we identified the top 10 central genes within the network, namely TP53, CCND1, MDM2, RB1, HIF1A, EP300, STAT3, CDK2, HSP90AA1, and PPARG (Fig. 4B). Furthermore, employing the MCODE plugin, we extracted four significant gene modules (Fig. 5). Notably, Module 1 encompasses all of the hub genes, with the exception of EP300.

TF-gene interactions and construction of TF-miRNA interaction network

The NetworkAnalyst tool was employed to predict TFs capable of interacting with the fourteen commonly expressed feature genes, and to construct a visually informative network diagram depicting these interaction relationships. The resultant network comprised 213 TFs, 227 nodes, and 402 edges. Notably, FTH1 and PCNA emerged as potential key players in the context of COVID-19 infection complicated by OSA (Fig. 6). Moreover, a TF-miRNA co-regulation network was constructed, which facilitated the prediction of connections among miRNAs, TFs, and hub genes. This comprehensive network encompassed 529 nodes and 671 edges, with 191 miRNAs and 324 TF genes interacting with the hub genes (Fig. 7).

Screening potential drugs

Following a rigorous screening process, we have successfully identified ten potential drug candidates that hold promise in the treatment of COVID-19 in conjunction with OSA. These drugs encompass acetaminophen (CTD 00005295), 5-Fluorouracil (CTD 00005987), genistein (CTD 00007324), estradiol (CTD 00005920), topotecan (CTD 00007293), POTASSIUM NITRATE (CTD 00001112), simvastatin (CTD 00007319), 7646–79-9 (CTD 00000928), Pinosylvin (CTD 00002139), and 2,4-dichlorophenoxyacetic acid (CTD 00007028). A comprehensive description of these drugs is provided in Table 3.
Table 3
Suggested top drug compounds for COVID-19 and OSA
Term
P-value
Adjusted P-value
Genes
acetaminophen CTD 00005295
5.67E-06
0.002117207
RB1;PCNA;CAV1;CD109;FTH1;UBE2T;
NF1;SMARCAD1;PLS3;PLOD2;NCAPD2
5-Fluorouracil CTD 00005987
6.57E-06
0.002117207
RB1;PCNA;CAV1;PLK2;UBE2T;PLOD2;HIF1A
genistein CTD 00007324
7.70E-06
0.002117207
RB1;PCNA;CAV1;PLK2;UBE2T;NCAPD2;HIF1A
estradiol CTD 00005920
9.25E-06
0.002117207
RB1;PCNA;CAV1;PLK2;CD109;FTH1;
UBE2T;NF1;PLS3;PLOD2;NCAPD2
topotecan CTD 00007293
2.16E-05
0.003806737
PCNA;FTH1;PLOD2;HIF1A
POTASSIUM NITRATE CTD 00001112
2.49E-05
0.003806737
PCNA;FTH1
simvastatin CTD 00007319
4.64E-05
0.004501554
RB1;PCNA;CAV1;HIF1A
7646–79-9 CTD 00000928
4.78E-05
0.004501554
PCNA;CAV1;PLK2;FTH1;UBE2T;PLS3;
PLOD2;NCAPD2;HIF1A
Pinosylvin CTD 00002139
4.91E-05
0.004501554
RB1;PCNA;FTH1
2,4-dichlorophenoxyacetic acid CTD 00007028
5.43E-05
0.004501554
PCNA;FTH1

Verification of the hub genes in clinical samples

Among the central genes, we observed the discernment of corresponding protein entities solely for STAT3, CDK2, and HSP90AA1. Within the confines of COVID-19 pulmonary tissue specimens, the expression profiles of CDK2 and HSP90AA1 exhibited an elevation vis-à-vis control samples, while the expression profile of STAT3 demonstrated a reduction in relation to control samples, albeit without attaining statistical significance (Fig. 8A-C). Moreover, we embarked on an exploration of the prognostic implications encapsulated within the HSP90AA1 proteins present within clinical serum samples originating from diverse COVID-19 cases, each marked by disparate outcomes (Fatal (F) = 20, Severe (S) = 14, Mild (M) = 20, Healthy (H) = 7). Our findings unveiled a notable escalation in the plasma concentration of HSP90AA1 in the context of more severe manifestations of COVID-19 (Fig. 8D).

Immune infiltration analysis and ROC curve

We undertook a comprehensive analysis of the intricate interrelationship existing between HSP90AA1 and immune cell populations, both in the context of COVID-19 and OSA. Within the ambit of COVID-19, a positive correlation was discerned between HSP90AA1 and activated CD4 memory T cells, as well as resting and activated dendritic cells, CD8 T cells, monocytes, and gamma delta T cells. Conversely, a negative correlation was observed between HSP90AA1 and neutrophils, along with M0 macrophages. In the landscape of OSA, HSP90AA1 exhibited positive correlations with resting CD4 memory T cells, memory B cells, M1 macrophages, eosinophils, and activated mast cells. On the other hand, a negative correlation was noted between HSP90AA1 and resting mast cells (Fig. 9A-B). The diagnostic potential of the HSP90AA1 gene was meticulously assessed through the construction of ROC curves. Within the confines of the COVID-19 dataset, the HSP90AA1 gene demonstrated commendable diagnostic efficacy (AUC: 0.745) in discriminating individuals afflicted by SARS-CoV-2 from their healthy counterparts. In the expanse of the OSA dataset, the HSP90AA1 gene achieved an AUC value of 0.620 (Fig. 9C).

Discussion

Various observational studies have indicated that individuals afflicted with OSA exhibit a heightened vulnerability to severe SARS-CoV-2 infection [10, 1517], albeit this association diminishes upon adjustment for other comorbidities (such as obesity and cardiovascular disorders) [33]. A Mendelian randomization study also yielded inconclusive evidence regarding the causal relationship between OSA and COVID-19. Conversely, a population-based study demonstrated that even after accounting for the effects of other comorbidities (e.g., obesity), OSA remains capable of doubling the risk of COVID-19-related adverse events, including hospitalization and death. Consequently, further comprehensive investigations are imperative to elucidate the intrinsic relationship between OSA and COVID-19. Within this study, fourteen feature genes shared by COVID-19 and OSA have been identified. Subsequently, through the construction of a protein–protein interaction network incorporating these common feature genes, we have identified 10 central genes (TP53, CCND1, MDM2, RB1, HIF1A, EP300, STAT3, CDK2, HSP90AA1, and PPARG). The subsequent analysis proceeded with functional enrichment analysis, TF-gene interactions, TF-miRNA interaction networks, and drug candidate screening.
Protein–protein interaction is the process by which two or more protein molecules form a protein complex through non-covalent bonds. At the molecular level, viruses invade cells and use a complex network of protein–protein interactions to manipulate cellular processes, and host cells initiate activation of innate antiviral defenses and adaptive immune systems to control viral replication through complex protein–protein interactions. Therefore, knowledge of protein–protein interactions is essential for understanding the virus-host relationship. Zhang et al. identified relevant genes in the virus-human protein interaction network, including UBL4A, GNB1, UMPS, POTEF, UBL4B, UBBP4, PARK2, PTPN11, and SCOC based on a random walk model [34]. In particular, Yang et al. developed the Human-Virus Interaction DataBase (HVIDB, http://​zzdlab.​com/​hvidb/​), which can accurately predict interactions between human host and viral proteins [35].
Currently, integrated machine learning based on multiple feature selection algorithms has been widely used to find biomarkers for diseases, which can screen potential biomarkers with high sensitivity, accuracy, and stability, and construct efficient and stable diagnostic models. Since the outbreak of COVID-19, numerous studies have analyzed transcriptomic, proteomic, metabolomic, and epigenomic data from COVID-19 patients based on machine learning to find reliable and stable COVID-19 biomarkers. Guo et al. developed a random forest machine learning model based on proteomic and metabolomic data from COVID-19 patients, which incorporated genes such as SAA2,ALB,CRP,SAA1,HABP2, and HP, and was able to discriminate well between non-severe and severe COVID-19 patients (AUC = 0.957) [36]. Using LASSO and ridge regression penalties, Zhou et al. identified a biomarker combination containing ORM1/AGP1, ORM2, FETUB, and CETP that demonstrated excellent reliability in distinguishing COVID-19 patients from healthy volunteers [31]. Cai et al. identified five biomarkers of COVID-19 by applying Boruta and minimum redundancy maximum relevance methods, which were PSMB8, COLCA2, FAM83A, LGALS3BP and IRF9. In addition, Cai et al. analyzed the methylation dataset of COVID-19 by utilizing the Monte Carlo feature selection method analyzed the methylation dataset of COVID-19 and found that EPSTI1, NACAP1, SHROOM3, C19ORF35 and MX1 as key features could significantly distinguish COVID-19 patients from non-COVID-19 patients [37, 38].
Prior investigations have identified five of these central genes (TP53, HIF1A, STAT3, HSP90AA1, PPARG) to be linked to the pathogenesis of both COVID-19 and OSA. The tumor suppressor gene TP53, a crucial participant in the apoptotic signaling cascade, assumes a significant role in monitoring cellular division and possesses reparative effects on aberrant division and cellular damage. Multiple studies have revealed a downregulation of TP53 gene expression in patients infected with SARS-CoV-2, consequently fostering viral replication and increasing the likelihood of tumorigenesis [3942]. Intriguingly, periodic intermittent hypoxia has been implicated as a causative factor for non-small cell lung cancer in TP53fl/fl mice, thereby facilitating accelerated tumor growth [43]. Hence, we postulate that individuals afflicted with the concomitant presence of these two ailments are predisposed to a heightened risk of cancer or tumor development, ultimately resulting in diminished overall survival rates.
HIF1A (hypoxia-inducible factor 1-alpha), serves as a prominent regulator of cellular and systemic homeostatic responses to hypoxia by orchestrating the transcriptional activation of numerous genes. These genes encompass those involved in energy metabolism, angiogenesis, apoptosis, and others whose protein products enhance oxygen delivery or facilitate metabolic adaptation to hypoxic conditions [44]. Notably, previous investigations have indicated that the prognosis of COVID-19 is significantly influenced by the cytokine storm triggered by SARS-CoV-2. Transcriptome analyses have unveiled that SARS-CoV-2 infection induces mitochondrial damage and the generation of mitochondrial-derived reactive oxygen species (Mito-ROS), thereby promoting the expression of HIF-1A and consequent SARS-CoV-2 infection and cytokine production [45, 46]. Furthermore, HIF-1A assumes a vital role in regulating metabolic pathways and inflammatory processes [47]. The dysregulation of the HIF-1A pathway contributes to the development of various diseases, including cancer, cardiovascular disease, and Alzheimer's disease [4851]. Importantly, many of these disorders have been demonstrated to be associated with severe SARS-CoV-2 infection, thereby suggesting the prognostic impact of HIF-1A on COVID-19 [52]. On the other hand, numerous studies have illustrated the close association between HIF-1A and OSA, given that OSA is characterized by chronic intermittent hypoxia (CIH). CIH stimulates the upregulation of the hypoxia-inducible transcription factor HIF1A, contributing to the independent risk of OSA for atherosclerosis and heightened cardiovascular mortality. Mechanistically, hypertension, dyslipidemia, insulin resistance, systemic inflammation, and oxidative stress are among the underlying mechanisms through which CIH accelerates atherosclerotic disease [53]. A recent study has demonstrated that mice exposed to CIH exhibited impaired clearance of triglyceride-rich lipoproteins. Subsequent investigations have suggested that this phenomenon may be attributed to the transcriptional regulation of HIF-1A, which upregulates the expression of the lipoprotein lipase (LPL) inhibitor (Angptl4), thereby inactivating LPL and impeding lipid clearance [54]. Additionally, CIH exacerbates non-alcoholic fatty liver disease (NAFLD) by inducing hepatic oxidative stress via the activation of the HIF-1A gene [55]. Furthermore, HIF-1 has been implicated in the pathological processes of hypoxia and has been associated with various other diseases such as diabetes mellitus, idiopathic pulmonary fibrosis, pulmonary arterial hypertension, systemic hypertension, myocardial injury, and cognitive deficits [5661]. In summary, the myriad metabolic and pathophysiological responses elicited by intermittent hypoxia in individuals with OSA may contribute to their susceptibility to COVID-19 and portend a poorer prognosis.
Inflammation constitutes a hallmark feature of both OSA and SARS-CoV-2 infections. Hypotheses suggest that alterations in the gut microbiota, which are associated with COVID-19 and OSA, may compromise the integrity of the intestinal barrier, thereby facilitating the translocation of bacteria into the systemic circulation and subsequently promoting systemic inflammation [8]. Notably, systemic inflammation is particularly pronounced in the context of COVID-19. STATs (signal transducer and activator of transcription) are well-known components of the JAK/STAT signaling pathway. Previous investigations have revealed that the viral component of SARS-CoV-2 induces malfunctioning of STAT1 and compensatory overactivation of STAT3. Consequently, a positive feedback loop between STAT3 and plasminogen activator inhibitor-1 (PAI-1) is established in SARS-CoV-2-infected cells, thereby perpetuating a cycle of activation that triggers the release of pro-inflammatory cytokines and chemokines [62].
In a transcriptomic analysis, the connection between HSP90A family class A member 1 (HSP90AA1) and the quantity of SARS-CoV-2 RNA was unveiled [63]. Moreover, HSP90AA1 was identified as a risk factor for the coexistence of COVID-19 and cardiovascular disease [64]. Notably, HSP90 levels were significantly elevated in patients with OSAS when compared to control subjects [65]. These findings suggest that HSP90 may potentially play a role in the progression of both of these diseases.
PPARG (peroxisome proliferator-activated receptor gamma), a crucial transcription factor, governs both inflammation and fatty acid metabolism. Studies have demonstrated that PPARG agonists can enhance the expression of ACE2, thereby contributing to the invasion of SARS-CoV-2 [66]. Furthermore, OSA is characterized by metabolic dysfunction and obesity, and the PPARG gene is implicated in the regulation of lipid metabolism and glucose homeostasis. This suggests that PPARG may also play a potential role in the pathophysiological mechanisms underlying the comorbidity of COVID-19 and OSA [67]. As for the other five central genes (CCND1, MDM2, RB1, EP300, CDK2) in the study, their associations with OSA patients have not been reported. Therefore, future investigations could delve into exploring the potential mechanisms that underlie the comorbidity between COVID-19 and OSA from additional perspectives.
Condensed chromosomes, centromeric regions, autolysosomes, and molecular functions related to DNA insertion or deletion was enriched in this study. The primary constriction along a condensed chromosome is known as the centromere region, and the end of the chromosome as the telomere. These regions are essential for the proper integrity and segregation of chromosomes, as well as for the hierarchy of order within the cell nucleus. A study conducted at the Federal University of São Paulo in Brazil shows that the telomere shortening that naturally occurs with aging and is accelerated by OSA can be mitigated by the use of continuous positive airway pressure, indicating that accelerated telomere shortening due to OSA can therefore lead to premature cell aging [68]. It is now well established that SARS-CoV-2 can enter host cells via the ACE2 receptor. Interestingly, the gene encoding ACE2 is located on an X chromosome, and according to Lyon's theory, one of the two X chromosomes is transcriptionally silenced, a process that results in the condensation of an X chromosome into a compact structure known as a barr body [69, 70]. Autolysosome is formed when an autophagosome or phagosome fuses with a lysosome [71]. As a marker of autolysosomal degradation, P62 protein expression in blood cytotoxic T cells was increased in OSA patients compared to primary snoring patients, indicating that OSA patients had impaired autophagy activity [72]. In COVID-19, the SARS-CoV-2 protein ORF3a affects autolysosome formation by hindering the assembly of the SNARE complex, and furthermore, SARS-CoV-2 infection-induced autolysosome accumulation facilitates progeny virus propagation [7375]. As for DNA insertion or deletion, it was shown that angiotensin-converting enzyme gene insertion/deletion polymorphisms differed significantly in OSA disease groups with different severities and may be involved in the pathogenesis of hypertension [76, 77]. In COVID-19, insertion-and-deletion mutations in several genes including Nsp2, Nsp3, S1, and ORF8 genes have been observed, which may provide further insights into the developmental process of SARS-CoV-2 [78, 79].
The study revealed that gas homeostasis may be a crucial shared pathogenic mechanism in COVID-19 and OSA, as indicated by the analysis of GO enrichment. Gas homeostasis refers to the tightly regulated maintenance of oxygen and carbon dioxide levels in the human body, involving multiple pathways. Recent research has suggested the involvement of these pathways in the pathogenesis of both COVID-19 and OSA. Hypoxia-inducible factors (HIFs) are known regulators of cellular responses to low oxygen levels. We have discussed in detail the significant role of HIF1A in the development of COVID-19 and OSA patients. Furthermore, OSA patients experience chronic intermittent hypoxia, while the increased production of pro-inflammatory cytokines in COVID-19 also affects gas exchange and disrupts gas homeostasis [80]. Oxidoreductase activity refers to the enzymatic activity involved in redox reactions, which is closely associated with oxidative stress [81]. It is increasingly recognized that COVID-19 pathogenesis and immunopathogenesis are closely linked to oxidative stress and cytokine storm. Oxidative stress contributes to the development of endothelial dysfunction, while the cytokine storm exacerbates this effect. Ultimately, these events trigger the activation of the blood clotting cascade, leading to blood coagulation and microvascular thrombosis. In the case of OSA, intermittent hypoxia and reoxygenation during sleep-disordered breathing also induce oxidative stress and inflammation[82, 83]. This resulting oxidative damage further contributes to the development and worsening of complications associated with OSA, including obesity, metabolic dysfunction, cardiovascular diseases, and cognitive impairment [82, 84, 85]. DNA repair and replication pathways play vital roles in maintaining the stability of the genome and ensuring cell survival. Recent studies have revealed that these pathways may also be involved in the pathogenesis of COVID-19 and OSA. Emerging evidence suggests that viral infection can induce DNA damage and trigger an altered DNA damage response. Specifically, certain products of SARS-CoV-2, such as ORF6 and NSP13 proteins, disrupt the DNA damage response kinase CHK1 and reduce the production of deoxynucleoside triphosphate (dNTP), which consequently leads to DNA damage response. In the case of OSA, increased levels of reactive oxygen species and repeated hypoxic conditions also disrupt DNA repair mechanisms and contribute to enhanced DNA damage [86]. In summary, gas homeostasis, oxidoreductase activity, and DNA repair and replication pathways may contribute to the shared pathogenesis of COVID-19 and OSA. Further studies are required to fully understand the precise roles of these pathways in these diseases and to explore their potential as therapeutic targets.
In this study, ten drug candidates were identified. To further strengthen the credibility of the results, we searched databases of information about drugs for the treatment of COVID-19, including DockCoV2 and DrugBank, assembled by Huang et al. [8790]. Of these, Topotecan and Estradiol had docking scores of -16.8 and -10.7, respectively, indicating potential as therapeutic agents.
It is important to acknowledge the limitations of this study. Firstly, the analysis was conducted using data from patients diagnosed with COVID-19 and OSA, rather than individuals specifically diagnosed with both disorders. This may impact the generalizability of the findings to the co-occurrence of COVID-19 and OSA. Additionally, it should be noted that the identified genes were not experimentally validated in OSA samples.

Conclusions

In conclusion, this investigation revealed a set of genes, molecular networks, and signaling pathways that are associated with both COVID-19 and OSA. These findings provide insights and potential avenues for further research into the molecular mechanisms underlying these two diseases. The study highlights a shared regulatory scheme and signaling mechanism between COVID-19 and OSA.

Acknowledgements

Not applicable.

Declarations

Not applicable.
Not applicable.

Competing interests

The authors declare no competing interests.
Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://​creativecommons.​org/​licenses/​by/​4.​0/​. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge

Supplementary Information

Literatur
1.
Zurück zum Zitat Williamson EJ, Walker AJ, Bhaskaran K, Bacon S, Bates C, Morton CE, Curtis HJ, Mehrkar A, Evans D, Inglesby P, et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature. 2020;584(7821):430–6.PubMedPubMedCentralCrossRef Williamson EJ, Walker AJ, Bhaskaran K, Bacon S, Bates C, Morton CE, Curtis HJ, Mehrkar A, Evans D, Inglesby P, et al. Factors associated with COVID-19-related death using OpenSAFELY. Nature. 2020;584(7821):430–6.PubMedPubMedCentralCrossRef
2.
Zurück zum Zitat Zhou Y, Chi J, Lv W, Wang Y. Obesity and diabetes as high-risk factors for severe coronavirus disease 2019 (Covid-19). Diabetes Metab Res Rev. 2021;37(2):e3377.PubMedCrossRef Zhou Y, Chi J, Lv W, Wang Y. Obesity and diabetes as high-risk factors for severe coronavirus disease 2019 (Covid-19). Diabetes Metab Res Rev. 2021;37(2):e3377.PubMedCrossRef
3.
Zurück zum Zitat Li X, Xu S, Yu M, Wang K, Tao Y, Zhou Y, Shi J, Zhou M, Wu B, Yang Z, et al. Risk factors for severity and mortality in adult COVID-19 inpatients in Wuhan. J Allergy Clin Immunol. 2020;146(1):110–8.PubMedPubMedCentralCrossRef Li X, Xu S, Yu M, Wang K, Tao Y, Zhou Y, Shi J, Zhou M, Wu B, Yang Z, et al. Risk factors for severity and mortality in adult COVID-19 inpatients in Wuhan. J Allergy Clin Immunol. 2020;146(1):110–8.PubMedPubMedCentralCrossRef
5.
Zurück zum Zitat Gottlieb DJ, Punjabi NM. Diagnosis and Management of Obstructive Sleep Apnea: A Review. JAMA. 2020;323(14):1389–400.PubMedCrossRef Gottlieb DJ, Punjabi NM. Diagnosis and Management of Obstructive Sleep Apnea: A Review. JAMA. 2020;323(14):1389–400.PubMedCrossRef
6.
Zurück zum Zitat Javaheri S, Barbe F, Campos-Rodriguez F, Dempsey JA, Khayat R, Javaheri S, Malhotra A, Martinez-Garcia MA, Mehra R, Pack AI, et al. Sleep Apnea: Types, Mechanisms, and Clinical Cardiovascular Consequences. J Am Coll Cardiol. 2017;69(7):841–58.PubMedPubMedCentralCrossRef Javaheri S, Barbe F, Campos-Rodriguez F, Dempsey JA, Khayat R, Javaheri S, Malhotra A, Martinez-Garcia MA, Mehra R, Pack AI, et al. Sleep Apnea: Types, Mechanisms, and Clinical Cardiovascular Consequences. J Am Coll Cardiol. 2017;69(7):841–58.PubMedPubMedCentralCrossRef
7.
8.
Zurück zum Zitat Mashaqi S, Kallamadi R, Matta A, Quan SF, Patel SI, Combs D, Estep L, Lee-Iannotti J, Smith C, Parthasarathy S et al: Obstructive Sleep Apnea as a Risk Factor for COVID-19 Severity-The Gut Microbiome as a Common Player Mediating Systemic Inflammation via Gut Barrier Dysfunction. Cells 2022, 11(9). Mashaqi S, Kallamadi R, Matta A, Quan SF, Patel SI, Combs D, Estep L, Lee-Iannotti J, Smith C, Parthasarathy S et al: Obstructive Sleep Apnea as a Risk Factor for COVID-19 Severity-The Gut Microbiome as a Common Player Mediating Systemic Inflammation via Gut Barrier Dysfunction. Cells 2022, 11(9).
9.
Zurück zum Zitat Tufik S, Gozal D, Ishikura IA, Pires GN, Andersen ML. Does obstructive sleep apnea lead to increased risk of COVID-19 infection and severity? J Clin Sleep Med. 2020;16(8):1425–6.PubMedPubMedCentralCrossRef Tufik S, Gozal D, Ishikura IA, Pires GN, Andersen ML. Does obstructive sleep apnea lead to increased risk of COVID-19 infection and severity? J Clin Sleep Med. 2020;16(8):1425–6.PubMedPubMedCentralCrossRef
10.
Zurück zum Zitat Maas MB, Kim M, Malkani RG, Abbott SM, Zee PC. Obstructive Sleep Apnea and Risk of COVID-19 Infection Hospitalization and Respiratory Failure. Sleep Breath. 2021;25(2):1155–7.PubMedCrossRef Maas MB, Kim M, Malkani RG, Abbott SM, Zee PC. Obstructive Sleep Apnea and Risk of COVID-19 Infection Hospitalization and Respiratory Failure. Sleep Breath. 2021;25(2):1155–7.PubMedCrossRef
11.
Zurück zum Zitat Peker Y, Celik Y, Arbatli S, Isik SR, Balcan B, Karataş F, Uzel FI, Tabak L, Çetin B, Baygül A, et al. Effect of High-Risk Obstructive Sleep Apnea on Clinical Outcomes in Adults with Coronavirus Disease 2019: A Multicenter, Prospective, Observational Clinical Trial. Ann Am Thorac Soc. 2021;18(9):1548–59.PubMedPubMedCentralCrossRef Peker Y, Celik Y, Arbatli S, Isik SR, Balcan B, Karataş F, Uzel FI, Tabak L, Çetin B, Baygül A, et al. Effect of High-Risk Obstructive Sleep Apnea on Clinical Outcomes in Adults with Coronavirus Disease 2019: A Multicenter, Prospective, Observational Clinical Trial. Ann Am Thorac Soc. 2021;18(9):1548–59.PubMedPubMedCentralCrossRef
12.
Zurück zum Zitat de Kruif MD, Voncken SFJ, Laven SAJS, Feron TMH, Kolfoort-Otte AAB. Obstructive sleep apnea and risk of COVID-19 infection, hospitalization and respiratory failure. Sleep Breath. 2021;25(4):2103.PubMedPubMedCentralCrossRef de Kruif MD, Voncken SFJ, Laven SAJS, Feron TMH, Kolfoort-Otte AAB. Obstructive sleep apnea and risk of COVID-19 infection, hospitalization and respiratory failure. Sleep Breath. 2021;25(4):2103.PubMedPubMedCentralCrossRef
13.
Zurück zum Zitat Kendzerska T, Povitz M, Gershon AS, Ryan CM, Talarico R, Franco Avecilla DA, Robillard R, Ayas NT, Pendharkar SR: Association of clinically significant obstructive sleep apnoea with risks of contracting COVID-19 and serious COVID-19 complications: a retrospective population-based study of health administrative data. Thorax 2023. Kendzerska T, Povitz M, Gershon AS, Ryan CM, Talarico R, Franco Avecilla DA, Robillard R, Ayas NT, Pendharkar SR: Association of clinically significant obstructive sleep apnoea with risks of contracting COVID-19 and serious COVID-19 complications: a retrospective population-based study of health administrative data. Thorax 2023.
14.
Zurück zum Zitat Rögnvaldsson KG, Eyþórsson ES, Emilsson ÖI, Eysteinsdóttir B, Pálsson R, Gottfreðsson M, Guðmundsson G, Steingrímsson V. Obstructive sleep apnea is an independent risk factor for severe COVID-19: a population-based study. Sleep. 2022;45(3):zsab272. Rögnvaldsson KG, Eyþórsson ES, Emilsson ÖI, Eysteinsdóttir B, Pálsson R, Gottfreðsson M, Guðmundsson G, Steingrímsson V. Obstructive sleep apnea is an independent risk factor for severe COVID-19: a population-based study. Sleep. 2022;45(3):zsab272.
15.
Zurück zum Zitat Strausz S, Kiiskinen T, Broberg M, Ruotsalainen S, Koskela J, Bachour A, Palotie A, Palotie T, Ripatti S, Ollila HM. Sleep apnoea is a risk factor for severe COVID-19. BMJ Open Respir Res. 2021;8(1):e000845. Strausz S, Kiiskinen T, Broberg M, Ruotsalainen S, Koskela J, Bachour A, Palotie A, Palotie T, Ripatti S, Ollila HM. Sleep apnoea is a risk factor for severe COVID-19. BMJ Open Respir Res. 2021;8(1):e000845.
16.
Zurück zum Zitat Hariyanto TI, Kurniawan A. Obstructive sleep apnea (OSA) and outcomes from coronavirus disease 2019 (COVID-19) pneumonia: a systematic review and meta-analysis. Sleep Med. 2021;82:47–53.PubMedPubMedCentralCrossRef Hariyanto TI, Kurniawan A. Obstructive sleep apnea (OSA) and outcomes from coronavirus disease 2019 (COVID-19) pneumonia: a systematic review and meta-analysis. Sleep Med. 2021;82:47–53.PubMedPubMedCentralCrossRef
17.
Zurück zum Zitat Miller MA, Cappuccio FP. A systematic review of COVID-19 and obstructive sleep apnoea. Sleep Med Rev. 2021;55:101382.PubMedCrossRef Miller MA, Cappuccio FP. A systematic review of COVID-19 and obstructive sleep apnoea. Sleep Med Rev. 2021;55:101382.PubMedCrossRef
18.
Zurück zum Zitat Fleming WE, Ferouz-Colborn A, Samoszuk MK, Azad A, Lu J, Riley JS, Cruz AB, Podolak S, Clark DJ, Bray KR, et al. Blood biomarkers of endocrine, immune, inflammatory, and metabolic systems in obstructive sleep apnea. Clin Biochem. 2016;49(12):854–61.PubMedCrossRef Fleming WE, Ferouz-Colborn A, Samoszuk MK, Azad A, Lu J, Riley JS, Cruz AB, Podolak S, Clark DJ, Bray KR, et al. Blood biomarkers of endocrine, immune, inflammatory, and metabolic systems in obstructive sleep apnea. Clin Biochem. 2016;49(12):854–61.PubMedCrossRef
19.
Zurück zum Zitat Tay MZ, Poh CM, Rénia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol. 2020;20(6):363–74.PubMedPubMedCentralCrossRef Tay MZ, Poh CM, Rénia L, MacAry PA, Ng LFP. The trinity of COVID-19: immunity, inflammation and intervention. Nat Rev Immunol. 2020;20(6):363–74.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Pena Orbea C, Wang L, Shah V, Jehi L, Milinovich A, Foldvary-Schaefer N, Chung MK, Mashaqi S, Aboussouan L, Seidel K, et al. Association of Sleep-Related Hypoxia With Risk of COVID-19 Hospitalizations and Mortality in a Large Integrated Health System. JAMA Netw Open. 2021;4(11):e2134241.PubMedPubMedCentralCrossRef Pena Orbea C, Wang L, Shah V, Jehi L, Milinovich A, Foldvary-Schaefer N, Chung MK, Mashaqi S, Aboussouan L, Seidel K, et al. Association of Sleep-Related Hypoxia With Risk of COVID-19 Hospitalizations and Mortality in a Large Integrated Health System. JAMA Netw Open. 2021;4(11):e2134241.PubMedPubMedCentralCrossRef
21.
Zurück zum Zitat Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, et al. Large-Scale Multi-omic Analysis of COVID-19 Severity. Cell Syst. 2021;12(1):23-40.e27.PubMedCrossRef Overmyer KA, Shishkova E, Miller IJ, Balnis J, Bernstein MN, Peters-Clarke TM, Meyer JG, Quan Q, Muehlbauer LK, Trujillo EA, et al. Large-Scale Multi-omic Analysis of COVID-19 Severity. Cell Syst. 2021;12(1):23-40.e27.PubMedCrossRef
22.
Zurück zum Zitat Chen YC, Chen KD, Su MC, Chin CH, Chen CJ, Liou CW, Chen TW, Chang YC, Huang KT, Wang CC, et al. Genome-wide gene expression array identifies novel genes related to disease severity and excessive daytime sleepiness in patients with obstructive sleep apnea. PLoS ONE. 2017;12(5):e0176575.PubMedPubMedCentralCrossRef Chen YC, Chen KD, Su MC, Chin CH, Chen CJ, Liou CW, Chen TW, Chang YC, Huang KT, Wang CC, et al. Genome-wide gene expression array identifies novel genes related to disease severity and excessive daytime sleepiness in patients with obstructive sleep apnea. PLoS ONE. 2017;12(5):e0176575.PubMedPubMedCentralCrossRef
23.
Zurück zum Zitat Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90-97.PubMedPubMedCentralCrossRef Kuleshov MV, Jones MR, Rouillard AD, Fernandez NF, Duan Q, Wang Z, Koplev S, Jenkins SL, Jagodnik KM, Lachmann A, et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 2016;44(W1):W90-97.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607-d613.PubMedCrossRef Szklarczyk D, Gable AL, Lyon D, Junge A, Wyder S, Huerta-Cepas J, Simonovic M, Doncheva NT, Morris JH, Bork P, et al. STRING v11: protein-protein association networks with increased coverage, supporting functional discovery in genome-wide experimental datasets. Nucleic Acids Res. 2019;47(D1):D607-d613.PubMedCrossRef
25.
Zurück zum Zitat Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.PubMedPubMedCentralCrossRef Shannon P, Markiel A, Ozier O, Baliga NS, Wang JT, Ramage D, Amin N, Schwikowski B, Ideker T. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498–504.PubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. CytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 2014;8 Suppl 4(Suppl 4):S11.PubMedCrossRefADS Chin CH, Chen SH, Wu HH, Ho CW, Ko MT, Lin CY. CytoHubba: identifying hub objects and sub-networks from complex interactome. BMC Syst Biol. 2014;8 Suppl 4(Suppl 4):S11.PubMedCrossRefADS
27.
28.
Zurück zum Zitat Davis CA, Hitz BC, Sloan CA, Chan ET, Davidson JM, Gabdank I, Hilton JA, Jain K, Baymuradov UK, Narayanan AK, et al. The Encyclopedia of DNA elements (ENCODE): data portal update. Nucleic Acids Res. 2018;46(D1):D794-d801.PubMedCrossRef Davis CA, Hitz BC, Sloan CA, Chan ET, Davidson JM, Gabdank I, Hilton JA, Jain K, Baymuradov UK, Narayanan AK, et al. The Encyclopedia of DNA elements (ENCODE): data portal update. Nucleic Acids Res. 2018;46(D1):D794-d801.PubMedCrossRef
29.
Zurück zum Zitat Liu ZP, Wu C, Miao H, Wu H. RegNetwork: an integrated database of transcriptional and post-transcriptional regulatory networks in human and mouse. Database (Oxford). 2015;2015:bav095.PubMedCrossRef Liu ZP, Wu C, Miao H, Wu H. RegNetwork: an integrated database of transcriptional and post-transcriptional regulatory networks in human and mouse. Database (Oxford). 2015;2015:bav095.PubMedCrossRef
30.
Zurück zum Zitat Yoo M, Shin J, Kim J, Ryall KA, Lee K, Lee S, Jeon M, Kang J, Tan AC. DSigDB: drug signatures database for gene set analysis. Bioinformatics. 2015;31(18):3069–71.PubMedPubMedCentralCrossRef Yoo M, Shin J, Kim J, Ryall KA, Lee K, Lee S, Jeon M, Kang J, Tan AC. DSigDB: drug signatures database for gene set analysis. Bioinformatics. 2015;31(18):3069–71.PubMedPubMedCentralCrossRef
31.
Zurück zum Zitat Shu T, Ning W, Wu D, Xu J, Han Q, Huang M, Zou X, Yang Q, Yuan Y, Bie Y, et al. Plasma Proteomics Identify Biomarkers and Pathogenesis of COVID-19. Immunity. 2020;53(5):1108-1122.e1105.PubMedPubMedCentralCrossRef Shu T, Ning W, Wu D, Xu J, Han Q, Huang M, Zou X, Yang Q, Yuan Y, Bie Y, et al. Plasma Proteomics Identify Biomarkers and Pathogenesis of COVID-19. Immunity. 2020;53(5):1108-1122.e1105.PubMedPubMedCentralCrossRef
32.
Zurück zum Zitat Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M, Alizadeh AA. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.PubMedPubMedCentralCrossRef Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M, Alizadeh AA. Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 2015;12(5):453–7.PubMedPubMedCentralCrossRef
33.
Zurück zum Zitat Mashaqi S, Lee-Iannotti J, Rangan P, Celaya MP, Gozal D, Quan SF, Parthasarathy S. Obstructive sleep apnea and COVID-19 clinical outcomes during hospitalization: a cohort study. J Clin Sleep Med. 2021;17(11):2197–204.PubMedPubMedCentralCrossRef Mashaqi S, Lee-Iannotti J, Rangan P, Celaya MP, Gozal D, Quan SF, Parthasarathy S. Obstructive sleep apnea and COVID-19 clinical outcomes during hospitalization: a cohort study. J Clin Sleep Med. 2021;17(11):2197–204.PubMedPubMedCentralCrossRef
34.
Zurück zum Zitat Zhang Y, Zeng T, Chen L, Ding S, Huang T, Cai YD. Identification of COVID-19 Infection-Related Human Genes Based on a Random Walk Model in a Virus-Human Protein Interaction Network. Biomed Res Int. 2020;2020:4256301.PubMedPubMedCentral Zhang Y, Zeng T, Chen L, Ding S, Huang T, Cai YD. Identification of COVID-19 Infection-Related Human Genes Based on a Random Walk Model in a Virus-Human Protein Interaction Network. Biomed Res Int. 2020;2020:4256301.PubMedPubMedCentral
35.
Zurück zum Zitat Yang X, Lian X, Fu C, Wuchty S, Yang S, Zhang Z. HVIDB: a comprehensive database for human-virus protein-protein interactions. Brief Bioinform. 2021;22(2):832–44.PubMedCrossRef Yang X, Lian X, Fu C, Wuchty S, Yang S, Zhang Z. HVIDB: a comprehensive database for human-virus protein-protein interactions. Brief Bioinform. 2021;22(2):832–44.PubMedCrossRef
36.
Zurück zum Zitat Shen B, Yi X, Sun Y, Bi X, Du J, Zhang C, Quan S, Zhang F, Sun R, Qian L, et al. Proteomic and Metabolomic Characterization of COVID-19 Patient Sera. Cell. 2020;182(1):59-72.e15.PubMedPubMedCentralCrossRef Shen B, Yi X, Sun Y, Bi X, Du J, Zhang C, Quan S, Zhang F, Sun R, Qian L, et al. Proteomic and Metabolomic Characterization of COVID-19 Patient Sera. Cell. 2020;182(1):59-72.e15.PubMedPubMedCentralCrossRef
37.
Zurück zum Zitat Zhang YH, Li H, Zeng T, Chen L, Li Z, Huang T, Cai YD. Identifying Transcriptomic Signatures and Rules for SARS-CoV-2 Infection. Front Cell Dev Biol. 2020;8:627302.PubMedCrossRef Zhang YH, Li H, Zeng T, Chen L, Li Z, Huang T, Cai YD. Identifying Transcriptomic Signatures and Rules for SARS-CoV-2 Infection. Front Cell Dev Biol. 2020;8:627302.PubMedCrossRef
38.
Zurück zum Zitat Li Z, Mei Z, Ding S, Chen L, Li H, Feng K, Huang T, Cai YD. Identifying Methylation Signatures and Rules for COVID-19 With Machine Learning Methods. Front Mol Biosci. 2022;9:908080.PubMedPubMedCentralCrossRef Li Z, Mei Z, Ding S, Chen L, Li H, Feng K, Huang T, Cai YD. Identifying Methylation Signatures and Rules for COVID-19 With Machine Learning Methods. Front Mol Biosci. 2022;9:908080.PubMedPubMedCentralCrossRef
39.
Zurück zum Zitat Jackson H, Rivero Calle I, Broderick C, Habgood-Coote D, D’Souza G, Nichols S, Vito O, Gómez-Rial J, Rivero-Velasco C, Rodríguez-Núñez N, et al. Characterisation of the blood RNA host response underpinning severity in COVID-19 patients. Sci Rep. 2022;12(1):12216.PubMedPubMedCentralCrossRefADS Jackson H, Rivero Calle I, Broderick C, Habgood-Coote D, D’Souza G, Nichols S, Vito O, Gómez-Rial J, Rivero-Velasco C, Rodríguez-Núñez N, et al. Characterisation of the blood RNA host response underpinning severity in COVID-19 patients. Sci Rep. 2022;12(1):12216.PubMedPubMedCentralCrossRefADS
40.
Zurück zum Zitat Ryan FJ, Hope CM, Masavuli MG, Lynn MA, Mekonnen ZA, Yeow AEL, Garcia-Valtanen P, Al-Delfi Z, Gummow J, Ferguson C, et al. Long-term perturbation of the peripheral immune system months after SARS-CoV-2 infection. BMC Med. 2022;20(1):26.PubMedPubMedCentralCrossRef Ryan FJ, Hope CM, Masavuli MG, Lynn MA, Mekonnen ZA, Yeow AEL, Garcia-Valtanen P, Al-Delfi Z, Gummow J, Ferguson C, et al. Long-term perturbation of the peripheral immune system months after SARS-CoV-2 infection. BMC Med. 2022;20(1):26.PubMedPubMedCentralCrossRef
41.
Zurück zum Zitat Gómez-Carballa A, Rivero-Calle I, Pardo-Seco J, Gómez-Rial J, Rivero-Velasco C, Rodríguez-Núñez N, Barbeito-Castiñeiras G, Pérez-Freixo H, Cebey-López M, Barral-Arca R, et al. A multi-tissue study of immune gene expression profiling highlights the key role of the nasal epithelium in COVID-19 severity. Environ Res. 2022;210:112890.PubMedPubMedCentralCrossRef Gómez-Carballa A, Rivero-Calle I, Pardo-Seco J, Gómez-Rial J, Rivero-Velasco C, Rodríguez-Núñez N, Barbeito-Castiñeiras G, Pérez-Freixo H, Cebey-López M, Barral-Arca R, et al. A multi-tissue study of immune gene expression profiling highlights the key role of the nasal epithelium in COVID-19 severity. Environ Res. 2022;210:112890.PubMedPubMedCentralCrossRef
42.
Zurück zum Zitat Wang X, Liu Y, Li K, Hao Z: Roles of p53-Mediated Host-Virus Interaction in Coronavirus Infection. Int J Mol Sci. 2023;24(7):6371. Wang X, Liu Y, Li K, Hao Z: Roles of p53-Mediated Host-Virus Interaction in Coronavirus Infection. Int J Mol Sci. 2023;24(7):6371.
43.
Zurück zum Zitat Guo X, Liu Y, Kim JL, Kim EY, Kim EQ, Jansen A, Li K, Chan M, Keenan BT, Conejo-Garcia J, et al. Effect of cyclical intermittent hypoxia on Ad5CMVCre induced solitary lung cancer progression and spontaneous metastases in the KrasG12D+; p53fl/fl; myristolated p110fl/fl ROSA-gfp mouse. PLoS ONE. 2019;14(2):e0212930.PubMedPubMedCentralCrossRef Guo X, Liu Y, Kim JL, Kim EY, Kim EQ, Jansen A, Li K, Chan M, Keenan BT, Conejo-Garcia J, et al. Effect of cyclical intermittent hypoxia on Ad5CMVCre induced solitary lung cancer progression and spontaneous metastases in the KrasG12D+; p53fl/fl; myristolated p110fl/fl ROSA-gfp mouse. PLoS ONE. 2019;14(2):e0212930.PubMedPubMedCentralCrossRef
44.
Zurück zum Zitat Semenza GL, Agani F, Booth G, Forsythe J, Iyer N, Jiang BH, Leung S, Roe R, Wiener C, Yu A. Structural and functional analysis of hypoxia-inducible factor 1. Kidney Int. 1997;51(2):553–5.PubMedCrossRef Semenza GL, Agani F, Booth G, Forsythe J, Iyer N, Jiang BH, Leung S, Roe R, Wiener C, Yu A. Structural and functional analysis of hypoxia-inducible factor 1. Kidney Int. 1997;51(2):553–5.PubMedCrossRef
45.
Zurück zum Zitat Appelberg S, Gupta S, Svensson Akusjärvi S, Ambikan AT, Mikaeloff F, Saccon E, Végvári Á, Benfeitas R, Sperk M, Ståhlberg M, et al. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg Microbes Infect. 2020;9(1):1748–60.PubMedPubMedCentralCrossRef Appelberg S, Gupta S, Svensson Akusjärvi S, Ambikan AT, Mikaeloff F, Saccon E, Végvári Á, Benfeitas R, Sperk M, Ståhlberg M, et al. Dysregulation in Akt/mTOR/HIF-1 signaling identified by proteo-transcriptomics of SARS-CoV-2 infected cells. Emerg Microbes Infect. 2020;9(1):1748–60.PubMedPubMedCentralCrossRef
46.
Zurück zum Zitat Tian M, Liu W, Li X, Zhao P, Shereen MA, Zhu C, Huang S, Liu S, Yu X, Yue M, et al. HIF-1α promotes SARS-CoV-2 infection and aggravates inflammatory responses to COVID-19. Signal Transduct Target Ther. 2021;6(1):308.PubMedPubMedCentralCrossRef Tian M, Liu W, Li X, Zhao P, Shereen MA, Zhu C, Huang S, Liu S, Yu X, Yue M, et al. HIF-1α promotes SARS-CoV-2 infection and aggravates inflammatory responses to COVID-19. Signal Transduct Target Ther. 2021;6(1):308.PubMedPubMedCentralCrossRef
47.
Zurück zum Zitat Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, Frezza C, Bernard NJ, Kelly B, Foley NH, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238–42.PubMedPubMedCentralCrossRefADS Tannahill GM, Curtis AM, Adamik J, Palsson-McDermott EM, McGettrick AF, Goel G, Frezza C, Bernard NJ, Kelly B, Foley NH, et al. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature. 2013;496(7444):238–42.PubMedPubMedCentralCrossRefADS
48.
49.
Zurück zum Zitat Semenza GL. Hypoxia-inducible factor 1 and cardiovascular disease. Annu Rev Physiol. 2014;76:39–56.PubMedCrossRef Semenza GL. Hypoxia-inducible factor 1 and cardiovascular disease. Annu Rev Physiol. 2014;76:39–56.PubMedCrossRef
50.
51.
Zurück zum Zitat Ashok BS, Ajith TA, Sivanesan S. Hypoxia-inducible factors as neuroprotective agent in Alzheimer’s disease. Clin Exp Pharmacol Physiol. 2017;44(3):327–34.PubMedCrossRef Ashok BS, Ajith TA, Sivanesan S. Hypoxia-inducible factors as neuroprotective agent in Alzheimer’s disease. Clin Exp Pharmacol Physiol. 2017;44(3):327–34.PubMedCrossRef
52.
Zurück zum Zitat Zhang L, Li M, Wang Z, Sun P, Wei S, Zhang C, Wu H, Bai H. Cardiovascular Risk After SARS-CoV-2 Infection Is Mediated by IL18/IL18R1/HIF-1 Signaling Pathway Axis. Front Immunol. 2021;12:780804.PubMedCrossRefADS Zhang L, Li M, Wang Z, Sun P, Wei S, Zhang C, Wu H, Bai H. Cardiovascular Risk After SARS-CoV-2 Infection Is Mediated by IL18/IL18R1/HIF-1 Signaling Pathway Axis. Front Immunol. 2021;12:780804.PubMedCrossRefADS
53.
54.
Zurück zum Zitat Drager LF, Yao Q, Hernandez KL, Shin M-K, Bevans-Fonti S, Gay J, Sussan TE, Jun JC, Myers AC, Olivecrona G, et al. Chronic intermittent hypoxia induces atherosclerosis via activation of adipose angiopoietin-like 4. Am J Respir Crit Care Med. 2013;188(2):240–8.PubMedPubMedCentralCrossRef Drager LF, Yao Q, Hernandez KL, Shin M-K, Bevans-Fonti S, Gay J, Sussan TE, Jun JC, Myers AC, Olivecrona G, et al. Chronic intermittent hypoxia induces atherosclerosis via activation of adipose angiopoietin-like 4. Am J Respir Crit Care Med. 2013;188(2):240–8.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Aron-Wisnewsky J, Clement K, Pépin J-L. Nonalcoholic fatty liver disease and obstructive sleep apnea. Metabolism. 2016;65(8):1124–35.PubMedCrossRef Aron-Wisnewsky J, Clement K, Pépin J-L. Nonalcoholic fatty liver disease and obstructive sleep apnea. Metabolism. 2016;65(8):1124–35.PubMedCrossRef
56.
Zurück zum Zitat Cai Z, Luo W, Zhan H, Semenza GL. Hypoxia-inducible factor 1 is required for remote ischemic preconditioning of the heart. Proc Natl Acad Sci U S A. 2013;110(43):17462–7.PubMedPubMedCentralCrossRefADS Cai Z, Luo W, Zhan H, Semenza GL. Hypoxia-inducible factor 1 is required for remote ischemic preconditioning of the heart. Proc Natl Acad Sci U S A. 2013;110(43):17462–7.PubMedPubMedCentralCrossRefADS
57.
Zurück zum Zitat Huang S, Chen P, Shui X, He Y, Wang H, Zheng J, Zhang L, Li J, Xue Y, Chen C, et al. Baicalin attenuates transforming growth factor-β1-induced human pulmonary artery smooth muscle cell proliferation and phenotypic switch by inhibiting hypoxia inducible factor-1α and aryl hydrocarbon receptor expression. J Pharm Pharmacol. 2014;66(10):1469–77.PubMedCrossRef Huang S, Chen P, Shui X, He Y, Wang H, Zheng J, Zhang L, Li J, Xue Y, Chen C, et al. Baicalin attenuates transforming growth factor-β1-induced human pulmonary artery smooth muscle cell proliferation and phenotypic switch by inhibiting hypoxia inducible factor-1α and aryl hydrocarbon receptor expression. J Pharm Pharmacol. 2014;66(10):1469–77.PubMedCrossRef
58.
Zurück zum Zitat Lei W, He Y, Shui X, Li G, Yan G, Zhang Y, Huang S, Chen C, Ding Y. Expression and analyses of the HIF-1 pathway in the lungs of humans with pulmonary arterial hypertension. Mol Med Rep. 2016;14(5):4383–90.PubMedCrossRef Lei W, He Y, Shui X, Li G, Yan G, Zhang Y, Huang S, Chen C, Ding Y. Expression and analyses of the HIF-1 pathway in the lungs of humans with pulmonary arterial hypertension. Mol Med Rep. 2016;14(5):4383–90.PubMedCrossRef
59.
Zurück zum Zitat Zhang Z, Li Z, Wang Y, Wei L, Chen H. Overexpressed long noncoding RNA CPS1-IT alleviates pulmonary arterial hypertension in obstructive sleep apnea by reducing interleukin-1β expression via HIF1 transcriptional activity. J Cell Physiol. 2019;234(11):19715–27.PubMedCrossRef Zhang Z, Li Z, Wang Y, Wei L, Chen H. Overexpressed long noncoding RNA CPS1-IT alleviates pulmonary arterial hypertension in obstructive sleep apnea by reducing interleukin-1β expression via HIF1 transcriptional activity. J Cell Physiol. 2019;234(11):19715–27.PubMedCrossRef
60.
Zurück zum Zitat Xiong M, Zhao Y, Mo H, Yang H, Yue F, Hu K. Intermittent hypoxia increases ROS/HIF-1α ’related oxidative stress and inflammation and worsens bleomycin-induced pulmonary fibrosis in adult male C57BL/6J mice. Int Immunopharmacol. 2021;100:108165.PubMedCrossRef Xiong M, Zhao Y, Mo H, Yang H, Yue F, Hu K. Intermittent hypoxia increases ROS/HIF-1α ’related oxidative stress and inflammation and worsens bleomycin-induced pulmonary fibrosis in adult male C57BL/6J mice. Int Immunopharmacol. 2021;100:108165.PubMedCrossRef
61.
Zurück zum Zitat Wang N, Shi X-F, Khan SA, Wang B, Semenza GL, Prabhakar NR, Nanduri J. Hypoxia-inducible factor-1 mediates pancreatic β-cell dysfunction by intermittent hypoxia. Am J Physiol Cell Physiol. 2020;319(5):C922–32.PubMedPubMedCentralCrossRef Wang N, Shi X-F, Khan SA, Wang B, Semenza GL, Prabhakar NR, Nanduri J. Hypoxia-inducible factor-1 mediates pancreatic β-cell dysfunction by intermittent hypoxia. Am J Physiol Cell Physiol. 2020;319(5):C922–32.PubMedPubMedCentralCrossRef
62.
Zurück zum Zitat Matsuyama T, Kubli SP, Yoshinaga SK, Pfeffer K, Mak TW. An aberrant STAT pathway is central to COVID-19. Cell Death Differ. 2020;27(12):3209–25.PubMedCrossRef Matsuyama T, Kubli SP, Yoshinaga SK, Pfeffer K, Mak TW. An aberrant STAT pathway is central to COVID-19. Cell Death Differ. 2020;27(12):3209–25.PubMedCrossRef
63.
Zurück zum Zitat Wyler E, Mösbauer K, Franke V, Diag A, Gottula LT, Arsiè R, Klironomos F, Koppstein D, Hönzke K, Ayoub S, et al. Transcriptomic profiling of SARS-CoV-2 infected human cell lines identifies HSP90 as target for COVID-19 therapy. iScience. 2021;24(3):102151.PubMedPubMedCentralCrossRefADS Wyler E, Mösbauer K, Franke V, Diag A, Gottula LT, Arsiè R, Klironomos F, Koppstein D, Hönzke K, Ayoub S, et al. Transcriptomic profiling of SARS-CoV-2 infected human cell lines identifies HSP90 as target for COVID-19 therapy. iScience. 2021;24(3):102151.PubMedPubMedCentralCrossRefADS
64.
Zurück zum Zitat Luo S, Zhang X, Xiao X, Luo W, Yang Z, Tang S, Huang W. Exploring Potential Biomarkers and Molecular Mechanisms of Ischemic Cardiomyopathy and COVID-19 Comorbidity Based on Bioinformatics and Systems Biology. Int J Mol Sci. 2023;24(7):6511. Luo S, Zhang X, Xiao X, Luo W, Yang Z, Tang S, Huang W. Exploring Potential Biomarkers and Molecular Mechanisms of Ischemic Cardiomyopathy and COVID-19 Comorbidity Based on Bioinformatics and Systems Biology. Int J Mol Sci. 2023;24(7):6511.
65.
Zurück zum Zitat İn E, Özdemir C, Kaman D, Sökücü SN. Heat Shock Proteins, L-Arginine, and Asymmetric Dimethylarginine Levels in Patients With Obstructive Sleep Apnea Syndrome. Arch Bronconeumol. 2015;51(11):544–50.PubMedCrossRef İn E, Özdemir C, Kaman D, Sökücü SN. Heat Shock Proteins, L-Arginine, and Asymmetric Dimethylarginine Levels in Patients With Obstructive Sleep Apnea Syndrome. Arch Bronconeumol. 2015;51(11):544–50.PubMedCrossRef
66.
Zurück zum Zitat Vallée A, Lecarpentier Y, Vallée J-N. Interplay of Opposing Effects of the WNT/β-Catenin Pathway and PPARγ and Implications for SARS-CoV2 Treatment. Front Immunol. 2021;12:666693.PubMedPubMedCentralCrossRef Vallée A, Lecarpentier Y, Vallée J-N. Interplay of Opposing Effects of the WNT/β-Catenin Pathway and PPARγ and Implications for SARS-CoV2 Treatment. Front Immunol. 2021;12:666693.PubMedPubMedCentralCrossRef
68.
Zurück zum Zitat Tempaku P, D’Almeida V, Silva S, Bittencourt L, Tufik S. 0466 Effect of Obstructive Sleep Apnea and CPAP on Telomere Length and its Associated Mechanisms. Sleep. 2023;46(Supplement_1):A207.CrossRef Tempaku P, D’Almeida V, Silva S, Bittencourt L, Tufik S. 0466 Effect of Obstructive Sleep Apnea and CPAP on Telomere Length and its Associated Mechanisms. Sleep. 2023;46(Supplement_1):A207.CrossRef
69.
Zurück zum Zitat Foresta C, Rocca MS, Di Nisio A. Gender susceptibility to COVID-19: a review of the putative role of sex hormones and X chromosome. J Endocrinol Invest. 2021;44(5):951–6.PubMedCrossRef Foresta C, Rocca MS, Di Nisio A. Gender susceptibility to COVID-19: a review of the putative role of sex hormones and X chromosome. J Endocrinol Invest. 2021;44(5):951–6.PubMedCrossRef
70.
Zurück zum Zitat Boumil RM, Lee JT. Forty years of decoding the silence in X-chromosome inactivation. Hum Mol Genet. 2001;10(20):2225–32.PubMedCrossRef Boumil RM, Lee JT. Forty years of decoding the silence in X-chromosome inactivation. Hum Mol Genet. 2001;10(20):2225–32.PubMedCrossRef
71.
Zurück zum Zitat Klionsky DJ, Eskelinen EL, Deretic V. Autophagosomes, phagosomes, autolysosomes, phagolysosomes, autophagolysosomes... wait, I’m confused. Autophagy. 2014;10(4):549–51.PubMedPubMedCentralCrossRef Klionsky DJ, Eskelinen EL, Deretic V. Autophagosomes, phagosomes, autolysosomes, phagolysosomes, autophagolysosomes... wait, I’m confused. Autophagy. 2014;10(4):549–51.PubMedPubMedCentralCrossRef
72.
Zurück zum Zitat Chen YC, Lin IC, Su MC, Hsu PY, Hsiao CC, Hsu TY, Liou CW, Chen YM, Chin CH, Wang TY, et al. Autophagy impairment in patients with obstructive sleep apnea modulates intermittent hypoxia-induced oxidative stress and cell apoptosis via hypermethylation of the ATG5 gene promoter region. Eur J Med Res. 2023;28(1):82.PubMedPubMedCentralCrossRef Chen YC, Lin IC, Su MC, Hsu PY, Hsiao CC, Hsu TY, Liou CW, Chen YM, Chin CH, Wang TY, et al. Autophagy impairment in patients with obstructive sleep apnea modulates intermittent hypoxia-induced oxidative stress and cell apoptosis via hypermethylation of the ATG5 gene promoter region. Eur J Med Res. 2023;28(1):82.PubMedPubMedCentralCrossRef
73.
Zurück zum Zitat Miao G, Zhao H, Li Y, Ji M, Chen Y, Shi Y, Bi Y, Wang P, Zhang H. ORF3a of the COVID-19 virus SARS-CoV-2 blocks HOPS complex-mediated assembly of the SNARE complex required for autolysosome formation. Dev Cell. 2021;56(4):427-442.e425.PubMedCrossRef Miao G, Zhao H, Li Y, Ji M, Chen Y, Shi Y, Bi Y, Wang P, Zhang H. ORF3a of the COVID-19 virus SARS-CoV-2 blocks HOPS complex-mediated assembly of the SNARE complex required for autolysosome formation. Dev Cell. 2021;56(4):427-442.e425.PubMedCrossRef
74.
Zurück zum Zitat Hou P, Wang X, Wang H, Wang T, Yu Z, Xu C, Zhao Y, Wang W, Zhao Y, Chu F, et al. The ORF7a protein of SARS-CoV-2 initiates autophagy and limits autophagosome-lysosome fusion via degradation of SNAP29 to promote virus replication. Autophagy. 2023;19(2):551–69.PubMedCrossRef Hou P, Wang X, Wang H, Wang T, Yu Z, Xu C, Zhao Y, Wang W, Zhao Y, Chu F, et al. The ORF7a protein of SARS-CoV-2 initiates autophagy and limits autophagosome-lysosome fusion via degradation of SNAP29 to promote virus replication. Autophagy. 2023;19(2):551–69.PubMedCrossRef
75.
Zurück zum Zitat Zhang Y, Sun H, Pei R, Mao B, Zhao Z, Li H, Lin Y, Lu K. The SARS-CoV-2 protein ORF3a inhibits fusion of autophagosomes with lysosomes. Cell discovery. 2021;7(1):31.PubMedPubMedCentralCrossRef Zhang Y, Sun H, Pei R, Mao B, Zhao Z, Li H, Lin Y, Lu K. The SARS-CoV-2 protein ORF3a inhibits fusion of autophagosomes with lysosomes. Cell discovery. 2021;7(1):31.PubMedPubMedCentralCrossRef
76.
Zurück zum Zitat Zhang J, Zhao B. Gesongluobu, Sun Y, Wu Y, Pei W, Ye J, Hui R, Liu L: Angiotensin-converting enzyme gene insertion/deletion (I/D) polymorphism in hypertensive patients with different degrees of obstructive sleep apnea. Hypertens Res. 2000;23(5):407–11.PubMedCrossRef Zhang J, Zhao B. Gesongluobu, Sun Y, Wu Y, Pei W, Ye J, Hui R, Liu L: Angiotensin-converting enzyme gene insertion/deletion (I/D) polymorphism in hypertensive patients with different degrees of obstructive sleep apnea. Hypertens Res. 2000;23(5):407–11.PubMedCrossRef
77.
Zurück zum Zitat Boström KB, Hedner J, Melander O, Grote L, Gullberg B, Råstam L, Groop L, Lindblad U. Interaction between the angiotensin-converting enzyme gene insertion/deletion polymorphism and obstructive sleep apnoea as a mechanism for hypertension. J Hypertens. 2007;25(4):779–83.PubMedCrossRef Boström KB, Hedner J, Melander O, Grote L, Gullberg B, Råstam L, Groop L, Lindblad U. Interaction between the angiotensin-converting enzyme gene insertion/deletion polymorphism and obstructive sleep apnoea as a mechanism for hypertension. J Hypertens. 2007;25(4):779–83.PubMedCrossRef
78.
Zurück zum Zitat Akaishi T. Insertion-and-Deletion Mutations between the Genomes of SARS-CoV, SARS-CoV-2, and Bat Coronavirus RaTG13. Microbiol Spectr. 2022;10(3):e0071622.PubMedCrossRef Akaishi T. Insertion-and-Deletion Mutations between the Genomes of SARS-CoV, SARS-CoV-2, and Bat Coronavirus RaTG13. Microbiol Spectr. 2022;10(3):e0071622.PubMedCrossRef
79.
Zurück zum Zitat Akaishi T, Fujiwara K, Ishii T. Insertion/deletion hotspots in the Nsp2, Nsp 3, S1, and ORF8 genes of SARS-related coronaviruses. BMC ecology and evolution. 2022;22(1):123.PubMedPubMedCentralCrossRef Akaishi T, Fujiwara K, Ishii T. Insertion/deletion hotspots in the Nsp2, Nsp 3, S1, and ORF8 genes of SARS-related coronaviruses. BMC ecology and evolution. 2022;22(1):123.PubMedPubMedCentralCrossRef
82.
Zurück zum Zitat Tuleta I, França CN, Wenzel D, Fleischmann B, Nickenig G, Werner N, Skowasch D. Hypoxia-induced endothelial dysfunction in apolipoprotein E-deficient mice; effects of infliximab and L-glutathione. Atherosclerosis. 2014;236(2):400–10.PubMedCrossRef Tuleta I, França CN, Wenzel D, Fleischmann B, Nickenig G, Werner N, Skowasch D. Hypoxia-induced endothelial dysfunction in apolipoprotein E-deficient mice; effects of infliximab and L-glutathione. Atherosclerosis. 2014;236(2):400–10.PubMedCrossRef
83.
Zurück zum Zitat Jelic S, Padeletti M, Kawut SM, Higgins C, Canfield SM, Onat D, Colombo PC, Basner RC, Factor P, LeJemtel TH. Inflammation, oxidative stress, and repair capacity of the vascular endothelium in obstructive sleep apnea. Circulation. 2008;117(17):2270–8.PubMedPubMedCentralCrossRef Jelic S, Padeletti M, Kawut SM, Higgins C, Canfield SM, Onat D, Colombo PC, Basner RC, Factor P, LeJemtel TH. Inflammation, oxidative stress, and repair capacity of the vascular endothelium in obstructive sleep apnea. Circulation. 2008;117(17):2270–8.PubMedPubMedCentralCrossRef
84.
Zurück zum Zitat Gozal D, Kheirandish-Gozal L. Cardiovascular morbidity in obstructive sleep apnea: oxidative stress, inflammation, and much more. Am J Respir Crit Care Med. 2008;177(4):369–75.PubMedCrossRef Gozal D, Kheirandish-Gozal L. Cardiovascular morbidity in obstructive sleep apnea: oxidative stress, inflammation, and much more. Am J Respir Crit Care Med. 2008;177(4):369–75.PubMedCrossRef
85.
Zurück zum Zitat Lavie L. Oxidative stress in obstructive sleep apnea and intermittent hypoxia–revisited–the bad ugly and good: implications to the heart and brain. Sleep Med Rev. 2015;20:27–45.PubMedCrossRef Lavie L. Oxidative stress in obstructive sleep apnea and intermittent hypoxia–revisited–the bad ugly and good: implications to the heart and brain. Sleep Med Rev. 2015;20:27–45.PubMedCrossRef
86.
Zurück zum Zitat Turkiewicz S, Ditmer M, Sochal M, Białasiewicz P, Strzelecki D, Gabryelska A. Obstructive Sleep Apnea as an Acceleration Trigger of Cellular Senescence Processes through Telomere Shortening. Int J Mol Sci. 2021;22(22):12536. Turkiewicz S, Ditmer M, Sochal M, Białasiewicz P, Strzelecki D, Gabryelska A. Obstructive Sleep Apnea as an Acceleration Trigger of Cellular Senescence Processes through Telomere Shortening. Int J Mol Sci. 2021;22(22):12536.
87.
Zurück zum Zitat Ren H, Ling Y, Cao R, Wang Z, Li Y, Huang T: Early warning of emerging infectious diseases based on multimodal data. Biosafety and health 2023. Ren H, Ling Y, Cao R, Wang Z, Li Y, Huang T: Early warning of emerging infectious diseases based on multimodal data. Biosafety and health 2023.
88.
Zurück zum Zitat Li T, Huang T, Guo C, Wang A, Shi X, Mo X, Lu Q, Sun J, Hui T, Tian G, et al. Genomic variation, origin tracing, and vaccine development of SARS-CoV-2: A systematic review. Innovation (Cambridge (Mass)). 2021;2(2):100116. Li T, Huang T, Guo C, Wang A, Shi X, Mo X, Lu Q, Sun J, Hui T, Tian G, et al. Genomic variation, origin tracing, and vaccine development of SARS-CoV-2: A systematic review. Innovation (Cambridge (Mass)). 2021;2(2):100116.
89.
Zurück zum Zitat Chen TF, Chang YC, Hsiao Y, Lee KH, Hsiao YC, Lin YH, Tu YE, Huang HC, Chen CY, Juan HF. DockCoV2: a drug database against SARS-CoV-2. Nucleic Acids Res. 2021;49(D1):D1152-d1159.PubMedCrossRef Chen TF, Chang YC, Hsiao Y, Lee KH, Hsiao YC, Lin YH, Tu YE, Huang HC, Chen CY, Juan HF. DockCoV2: a drug database against SARS-CoV-2. Nucleic Acids Res. 2021;49(D1):D1152-d1159.PubMedCrossRef
90.
Zurück zum Zitat Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, Sajed T, Johnson D, Li C, Sayeeda Z, et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1):D1074-d1082.PubMedCrossRef Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, Sajed T, Johnson D, Li C, Sayeeda Z, et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1):D1074-d1082.PubMedCrossRef
Metadaten
Titel
Identification of biomarkers and pathways for the SARS-CoV-2 infections in obstructive sleep apnea patients based on machine learning and proteomic analysis
verfasst von
Hong Luo
Jisong Yan
Rui Gong
Dingyu Zhang
Xia Zhou
Xianguang Wang
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
BMC Pulmonary Medicine / Ausgabe 1/2024
Elektronische ISSN: 1471-2466
DOI
https://doi.org/10.1186/s12890-024-02921-1

Weitere Artikel der Ausgabe 1/2024

BMC Pulmonary Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Erhebliches Risiko für Kehlkopfkrebs bei mäßiger Dysplasie

29.05.2024 Larynxkarzinom Nachrichten

Fast ein Viertel der Personen mit mäßig dysplastischen Stimmlippenläsionen entwickelt einen Kehlkopftumor. Solche Personen benötigen daher eine besonders enge ärztliche Überwachung.

Nach Herzinfarkt mit Typ-1-Diabetes schlechtere Karten als mit Typ 2?

29.05.2024 Herzinfarkt Nachrichten

Bei Menschen mit Typ-2-Diabetes sind die Chancen, einen Myokardinfarkt zu überleben, in den letzten 15 Jahren deutlich gestiegen – nicht jedoch bei Betroffenen mit Typ 1.

15% bedauern gewählte Blasenkrebs-Therapie

29.05.2024 Urothelkarzinom Nachrichten

Ob Patienten und Patientinnen mit neu diagnostiziertem Blasenkrebs ein Jahr später Bedauern über die Therapieentscheidung empfinden, wird einer Studie aus England zufolge von der Radikalität und dem Erfolg des Eingriffs beeinflusst.

Costims – das nächste heiße Ding in der Krebstherapie?

28.05.2024 Onkologische Immuntherapie Nachrichten

„Kalte“ Tumoren werden heiß – CD28-kostimulatorische Antikörper sollen dies ermöglichen. Am besten könnten diese in Kombination mit BiTEs und Checkpointhemmern wirken. Erste klinische Studien laufen bereits.

Update Innere Medizin

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.