Next Article in Journal
Detection of Human Papillomavirus in Squamous Lesions of the Conjunctiva Using RNA and DNA In-Situ Hybridization
Next Article in Special Issue
Aggregated Genomic Data as Cohort-Specific Allelic Frequencies can Boost Variants and Genes Prioritization in Non-Solved Cases of Inherited Retinal Dystrophies
Previous Article in Journal
LIN28 Family in Testis: Control of Cell Renewal, Maturation, Fertility and Aging
Previous Article in Special Issue
A Genomic Information Management System for Maintaining Healthy Genomic States and Application of Genomic Big Data in Clinical Research
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Gene Networks of Hyperglycemia, Diabetic Complications, and Human Proteins Targeted by SARS-CoV-2: What Is the Molecular Basis for Comorbidity?

by
Olga V. Saik
1,2,* and
Vadim V. Klimontov
1
1
Laboratory of Endocrinology, Research Institute of Clinical and Experimental Lymphology—Branch of the Institute of Cytology and Genetics, Siberian Branch of Russian Academy of Sciences (RICEL—Branch of ICG SB RAS), Novosibirsk 630060, Russia
2
Laboratory of Computer Proteomics, Federal Research Center Institute of Cytology and Genetics, Siberian Branch of the Russian Academy of Sciences (ICG SB RAS), Novosibirsk 630090, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 7247; https://doi.org/10.3390/ijms23137247
Submission received: 4 June 2022 / Revised: 25 June 2022 / Accepted: 27 June 2022 / Published: 29 June 2022
(This article belongs to the Special Issue Medical Genetics, Genomics and Bioinformatics—2022)

Abstract

:
People with diabetes are more likely to have severe COVID-19 compared to the general population. Moreover, diabetes and COVID-19 demonstrate a certain parallelism in the mechanisms and organ damage. In this work, we applied bioinformatics analysis of associative molecular networks to identify key molecules and pathophysiological processes that determine SARS-CoV-2-induced disorders in patients with diabetes. Using text-mining-based approaches and ANDSystem as a bioinformatics tool, we reconstructed and matched networks related to hyperglycemia, diabetic complications, insulin resistance, and beta cell dysfunction with networks of SARS-CoV-2-targeted proteins. The latter included SARS-CoV-2 entry receptors (ACE2 and DPP4), SARS-CoV-2 entry associated proteases (TMPRSS2, CTSB, and CTSL), and 332 human intracellular proteins interacting with SARS-CoV-2. A number of genes/proteins targeted by SARS-CoV-2 (ACE2, BRD2, COMT, CTSB, CTSL, DNMT1, DPP4, ERP44, F2RL1, GDF15, GPX1, HDAC2, HMOX1, HYOU1, IDE, LOX, NUTF2, PCNT, PLAT, RAB10, RHOA, SCARB1, and SELENOS) were found in the networks of vascular diabetic complications and insulin resistance. According to the Gene Ontology enrichment analysis, the defined molecules are involved in the response to hypoxia, reactive oxygen species metabolism, immune and inflammatory response, regulation of angiogenesis, platelet degranulation, and other processes. The results expand the understanding of the molecular basis of diabetes and COVID-19 comorbidity.

1. Introduction

The coronavirus disease 2019 (COVID-19) pandemic has had a huge impact on morbidity and mortality worldwide. Globally, as of 30 May 2022, there have been 525,467,084 cumulative cases of COVID-19, including 6,285,171 deaths, reported to the WHO [1]. During the outbreak of the epidemic, individuals with diabetes turned out to be one of the most vulnerable cohorts. Though there is no strong evidence that diabetes predisposes to infection with SARS-CoV-2, patients with diabetes demonstrated more severe COVID-19 and higher intensive care unit admission and mortality rates [2,3]. Hyperglycemia has been repeatedly recognized as a risk factor for poor outcomes from COVID-19 in patients with pre-existing diabetes [4,5]. On the other hand, COVID-19 may cause hyperglycemia through the induction of insulin resistance and/or beta cell injury [6,7].
Being completely different in etiology, diabetes and COVID-19 demonstrate a certain parallelism in their mechanisms and organ damage. Specifically, the SARS-CoV-2-induced acute inflammatory response and acute tissue damage that may involve the cardiovascular system, kidneys, and brain correspond to chronic low-grade inflammation, macrovascular disease, chronic kidney disease, neuropathy, and brain changes in diabetes. Hypercoagulability, endothelial dysfunction, oxidative stress, and fibrosis are the common hallmarks of both diseases [8].
In subjects with diabetes, post-COVID-19 syndrome, or long COVID-19, has become a new challenge. The syndrome includes debilitating symptoms and signs that develop during or after an infection consistent with COVID-19, persist for more than 12 weeks, and cannot be explained by an alternative diagnosis [9]. Clinical manifestations of post-COVID-19 syndrome are very diverse, with weakness, general malaise, fatigue, concentration impairment, and breathlessness being the most common symptoms reported [10]. Although the hypothesis that patients with diabetes are more likely to develop post-COVID-19 syndrome had not been confirmed [11], the association between the two disorders indicates that they may be mutually aggravating [12]. Taken into account SARS-CoV-2-induced pathophysiological changes, such as dysregulation of inflammatory and immune response, oxidative stress, hypercoagulability, capillary damage, and tissue hypoxia [13], as well as instability in glycemic control [14], one can assume that long COVID-19 may contribute to the progression of diabetic complications. Identification of the molecular mechanisms of metabolic disorders and organ damage in subjects with diabetes during and after COVID-19 is an urgent task.
According to the Human Protein Atlas [15], human proteins related to SARS-CoV-2 can be divided into three groups: SARS-CoV-2 entry receptors, SARS-CoV-2 entry associated proteases, and intracellular SARS-CoV-2 interacting proteins (https://www.proteinatlas.org/humanproteome/sars-cov-2, accessed on 10 February 2022). The most known SARS-CoV-2 entry receptor is angiotensin-converting enzyme 2 (ACE2) [16]. Dipeptidyl peptidase-4 (DPP4) is also involved in the entry [17]. SARS-CoV-2 entry-associated proteases include transmembrane protease, serine 2 (TMPRSS2) [18], cathepsin B (CTSB) [19], and cathepsin L (CTSL) [20]. A set of 332 human intracellular proteins interacting with SARS-CoV-2 was identified by Gordon et al. [21].
In modern biology and medicine, gene network analysis is considered a useful tool for studying the molecular mechanisms of physiological processes and diseases [22]. Thus, in this study, we matched the molecular networks of hyperglycemia and diabetic complications with the networks of human proteins related to SARS-CoV-2. To build molecular (gene) networks, we used the ANDSystem, a bioinformatics tool that performs text-mining of PubMed/Medline-indexed publications. The ANDSystem reconstructs molecular networks as data graphs with nodes including molecules and edges showing the types of connections between nodes [23,24]. The system can be used for the analysis of molecular mechanisms of human diseases and their associations [25,26]. Recently, we applied this tool to reconstruct and analyze the gene networks of diabetic complications, glucose variability, and hypoglycemia [27,28].
The aim of this study was to find key molecules and biological processes that mediate the development of SARS-CoV-2-induced metabolic disorders and tissue damage in patients with diabetes by matching the gene networks of hyperglycemia, diabetic complications, insulin resistance, and beta cell dysfunction with the networks of human proteins targeted by SARS-CoV-2.

2. Results and Discussion

2.1. Network Associated with Hyperglycemia

In the first step, we updated a previously reconstructed gene network associated with hyperglycemia [27]. The updated network included 430 genes/proteins and 46,855 interactions between them (Figure 1, Table S1).
The functional diversity of the molecules included in the network is presented in Figure 2. Among the nodes of the network, hormones, receptors, enzymes, binding proteins, cytokines, growth factors, cell adhesion molecules, receptors, transcription factors, signal transducers, solute carriers, microRNAs, and other molecules were identified.
The variants of the associations between high glucose (HG) and identified genes are presented in Figure 3. It was shown that hyperglycemia upregulates the expression of 179 genes and downregulates 75 genes. On the other hand, 44 molecules contribute to hyperglycemia development and 54 demonstrate antihyperglycemic activity.
Insulin (INS), interleukin-6 (IL6), tumor protein P53 (TP53), mitogen-activated protein kinase 1 (MAPK1), tumor necrosis factor (TNF), glyceraldehydes-3-phosphate dehydrogenase (GAPDH), epidermal growth factor receptor (EGFR), signal transducer and activator of transcription 3 (STAT3), matrix metalloproteinase-9 (MMP9), and leptin (LEP) genes were the central hubs of the hyperglycemia-associated network with the highest betweenness centrality values (Table S2). The betweenness centrality reflects the involvement of a node (molecule) in signal transduction through a network. This measure is calculated based on the number of the shortest paths connecting all pairs of nodes in the network that go through the analyzed node. The high value of betweenness centrality means that the node is a key player or a “bridge” between different parts of a network [29]. Products of the identified genes with the highest betweenness centrality values participate in the regulation of glucose and lipid metabolism (INS, GAPDH, LEP), cell cycle and apoptosis (TP53, MAPK1, STAT3), immune and inflammatory response (IL6, TNF, MMP9), cell proliferation, differentiation, and survival (INS, LEP, EGFR, STAT3). The role of these molecules in glucose metabolism and hyperglycemia-related biochemical abnormalities have been discussed previously [27,30].
We have also identified the genes with the highest crosstalk specificity (CTS) values. The CTS is calculated as a number of neighbors of a particular node (molecule) in a network divided by the number of all neighbors of the node in the global human gene network of the ANDSystem. A higher CTS value means that a node is closely and specifically related to a studied network [24,26,27,28]. The highest crosstalk specificity (CTS) values (Table S3) were demonstrated by uncoupling protein 2 (UCP2), hydroxisteroid 11-beta dehydrohenase-1 (HSD11B1), interleukin-19 (IL19), fatty acid binding protein 1 (FABP1), pyroglutamylated RFamide peptide (QRFP), resistin (RETN), solute carrier family 2 member 2 (SLC2A2), leptin receptor overlapping transport (LEPROT), endothelial cell-specific molecule 1 (ESM1), and cholecystokinin (CCK) genes.
Mitochondrial uncoupling protein 2 (UCP2) is a member of the mitochondrial anion carrier proteins family. It is involved in the separation of oxidative phosphorylation from ATP synthesis with heat release. Hyperglycemia was shown to increase UCP2 levels [31,32] that could be a compensatory effect to the elevated reactive oxygen species generation [33]. UCP2 was shown to negatively regulate insulin secretion. It was proposed as a key factor in beta cell glucose sensing, and a critical interlayer connecting obesity, beta cell dysfunction, and type 2 diabetes [34]. Hydroxysteroid 11-beta dehydrogenase 1 (HSD11B1) catalyzes the bidirectional reaction of the conversion of cortisol to cortisone. The activity of this enzyme is impaired in hyperglycemic conditions [35]. Interleukin-19 (IL19) is a cytokine that can bind the IL20 receptor complex and lead to STAT3 activation. It is also involved in apoptosis induction and inflammatory response. Long-term hyperglycemia may increase IL19 expression in endothelial cells, resulting in local inflammation and accelerated endothelial damage [36]. Fatty acid-binding protein 1 (FABP1) binds long-chain fatty acids, bile acids, and other hydrophobic ligands; it is involved in fatty acid uptake, transport, and metabolism. The expression of FABP1 increases during hyperglycemia [37]. Pyroglutamylated RFamide peptide (QRFP) is a precursor of the members of the RFamide neuropeptides family, some of which are able to regulate blood pressure, reproduction, and food intake. The 26RFa product of QRFP reduces glucose-induced hyperglycemia and increases insulin sensitivity and insulin levels [38]. Resistin (RETN) is an adipokine that reduces insulin sensitivity, enhances hepatic gluconeogenesis, and increases lipolysis and serum-free fatty acid levels [39]. Solute carrier family 2 member 2 (SLC2A2) is an integral plasma membrane glycoprotein of the liver, islet beta cells, intestine, and kidney epithelium that mediates facilitated bidirectional glucose transport. It is also discussed as a glucose sensor with low glucose affinity. The SLC2A2 level is increased in hyperglycemia and decreased in hyperinsulinemia [40]. Leptin receptor overlapping transcript (LEPROT) is involved in the expression of growth hormone and leptin receptors, which are associated with hyperglycemia [41]. Endothelial cell-specific molecule 1 (ESM1) is regulated by cytokines and could have a role in the endothelial dysfunction [42]. Cholecystokinin (CCK) is further processed to multiple protein products such as cholecystokinin-8, -12, -33 peptide hormones, which are able to regulate gastric acid secretion and food intake. Cholecystokinin regulates postprandial hyperglycemia [43]; hyperglycemia, in its turn, could influence the satiating effect of cholecystokinin [44].
The Gene Ontology (GO) enrichment analysis performed by DAVID [45] revealed glucose homeostasis, inflammatory response, response to hypoxia, regulation of cell proliferation, angiogenesis and apoptosis, aging, and response to drugs among the most overrepresented biological processes associated with hyperglycemia (Table 1, Table S4).
These data demonstrate that hyperglycemia is associated with the upregulation or downregulation of a large number of genes whose products are involved in a wide range of physiological and pathophysiological processes. This may lead to an abnormal response to other stressor factors, including a viral infection.

2.2. Networks Associated with Diabetic Complications, Insulin Resistance, and Beta-Cell Dysfunction

In this work, we operated on gene networks associated with cardiovascular disease (CVD), diabetic nephropathy, diabetic retinopathy, and diabetic neuropathy that have been reconstructed and described previously [27,28]. These networks include 494, 685, 424, and 130 genes/proteins, respectively (Table S5).
Gene networks associated with insulin resistance and beta-cell dysfunction were built using the ANDSystem. These networks contain 1452 and 72 genes/proteins, respectively (Tables S6 and S7).

2.3. Networks of Human Proteins Related to SARS-CoV-2

2.3.1. SARS-CoV-2 Entry Receptors

ACE2-Related Network

ACE2, a carboxypeptidase, mediates vasodilation by cleaving angiotensin II and takes part in the negative regulation of the renin–angiotensin system. It was postulated that ACE2 is the main target of SARS-CoV-2 that supports its entrance into the human cells [16]. Interestingly, the loss of ACE2 in mice results in alterations in glucose tolerance and reduces the first phase of insulin secretion [46]. In db/db mice, the progression of type 2 diabetes was accompanied by ACE2 depletion; ACE2 restoration improved glycemia [47].
According to the ANDSystem, ACE2 directly interacts with 147 genes/proteins in the global human network (Table S8). Among them, 34 are participants of the hyperglycemia network (Table 2). The enrichment of the hyperglycemia network with genes/proteins interacting with ACE2 was statistically significant (p-value < 10−24).
It could be assumed that the interaction between SARS-CoV-2 and ACE2 disrupts the function of ACE2 and activity of ACE2-interacting molecules (Table 2). Among these molecules, of greatest interest are those that are upregulated by HG and downregulated by ACE2. This group includes angiopoietin-2 (ANGPT2), monocyte chemoattractant protein 1 (CCL2), connective tissue growth factor (CCN2), high-mobility group protein B1 (HMGB1), inter-cellular adhesion molecule 1 (ICAM1), vascular cell adhesion molecules 1 (VCAM1), miRNA-21 (MIR21), matrix metalloproteinase-9 (MMP9), and signal transducer and activator of transcription 3 (STAT3). As the binding of viral particles to ACE2 could lead to attenuation of ACE2’s ability to downregulate these genes/proteins, the upregulation can be assumed. In diabetes, HG can also upregulate the expression of these genes. This double effect can significantly activate the synthesis of the products of these genes, creating a background for comorbidity.
Some clinical evidence supports this assumption. It was shown that angiopoietin-2 levels were increased in COVID-19 patients and demonstrated relations with the disease severity, hypercoagulation, and mortality [48,49]. Monocyte chemoattractant protein 1 was also linked to COVID-19 severity; it was upregulated during the early phase of SARS-CoV-2 infection and increased further at the late stages in fatal cases [50]. Connective tissue growth factor is considered a fibrotic biomarker [51]. The serum levels of ICAM-1 and VCAM-1 were elevated in patients with COVID-19, especially in severe cases; the molecules demonstrated relations with coagulation disorders [52]. Enhanced ICAM-1 concentration was an independent predictor of mortality in COVID-19 [53]. The fibrosis-associated miRNA-21 was increased in the acute phase of COVID-19 infection and its upregulation turned out to be a predictor of chronic myocardial damage and inflammation in COVID-19 survivors [54]. The levels of MMP-9 were higher in COVID-19 patients and were considered an early indicator of respiratory failure and mortality [55,56]. It was supposed that the hyperactivation of STAT3 participates in the induction of a cytokine storm, the suppression of the antivirus interferon response, M2 macrophage polarization, and lung fibrosis and thrombosis in COVID-19 [57]. The HMGB-1 is thought to initiate inflammation in COVID-19 patients by triggering TLR4 pathway [58]; its serum level is elevated in severe COVID-19 cases [59]. It is also important to mention that HMGB1 itself is able to induce the expression of ACE2 in alveolar epithelial cells [58,59], forming a positive feedback loop in a gene network and amplifying the pathological signals during COVID-19 and hyperglycemia (Figure 4). Accordingly, HMGB1 inhibitors were discussed as promising candidates for the treatment of COVID-19 [59].
Among other components of the network, sirtuin 1 (SIRT1), angiotensin II receptor type 1 (AGTR1), apolipoprotein E (APOE), and ACE (ACE) are also worth mentioning. Hyperglycemia is known to induce the downregulation of SIRT1 [60]; in turn, sirtuin 1 downregulates ACE2 expression [61]. Thus, it could be suggested that hyperglycemia can induce ACE2 by blocking its repressor, leading to the more effective entrance of viral particles into the cells. Indeed, a deficiency of SIRT1 was linked with the hyperinflammatory response and increased mortality in COVID-19 [62,63]. It was shown that AGTR1 is normally downregulated by ACE2 [64] and possesses hyperglycemic activity [65]. The A/A genotype of rs5183 SNP in the AGTR1 gene was associated with higher hospitalization risk in patients with COVID-19 and comorbidities [66]. The apolipoprotein E ε4 allele (APOE4) was associated with ACE2 reduction [67] and blood glucose level [68]. It was linked to increased susceptibility to SARS-CoV-2 infection, severe COVID-19 course, post-COVID mental fatigue, and COVID-19 mortality [69,70]. ACE is able to downregulate ACE2 [71] and, in turn, it is downregulated by ACE2 [72]. This reciprocal regulation constitutes a loop in the gene network that modulates the balance between vasoconstriction and vasodilation. ACE rs4646994 SNP was shown to increase the risk of COVID-19 infection [73].
ACE2 is known as a critical participant in cardiovascular homeostasis and its altered expression is associated with CVD [74]. The inhibition of ACE2 accelerates diabetic kidney injury and renal ACE2 is downregulated in diabetic nephropathy [75]. The loss of ACE2 aggravates diabetic retinopathy by promoting bone marrow dysfunction [76]. The absence of ACE2 resulted in exaggerated glucose intolerance with insulin resistance [77].
The genes/proteins interacting with ACE2 were found in the gene networks of diabetes complications: there were 44 genes/proteins in the CVD network, 9 in the diabetic neuropathy network, 51 in the diabetic nephropathy network, 40 in the diabetic retinopathy network, 75 in the insulin resistance network, and 4 in the beta-cell dysfunction network. All of these networks were enriched by ACE2-interacting genes/proteins with statistically significant p-values less than 10−34, 10−6, 10−36, 10−32, 10−45, and 0.002, respectively.
According to the GO enrichment analysis, these genes are involved in the regulation of cell migration, gene expression, cell proliferation, phosphatidylinositol 3-kinase signaling, apoptosis, response to hypoxia and lipopolysaccharide, nitric oxide signaling, and the regulation of vascular endothelial cell proliferation (Table 3). Moreover, there were inflammatory response, blood vessel remodeling, angiogenesis, regulation of vascular tone and blood pressure, fatty acid and glucose homeostasis, and aging (Table S9).

DPP4-Related Network

DPP4 is an enzyme involved in glucose and insulin metabolism, as well as in immune regulation. It is thought to be a functional receptor of human coronavirus; it can directly bind with the S protein of SARS-CoV-2 [17]. In the global human gene network of the ANDSystem, DPP4 is linked with 251 genes/proteins (Table S10), and 48 of them are also involved in the hyperglycemia network. The enrichment of the hyperglycemia network with genes/proteins linked with DPP4 was statistically significant (p-value < 10−30).
The types of associations of the genes/proteins from hyperglycemia-related and DPP4-related networks with HG and DPP4 are presented in Table 4. Some identified genes, including CCL11, FGF2, HMGB1, and MMP9, are upregulated by HG [78,79,80,81] and downregulated by DPP4 [82,83,84,85]. In COVID-19, the ability of DPP4 to downregulate these genes could be attenuated. In COVID-19 patients, including those with long COVID with cognitive symptoms, the level of eotaxin-1 encoded by CCL11 was significantly increased [86,87]. Intradermal administration of eotaxin-1 upregulated DPP4 in rats [88], constituting a regulatory loop. The elevation of the serum level of fibroblast growth factor 2 (FGF2) in COVID-19 patients was closely associated with disease severity and admission to an intensive care unit [89]. The clinical significance of MMP-9 and HMGB1 in COVID-19 was mentioned above [55,56,59].
Tumor necrosis factor (TNF) and peroxisome proliferator-activated receptor gamma (PPARγ) could be important players in the hyperglycemia–COVID-19 relationship (Figure 5). Hyperglycemia induces the overproduction of TNF [90] and circulating plasma DPP4 levels are significantly upregulated by this factor [91]. The cytokine storm in COVID-19, associated with the severity of the disease, is characterized by the increase in TNF production; TNF is upregulated in acute lung injury and facilitates SARS-CoV-2 interaction with ACE2. Accordingly, TNF inhibitors were discussed as a therapeutic strategy in severe COVID-19 [92].
PPAR-γ has therapeutic potential against hyperglycemia [93], and its expression is increased by DPP4 [94]. In lung biopsies from patients with COVID-19, the gene enrichment patterns were similar to that of PPARG-knockout macrophages. There was a relation between the disease severity and reduced expression of several members of the PPARγ complex [95]. If DPP4 function is reduced by viral expansion, the expression of PPARG could be lowered, promoting insulin resistance and hyperglycemia.
DPP4 and the genes/proteins interacting with it were also found in the analyzed gene networks of diabetes complications. Sixty genes/proteins were identified in the CVD network, 23 in the network of diabetic neuropathy, 79 in the diabetic nephropathy network, 56 in the diabetic retinopathy network, 126 in the insulin resistance network, and 6 in the beta-cell dysfunction network. All of these networks were enriched by DPP4-interacting genes/proteins with statistically significant p-values less than 10−42, 10−19, 10−54, 10−40, 10−76, and 0.0003, respectively.
The GO enrichment analysis revealed the response to hypoxia, regulation of ERK1 and ERK2 cascade, phosphatidylinositol 3-kinase signaling, interleukin-8 production, lipid storage and smooth muscle cell proliferation, aging, cellular response to lipopolysaccharide, and acute-phase response among principal biological processes regulated by the genes linked to DPP4, hyperglycemia, and diabetic complications (Table 5). The regulation of insulin secretion, glucose homeostasis, regulation of MAPK cascade, vasodilation, inflammatory response, and regulation of cytokine production added to the list of overrepresented processes (Table S11).

2.3.2. SARS-CoV-2 Entry-Associated Protease Receptors

TMPRSS2-Related Network

TMPRSS2, a serine protease, is involved in SARS-CoV-2 host cells entry by S protein priming [18]. In the ANDSystem global human gene network, TMPRSS2 was linked to 52 genes/proteins (Table S12). Among these molecules, the androgen receptor (AR) was the only one that was also present in the hyperglycemia network. It was shown that high glucose downregulates AR mRNA and protein levels in LNCaP cells through NF-κB activation [96]. In turn, AR stimulates TMPRSS2 expression [97], facilitating the SARS-CoV-2 entry (Figure 6). It was postulated that the sex differences in COVID-19 severity could be related to androgen sensitivity [98].
Some genes/proteins linked with TMPRSS2 were also present in the networks of diabetic complications and diabetes-related impaired insulin sensitivity and insulin secretion. Only 3 genes/proteins were found in the CVD network, 4 in the network of diabetic neuropathy, 10 in the diabetic nephropathy network, 6 in the diabetic retinopathy network, 12 in the insulin resistance network, and 1 in the network of beta-cell dysfunction. Except for the gene networks of CVD and beta-cell dysfunction, there was some enrichment of the analyzed networks with the TMPRSS2-interacting genes/proteins with p-values less than 0.0004 for diabetic neuropathy, 10−5 for diabetic nephropathy, 0.0008 for diabetic retinopathy, and 0.0003 for the insulin resistance network.
According to the obtained results, the role of TMPRSS2 in the crosstalk between diabetes-related metabolic disorders, diabetic complications, and COVID-19 seems to be modest.

CTSB-Related Network

Cathepsin B (CTSB), a cysteine protease, facilitates the entry of SARS-CoV-2 into the target host cells by the activation of the viral surface protein S [19]. CTSB was directly linked to 329 genes/proteins in the global human network reconstructed by the ANDSystem (Table S13). Among these molecules, 48 were the components of the hyperglycemia-related network (Table 6). The enrichment of hyperglycemia network with genes/proteins interacting with CTSB was statistically significant (p-value < 10−25).
The associations of the gene/proteins from hyperglycemia-related and cathepsin B (CTSB)-related networks with HG and cathepsin B are presented in Table 6. As shown in Figure 7, the expression of caspase 8 (CASP8), interleukin-6 (IL6), interleukin-8 (CXCL8), Sp1 transcription factor (SP1), toll-like receptor 4 (TLR4), TNF, STAT3, and prolactin (PRL) are upregulated by hyperglycemia [99,100,101,102,103,104,105,106] and are known to induce cathepsin B [107,108,109,110,111,112,113,114]. In COVID-19, the inflammatory response and cell death are triggered via caspase 8 activation [115] and cathepsin B is able to activate this enzyme [116]. Interleukin-8 participates in the signaling axis, determining the severity of COVID-19 [117]. Interleukin-6 was proposed as a biomarker for the development of fatal severe acute respiratory syndrome in COVID-19 [118]. It was reported that prolactin serum levels are increased in COVID-19 patients [119]. Cathepsin B stimulates prolactin release [120]. The excess of prolactin can contribute to hyperglycemia by the reduction of insulin sensitivity [121]. On the other hand, prolactin may reduce the hyperinflammatory status in COVID-19 as it has an anti-inflammatory activity [119]. The Sp1 transcription factor could be linked with cytokine expression and the inflammatory response in COVID-19 via miR-155-5p [122]. STAT3 hyperactivation is related to the cytokine storm in COVID-19 [57]. TLR4 was discussed as a prime regulatory factor associated with the immunity and pathogenesis of SARS-CoV-2 infection [123].
Some other genes/proteins listed in Table 6 have been studied in COVID-19 [124,125,126,127,128,129,130,131,132,133,134,135,136,137,138,139,140,141,142,143,144,145,146,147]. It was shown that serum brain-derived neurotrophic factor (BDNF) is associated with poor prognosis of the disease [124]. The activation of caspase 1 (CASP1) was related to a severe course of COVID-19 [125]. In red blood cells obtained from COVID-19 patients, the levels of caspase-3/7 were elevated [126] and the CASP3 gene was a prognostic marker for COVID-19 severity [127]. The level of interleukin-18 (IL18) was significantly higher in patients with severe COVID-19 than in those with milder disease [128]. A dramatic and early rise in IL-10 was observed in severe SARS-CoV-2 infection [129]. It was reported that a TGFB1-related chronic immune response is induced in severe COVID-19 [130]. The levels of anti-ANXA2 antibodies predicted mortality among hospitalized COVID-19 patients [131,132].
It was found that cathepsin B participates in the conversion of proinsulin to insulin. It is also involved in some diabetic complications, including CVD [148,149]. The down-regulation of CTSB suppresses autophagy and promotes apoptosis contributing to the development of proliferative diabetic retinopathy [150]. The insulin resistance causes the downregulation of CTSB [151].
The genes/proteins interacting with CTSB were found in the analyzed gene networks: 59 genes/proteins were revealed in the CVD network, 23 in the diabetic neuropathy, 84 in the diabetic nephropathy, 58 in the diabetic retinopathy, 124 in the insulin resistance, and 12 in the beta-cell dysfunction network. All of these networks were enriched by CTSB-interacting genes/proteins with statistically significant p-values (less than 10−32, 10−16, 10−48, 10−35, 10−56, and 10−8 respectively).
According to the GO enrichment analysis, the genes linked with CTSB and incorporated in the discussed networks were involved in the regulation of cell proliferation, gene expression, protein phosphorylation, and interleukin-8 production, protein kinase B and lipopolysaccharide-mediated signaling pathways, response to drug, and apoptosis (Table 7). The insulin secretion, inflammatory response, regulation of cytokine production, and response to hypoxia were also overrepresented (Table S14).

CTSL-Related Network

Cathepsin L, a lysosomal cysteine proteinase encoded by the CTSL gene, was shown to cleave the SARS-CoV-2 spike protein and enhance virus entry. Its circulating level is elevated in SARS-CoV-2 infection and it is positively correlated with the disease course and severity [20]. In the global human network estimated by the ANDSystem, CTSL is directly linked to 212 genes/proteins (Table S15). Among them, 22 molecules were also revealed in the hyperglycemia network (Table 8). The enrichment of the hyperglycemia network with genes/proteins interacting with CTSL was statistically significant (p-value < 10−8).
As shown in Table 8, FGF2, IL6, FOXO1, HPSE, JUN, and MAPK1 are upregulated by hyperglycemia [79,101,152,153,154,155] and can activate the cathepsin L [156,157,158,159,160,161]. For some of these molecules, there is clinical evidence of an association with COVID-19 (Figure 8). Specifically, the levels of fibroblast growth factor 2 (FGF2), interleukin-6 (IL6), and heparanase (HPSE) were associated with COVID-19 disease severity [89,118,162]. The activation of the MAPK1 signaling pathway was involved in cytokine production in SARS-CoV-2 [163]. Cadherin 1 (CDH1) is downregulated by both HG [164] and cathepsin L [165]. It was found that in cells infected by SARS-CoV-2, the expression of CDH1 was significantly lowered [166].
Some data indicate the involvement of cathepsin L in the pathogenesis of diabetic kidney disease [167,168,169]. In proliferative diabetic retinopathy, the protein level of cathepsin L is significantly downregulated [150]. The comparative analysis of the CTSL-related gene network and networks of diabetic complications and metabolic abnormalities showed the presence of 47 CTSL-related genes/proteins in the CVD network, 14 genes/proteins in the diabetic neuropathy network, 47 in diabetic nephropathy, 30 in diabetic retinopathy, 83 in insulin resistance, and 7 in the beta-cell dysfunction network. All analyzed networks were enriched by CTSL-interacting genes/proteins with statistically significant p-values (less than 10−30, 10−9, 10−24, 10−15, 10−39, and 10−4, respectively).
According to the GO enrichment analysis, the identified genes are involved in the regulation of cell proliferation and migration, chemotaxis, gene expression, protein phosphorylation, the MAPK cascade, protein kinase B signaling, lipopolysaccharide-mediated signaling, protein import into the nucleus, and silencing by miRNA (Table 9). Other important processes include angiogenesis, the reactive oxygen species metabolic process, glucose homeostasis, acute-phase response, regulation of vascular endothelial growth factor production, apoptotic process, inflammatory response, and aging (Table S16).

2.3.3. Intracellular Proteins Targeted by SARS-CoV-2

Network of Intracellular Proteins Targeted by SARS-CoV-2

According to Gordon et al. [21], 332 human proteins are targeted by SARS-CoV-2. We reconstructed a gene network for these proteins (Figure 9) and found 1664 interactions within it (Table S17).
Most of the nodes in the network (203 of 332) turned out to be proteins with binding activity. The network included the molecules that bind RNAs, macromolecular complexes, chaperones, enzymes, microtubules, guanosine triphosphate (GTP), and other molecules. There were some proteins with ATPase and GTPase activity, oxidoreductases, metalloendopeptidases, kinases, nucleoporins, and fibrillins (Figure 10).
As expected, viral process and intracellular transport were identified among enriched GO biological processes in which SARS-CoV-2-targeted proteins participate (Table 10). The list of overrepresented processes included protein transport, folding and heterotrimerization, protein targeting to mitochondrion, tRNA and mRNA transport, regulation of the mitotic cell cycle, regulation of cellular response to heat, and others. In addition, we found the regulation of glucose transport among the overrepresented processes.
We identified mov10 RISC complex RNA helicase (MOV10), Golgi reassembly stacking protein 1 (GORASP1), nucleoporin 62 (NUP62), cullin 2 (CUL2), golgin A2 (GOLGA2), OS9 endoplasmic reticulum lectin (OS9), Ras homolog family member A (RHOA), G3BP stress granule assembly factor 1 (G3BP1), RAB7A, member RAS oncogene family (RAB7A), and centrosomal protein 250 (CEP250) as the network components with the highest betweenness centrality values (Table S18). Among them, products of G3BP1, MOV10, RAB7A, and RHOA pose hydrolase activity; CEP250 and NUP62 regulate the protein localization to centrosomes; GOLGA2 and GORASP1 are associated with transport through the Golgi complex; and CUL2 and OS9 are involved in the protein ubiquitination.
The NADH: ubiquinone oxidoreductase complex assembly factor 1 (NDUFAF1), TM2 domain containing 3 (TM2D3), fatty acyl-CoA reductase 2 (FAR2), centrosomal protein 68 (CEP68), golgin A7 (GOLGA7), nucleolar protein 10 (NOL10), nucleoporin 58 (NUP58), centrosomal protein 112 (CEP112), nucleoporin 54 (NUP54), and quiescin sulfhydryl oxidase 2 (QSOX2) genes demonstrated the highest crosstalk specificity values (Table S19). The functions of the molecules encoded by these genes are quite diverse: nucleoporins are responsible for the transport of molecules across the nuclear envelope; fatty acyl-CoA reductase 2 is a fatty acid to fatty alcohols-converting enzyme; quiescin sulfhydryl oxidase 2 is an enzyme catalyzing the oxidation of sulfhydryl groups in peptide and protein thiols to disulfides with the reduction of oxygen to hydrogen peroxide; centrosomal proteins are components of the human centrosomes and are involved in cell division control; NADH: ubiquinone oxidoreductase complex assembly factor 1 is involved in the mitochondrial respiratory chain catalyzing the transfer of electrons from NADH to ubiquinone; TM2 domain-containing 3 regulates the signal cascades of cell death/proliferation; golgin A7 participates in the transport of proteins from the Golgi complex to the cell surface; and finally, nucleolar protein 10 is associated with late ribosomal RNA-processing events and the assembly of ribosomal particles [170].
Therefore, the key players of the network of SARS-CoV-2-targeted proteins are involved in the protein transport, ubiquitination and cleavage, biogenesis of ribosomes, response to reactive oxygen species and mitochondrial respiration and signal cascades of cell death/proliferation.

Comparative Analysis of the Network of Hyperglycemia and Network of Human Proteins Targeted by SARS-CoV-2

At the next step, we performed the mapping and comparative analysis of both reconstructed networks with assessment of network centralization, average number of neighbors, and network density. Among these parameters, the network centralization is a measure of how the nodes with high to low centrality are distributed. The centralization is higher if there are many clustered hubs in a network. The average number of neighbors reflects the overall connectivity of the nodes in a network. The network density describes the proportion of all possible links between nodes that are in fact observed in a network. A high network density measure shows the signal transduction effectiveness in a network [171].
It was revealed that molecules that make up the hyperglycemia-associated network are more tightly interconnected than those in the network of SARS-CoV-2-targeted proteins. The network centralization values were 0.61 and 0.046, average numbers of neighbors 84.764 and 4.322, and network density values 0.1 and 0.007, respectively. The obtained results indicate that the hyperglycemia-associated network represents close interactions between genes/proteins and it could be considered as a single module in the global human gene network. Oppositely, the participants of the network of SARS-CoV-2-targeted proteins seem to be not so tightly connected to each other. This is consistent with the potential of the virus to affect a huge number of cellular and physiological processes [172,173].
The intersection of the two analyzed networks revealed that eight genes (DNMT1, FBN1, GDF15, GPX1, HMOX1, IDE, PLAT, and RHOA) are common for them. The proteins encoded by these genes are very different in functional specialization.
DNA methyltransferase 1 encoded by DNMT1 gene transfers methyl groups to DNA cytosine nucleotides that are responsible for maintaining DNA methylation patterns. Hyperglycemia increases the enzyme levels in retinal endothelial cells [174]. In turn, the changes in the retinal DNA methylation machinery induced by high glucose are involved in mitochondrial damage and persist after normoglycemia is restored, and therefore may be involved in the metabolic memory in diabetes [175]. It was shown that transient hyperglycemia directly upregulated DNMT1 expression, leading to the hypermethylation of angiopoietin-1, long-lasting activation of NF-κB, and endothelial dysfunction [176]. In cultured SARS-CoV-2-infected lung epithelial cells, DNMT1 was downregulated; however, this inhibition was not detected in COVID-19 patient’s lung tissues [177].
The fibrillin 1 gene (FBN1) encodes a preproprotein that further processes to fibrillin-1, an extracellular matrix glycoprotein, and a hormone asprosin. Fibrillin-1 is a structural component of calcium-binding microfibrils of the connective tissue. Asprosin, a fasting-induced glucogenic hormone, is secreted by white adipose tissue and is recruited to the liver, where it stimulates rapid glucose release into the circulation via the G protein-cAMP-PKA pathway. Humans and mice with insulin resistance show dramatically elevated plasma asprosin levels [178]. Under hyperglycemia conditions, the expression of FBN1 is increased in the kidneys and decreased in the heart due to the epigenetic modifications [179]. A decrease in asprosin serum levels has been reported in patients with COVID-19 [180].
Growth differentiation factor 15 (GDF15) is a secreted ligand that binds to various transforming growth factor beta (TGF-β) receptors resulting in SMAD transcription factor activation. In addition to the signaling patterns of TGF-β, it also acts as a pleiotropic cytokine that participates in the response to cellular injury. Some data demonstrate that GDF-15 is involved in the regulation of inflammation, endothelial cell function, insulin sensitivity, and weight gain [181]. In COVID-19, GDF15 levels are associated with the disease severity and progression [182,183].
Glutathione peroxidase 1 (GPX1) is a selenium-dependent antioxidant enzyme essential for cell survival in oxidative stress. The GPX1 expression is induced by hyperglycemia [184]. The increased GPX1 activity in hyperglycemic conditions could be adaptive and aimed at compensating a decrease in the enzyme protein level due to enhanced proteasome degradation [185]. Recent experimental data indicate the dual role of GPX1 in glucose and lipid metabolism: GPX1 overexpression in the beta cells and insulin-responsive tissues lead to metabolic phenotypes similar to type 2 diabetes; meanwhile, Gpx1−/− mice develop insulin-dependent diabetes [186].
Heme oxygenase 1 (HO-1, HMOX1) is a key rate-limiting enzyme in the process of degradation of heme, the iron-containing molecule. HO-1 acts as antioxidant, anti-inflammatory, antiapoptotic and angiogenic factor through its by-products carbon monoxide (CO) and bilirubin, and can affect multiple cellular pathways involved in endothelial dysfunction and oxidative stress [187]. HO-1 demonstrates antiviral activity by interfering with the replication or activation of the interferon pathway [188]. It was shown that quercetin, a HO-1 inducer, reduced SARS-CoV-2 spike protein expression in kidney cell lines [189]. High glucose decreases HMOX1 expression and protein activity in endothelial cells [190]. In turn, the induction of HO-1 alleviated oxidative and inflammatory response and endoplasmic reticulum stress induced by high glucose in cultured endothelial cells [191]. In different diabetic models, upregulating the HO system increases insulin secretion and reduces hyperglycemia. Similarly, CO also enhances insulin production and improves glucose metabolism [192].
Tissue-type plasminogen activator (tPA) encoded by the PLAT gene is a secreted serine protease that converts the proenzyme plasminogen to plasmin, a fibrinolytic enzyme. It is also involved in cell migration and tissue remodeling. The abnormal activity of the enzyme causes the disruptions in fibrinolysis, leading to excessive bleeding or thrombosis. The decreased activity of tPA could be a risk factor for type 2 diabetes [193]; its dysregulation can aggravate adverse cardiovascular events in hyperglycemia [194]. It was estimated that COVID-19 is associated with increased plasma thrombin generation [195]. Plasma tPA is elevated in COVID-19 patients. At the same time, the plasminogen activator inhibitor-1 (PAI-1) level is reduced [196] and is associated with increased mortality [197].
Insulin-degrading enzyme (IDE) is a zinc metallopeptidase that breaks down the intracellular insulin; it is also able to degrade glucagon, amylin, β-amyloid, and bradykinin. Moreover, IDE behaves as a heat shock protein and modulates the ubiquitin–proteasome system. Current data indicate that IDE acts as a regulator of insulin secretion and hepatic insulin sensitivity, and may participate in the crosstalk between the liver and beta cells. There is increasing evidence that improper IDE function, regulation, or trafficking might be involved in the pathogenesis of metabolic diseases [198,199].
Ras homolog family member A (RhoA, RHOA) is a small GTPase that regulates cell shape, attachment, and motility by promoting the reorganization of the actin cytoskeleton. RhoA participates in the regulation of smooth muscle tone and activates many downstream kinases. It was revealed that the RhoA/Rho-kinase pathway plays an important role in endothelial function and is implicated in cardiovascular disease, erectile dysfunction [200], and diabetic nephropathy [201]. Hyperglycemia causes the increase in RHOA expression in smooth muscles [202]. RHOA was identified among the hub genes playing a central role in COVID-19 immunopathogenesis [203].
Recently, Sardar et al. identified HMOX1, DNMT1, PLAT, GDF1, and ITGB1 as hub genes that are involved in the host–virus interactions in SARS-CoV-2 infection [204]. According to our results, three of them (HMOX1, DNMT1, and PLAT) are common for the networks of hyperglycemia and SARS-CoV-2-targeted proteins.
We revealed, by the comparative analysis of two networks, that SARS-CoV-2-targeted proteins directly interact with 381 gene/proteins of the hyperglycemia network, i.e., almost all of them (Table S20). Among these interactions, there were a large number of protein–protein interactions, as well as regulatory relationships that concern the regulation of protein activity, expression, transport, and degradation. The GO enrichment analysis showed the involvement of these genes/proteins in the response to hypoxia, the apoptotic process, inflammatory response, regulation of angiogenesis, nitric oxide-mediated signal transduction, regulation of reactive oxygen species metabolism, response to tumor necrosis factor, immune response, leukocyte migration, platelet degranulation, regulation of endothelial cell functions, and others processes (Table S21). These findings are in consistence with the estimated pathophysiological abnormalities, such as hypercoagulability, endothelial dysfunction, oxidative stress, and dysregulation of the inflammatory and immune response, which characterize acute and long COVID-19 (Figure 11).

Comparative Analysis of the Networks of Diabetic Complications and Network of Human Proteins Targeted by SARS-CoV-2

In this work, we revealed some intersections between genes/proteins associated with diabetes complications and those of SARS-CoV-2-targeted proteins (Figure 12). Seven molecules were found when assessing the CVD network (COMT, GDF15, GPX1, HMOX1, LOX, PLAT, and SELENOS), eight in the network of diabetic nephropathy (DNMT1, F2RL1, GDF15, HDAC2, HMOX1, HYOU1, LOX, and RHOA), three in the network of diabetic retinopathy (HMOX1, NUTF2, and PLAT), and two in the diabetic neuropathy network (HMOX1 and PLAT).
The role of DNMT1, GDF15, GPX1, HMOX1, PLAT, and RHOA were considered in the previous section. We identified HMOX1 as a shared hub for all analyzed networks. This corresponds to the broad biological functions of HO-1. The beneficial effects of HO-1 and its reaction products in diabetic vascular complications include anti-inflammatory, antiproliferative, antiapoptotic, and immunomodulatory activity [205]. It was revealed that polymorphism in the HMOX1 promoter is associated with CVD in subjects with diabetes [206]. The serum levels of HO-1 are reduced in patients with diabetic retinopathy [207]. In mice, HO-1 deficiency contributes to diabetic kidney disease [208]. Accordingly, the induction of the enzyme demonstrated a protective effect in diabetic nephropathy [209]. HO-1 mitigates cytokine storm and lung injury in mouse models of sepsis and may exert antivirus activity [210]. Therefore, it could be speculated that the suppression of HO-1 in hyperglycemia is a promoting mechanism for diabetic complications and a more severe COVID-19 course.
The PLAT gene was found in the networks of hyperglycemia, CVD, diabetic neuropathy, and retinopathy. This is consistent with data on the important role of fibrinolysis disorders in the development of diabetic vascular complications [211] and COVID-19 [212].
We found GDF15 in the networks of CVD and diabetic nephropathy. In type 2 diabetes, GDF15 is associated with both macrovascular and microvascular complications [213,214]. At the same time, GDF-15 is considered an indicator of COVID-19 severity [182,183].
The lysyl oxidase gene (LOX) was also revealed in the networks of CVD and diabetic nephropathy. The enzyme is involved in the crosslinking of collagens and elastin, and is supposed to be involved in the impairment of the elastic component of lungs in COVID-19 [215]. The LOX gene polymorphisms are associated with CVD [216] and it was proposed as a drug target for CVD therapy [217]. An enhanced LOX expression in the kidneys was found in rats with diabetic nephropathy [218].
Selenoprotein S (SELENOS), which was found in the network of CVD, is a transmembrane protein involved in the degradation of misfolded proteins in the endoplasmic reticulum; it is involved in inflammation, oxidative stress, endoplasmic reticulum stress, and glucose metabolism [219]. Selenoprotein S is highly expressed in the blood vessels [220] and is supposed to be a target in diabetic macroangiopathy [219].
Three more genes (F2RL1, HDAC2, and HYOU1) identified in the diabetic nephropathy network deserve to be mentioned. F2R-like trypsin receptor 1, or protease-activated receptor 2 (F2RL1), is a G-protein coupled receptor; it stimulates vascular smooth muscle relaxation, the dilation of blood vessels, and increases blood flow; it is also involved in the inflammatory and immune response. F2R-like trypsin receptor 1 was shown to aggravate diabetic nephropathy progression [221]. SARS-CoV-2 viral protein ORF9c directly interacts with PAR2 with F2R-like trypsin receptor 1; moreover, it was speculated that the activation of protease-activated receptors by proteases plays a role in COVID-19-induced hyperinflammation [222]. Histone Deacetylase 2 (HDAC2) determines the acetylation status of histones and plays an important role in diabetic nephropathy via the excessive accumulation of the extracellular matrix in the kidneys and epithelial-to-mesenchymal transition of renal tubular epithelial cells [223,224]. Hypoxia Upregulated 1 (HYOU1) is a heat shock protein accumulated in the endoplasmic reticulum under hypoxic conditions, which is important for protein folding and secretion in the endoplasmic reticulum and is associated with apoptosis. In patients with diabetic nephropathy, HYOU1 was upregulated in tubular epithelial cells [225].
Nuclear transport factor 2 (NUTF2), identified in the diabetic retinopathy network, is a cytosolic factor that facilitates the transport of the proteins into the nucleus. The level of the factor was lower in patients with diabetic retinopathy; its overexpression showed a protective effect against diabetic retinopathy [226].
The participants of the gene networks of diabetic complications were directly linked to some SARS-CoV-2-targeted proteins (Table S22). It turned out that SARS-CoV-2-targeted proteins had interactions with 415, 583, 339, and 110 genes/proteins in the network of CVD, diabetic nephropathy, diabetic retinopathy, and diabetic neuropathy, respectively. The most overrepresented GO biological processes for these gene sets were cytokine-mediated signaling, response to hypoxia, inflammatory response, regulation of blood pressure and angiogenesis, regulation of cell proliferation, migration and apoptosis, as well as protein kinase B, ERK1 and ERK2, and phosphatidylinositol 3-kinase signaling (Table 11 and Table S23).

Comparative Analysis of the Networks of Insulin Resistance, Beta-Cell Dysfunction, and Human Proteins Targeted by SARS-CoV-2

The gene network associated with insulin resistance contained 1452 genes/proteins (Table S6). The intersection of this network with the network of human proteins targeted by SARS-CoV-2 included 13 genes/proteins: HMOX1, PLAT, GDF15, DNMT1, F2RL1, GPX1, SELENOS, IDE, BRD2, ERP44, PCNT, RAB10, and SCARB1. Among them, HMOX1, PLAT, GDF15, DNMT1, F2RL1, GPX1, SELENOS, and IDE were also involved in the networks of diabetic complications and hyperglycemia.
It was found that the induction or overexpression of HMOX1 improves the insulin sensitivity and glucose tolerance [227,228]. The elevated levels of PLAT and GDF15 were associated with insulin resistance [229,230,231]. The inverse correlation of DNMT1 expression with insulin sensitivity was observed in adipose tissue [232]. The lack of F2rl1 in mice was associated with the protection from the insulin resistance induced by a high-fat diet [233]. The overexpression of GPX1 was shown to cause insulin resistance [234,235]. The expression of SELENOS and a number of SNPs in it were associated with the homeostasis model assessment of insulin resistance [219,236]. The IDF inhibition improves insulin sensitivity [237] and the upregulation of IDE could be used as a treatment for insulin resistance [238].
Bromodomain-containing 2 (BRD2) is a transcriptional regulator that participates in mitosis. BRD2 can induce insulin resistance through the mTOR/Akt signaling pathway and an inflammatory response in adipose tissue [239]. Endoplasmic Reticulum Protein 44 (ERP44) is a pH-regulated chaperone and could participate in protein quality control at the endoplasmic reticulum–Golgi interface. The decreased cellular level of ERP44 is associated with insulin resistance [240]. Pericentrin (PCNT) is an integral component of the pericentriolar material and is involved in the functioning of the centrosomes, cytoskeleton, and cell-cycle progression. Mutations in PCNT are associated with severe insulin resistance and diabetes [241]. RAB10 is a member of the RAS oncogene family and a small GTPase that regulates intracellular vesicle trafficking. The adipose RAB10 is involved in systemic insulin sensitivity, as RAB10 is required for insulin-stimulated GLUT4 translocation to the plasma membrane that is responsible for glucose uptake [242]. Scavenger receptor class B member 1 (SCARB1) is a high-density lipoprotein cholesterol plasma membrane receptor and the polymorphisms in this gene are associated with insulin resistance [243,244].
In the gene network associated with insulin resistance, there were 1163 genes/proteins directly linked with participants of the network of proteins targeted by SARS-CoV-2 (Table S24). The GO enrichment analysis showed that these genes are involved in the cytokine-mediated signaling pathway, apoptotic process, response to inflammation and hypoxia, and other processes (Table S24).
The gene network associated with beta-cell dysfunction included 72 genes/proteins (Table S7). Fifty-four of them demonstrated interactions with the proteins targeted by SARS-CoV-2 (Table S25). These 54 genes/proteins are involved in the GO biological processes related to the cytokine-mediated signaling pathway, apoptotic process, release of cytochrome c from mitochondria, T cell homeostasis, cell proliferation, and others (Table S25). Among the identified genes, TNF and CASP3 were associated with COVID-19-related networks [92,127].

2.4. Discussion

The results of our study indicate that in patients with diabetes, SARS-CoV-2 triggers a cascade of molecular events that can be considered in terms of molecular networks with a number of positive and negative feedback loops, bypasses, and parallel regulatory pathways. In diabetes, HG induces a wide range of changes in the gene expression, forming a pathophysiological basis for an inappropriate response to stressors including SARS-CoV-2. According to our data, the hyperglycemia-related network includes 430 genes/proteins that are involved in the inflammatory pathways, response to hypoxia, regulation of cell proliferation, angiogenesis, apoptosis, and other processes. The virus can induce further disturbances in the biochemical and pathophysiological processes induced by hyperglycemia.
We have shown that the networks of SARS-CoV-2 entry-supporting proteins (ACE2, DPP4, CTSB and CTSL) are significantly enriched with the genes/proteins associated with hyperglycemia. In addition, the molecules forming the networks of human proteins related to SARS-CoV-2 were found to be significantly overrepresented in the gene networks of the diabetes complications (CVD, diabetic neuropathy, diabetic nephropathy, and diabetic retinopathy), as well as in the insulin resistance and beta-cell dysfunction networks. These findings are consistent with clinical data on more severe courses and poorer outcomes of COVID-19 in subjects with diabetes [2,3] and give further support to notion of parallels between COVID-19 and diabetes pathology [8].
The clinical evidence supports the role of some molecules revealed in this work in COVID-19 pathogenesis: ANGPT2 [48,49], CCL2 [50], ICAM1 [52,53], VCAM-1 [52], MIR21 [54], MMP9 [55,56], STAT3 [57], HMGB1 [58,59], SIRT1 [62,63], AGTR1 [66], APOE4 [69,70], ACE [73], CCL11 [86,87], FGF2 [89], TNF [92], PPAR-γ [95], CASP8 [115], IL8 [117], IL6 [118], PRL [119], SP1 [122], TLR4 [123], BDNF [124], CASP1 [125], CASP3 [126,127], IL18 [128], IL10 [129], TGFB1 [130], ANXA2 [131,132], HPSE [162], MAPK1 [163], CDH1 [166], FBN1 [180], GDF15 [182,183], PLAT [195,196,197], RHOA [203], HMOX1 [204], LOX [215], and others.
According to the GO enrichment analysis, the molecules associated with the proteins related to SARS-CoV-2 are involved in the immune and inflammatory response, acute-phase response, interleukin-8 production, oxidative stress, regulation of cytokine production, response to hypoxia, regulation of vascular endothelial cell proliferation, glucose homeostasis, fibrinolysis, extracellular matrix formation, tissue remodeling, apoptosis, regulation of cell proliferation and migration, angiogenesis, aging, gene expression, phosphatidylinositol 3-kinase signaling, protein kinase B signaling, DNA methylation, and protein phosphorylation. These processes could provide a pathophysiological basis for a more severe clinical course of COVID-19 in subjects with diabetes [172,173].

2.5. Study Limitations

Our study is not without limitations. The gene network reconstruction was based on the text-mining of PubMed/Medline-indexed publications only. Therefore, we cannot exclude that some relevant information has been missed or some of the revealed interactions are false-positive. The study is a hypothesis-generating one. The role of some identified genes/proteins as mediators of a more severe clinical course and worse outcomes of COVID-19 in patients with diabetes needs further experimental verification.

3. Materials and Methods

The study design is presented as a flowchart in Figure 13.
The gene networks were automatically reconstructed by the ANDSystem [23,24], version: 22.0118b686_2022 (ICG SB RAS, Novosibirsk, Russia), available online at http://www-bionet.sscc.ru/and/cell/ (accessed on 10 January 2022).
The structural characteristics of the studied networks were analyzed by the ANDSystem function “Statistics” of the “Analysis” section. It was used to find the betweenness centrality coefficients, the network centralization, the average number of neighbors, the network density, and the CTS values. The CTS reflects the degree to which a particular node is specifically involved in the studied network. CTS is calculated as following: CTS = Ki/Mi, where Ki stands for the number of interactions of a particular i-th gene in the analyzed gene network and Mi stands for the number of interactions of this i-th gene in the global human gene network of the ANDSystem [24,26].
The enrichment of the analyzed gene networks by lists of selected genes was assessed according to the hypergeometric distribution by the “hypergeom.sf” function of the “scipy” library of the Python programming language [245].
The GO enrichment analysis for gene sets was performed by the web-tool DAVID, version 6.8 (LHRI, Frederick, MD, USA) [45]. It is available online: https://david.ncifcrf.gov/home.jsp (accessed on 20 February 2022). The used parameters were: organism, “Homo sapiens”; Gene_Ontology, “GOTERM_BP_DIRECT”; the cut-off for the statistical significance was set as p-values with FDR correction lower than 0.05.
The information on the function of the identified genes was obtained from the database GeneCards [170]. It is available online at https://www.genecards.org/ (accessed on 25 February 2022).
The Venn diagram demonstrating the interactions of genes from the gene networks (Figure 11) was made by the BioVenn web application (available at https://www.biovenn.nl/index.php, accessed on 18 February 2022). The Venn diagram showing the interactions of the gene lists associated with the diabetes complications and the proteins targeted by SARS-CoV-2 (Figure 12) was made by the “Bioinformatics & Evolutionary Genomics” resource (available online at http://bioinformatics.psb.ugent.be/webtools/Venn/, accessed on 19 February 2022).

4. Conclusions

In this work, we have demonstrated, for the first time, that the hyperglycemia network and the networks of SARS-CoV-2-targeted proteins have a number of paths that interact with each other. We revealed that SARS-CoV-2-targeted proteins directly regulate physical interactions with 381 gene/proteins of the hyperglycemia network, i.e., almost all of them. The proteins associated with hyperglycemia and targeted by SARS-CoV-2 proteins are involved in glucose homeostasis, fibrinolysis, extracellular matrix formation, cell migration, tissue remodeling, DNA methylation, response to cellular injury, hypoxia, immune response, inflammation, and oxidative stress. We identified HMOX1 as a shared hub for all analyzed networks. The PLAT gene could be a possible hub that links hyperglycemia, COVID-19, and negative cardiovascular events. Most elements of the hyperglycemia-associated network demonstrate protein–protein or regulatory links with the SARS-CoV-2-targeted proteins. The involvement of these interactions in the cytokine network, inflammation and immunity, angiogenesis and response to hypoxia, oxidative stress, apoptosis, and endothelial cell functions seems to form a pathogenic basis for a more a severe course of COVID-19 in subjects with diabetes.
A number of genes/proteins targeted by SARS-CoV-2 (ACE2, BRD2, COMT, CTSB, CTSL, DNMT1, DPP4, ERP44, F2RL1, GDF15, GPX1, HDAC2, HMOX1, HYOU1, IDE, LOX, NUTF2, PCNT, PLAT, RAB10, RHOA, SCARB1, and SELENOS) were found in the networks of vascular diabetic complications and insulin resistance. According to the GO enrichment analysis, the identified molecules are involved in cytokine-mediated signaling, response to hypoxia, inflammatory response, regulation of blood pressure and angiogenesis, regulation of cell proliferation, migration and apoptosis, as well as protein kinase B, ERK1 and ERK2, and phosphatidylinositol 3-kinase signaling, and other processes.
The results obtained contribute to the deeper understanding of the molecular pathophysiology of COVID-19-induced disorders in subjects with diabetes. The functional significance of the identified hub molecules and their potential value as therapeutic targets requires further research.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23137247/s1.

Author Contributions

Conceptualization, O.V.S. and V.V.K.; investigation, O.V.S.; methodology, O.V.S. and V.V.K.; supervision, V.V.K.; visualization, O.V.S.; writing—original draft, O.V.S. and V.V.K.; writing—review and editing, O.V.S. and V.V.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by Russian Science Foundation (grant No. 20-15-00057).

Institutional Review Board Statement

The study was conducted according to the guidelines of the Declaration of Helsinki, and approved by the Ethics Committee of the RICEL—Branch of IC&G SB RAS (protocol No. 158, date of approval 1 June 2020).

Informed Consent Statement

Not applicable.

Data Availability Statement

The data supporting the reported results are available in the Supplementary Materials.

Acknowledgments

We acknowledge Vladimir Ivanisenko (IC&G SB RAS, Novosibirsk, Russia) and Pavel Demenkov (IC&G SB RAS, Novosibirsk, Russia) for the advice concerning bioinformatics approaches valuable for the performed analysis.

Conflicts of Interest

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Abbreviations

ACEAngiotensin-converting enzyme
ARAndrogen receptor
CTSCrosstalk specificity
CTSBCathepsin B
CTSLCathepsin L
CVDCardiovascular disease
DPP-4Dipeptidyl peptidase-4
EGFEpidermal growth factor
ERKExtracellular signal-regulated kinase
GDF15Growth differentiation factor 15
GOGene ontology
GPX1Glutathione peroxidase 1
HGHigh glucose
HMGB1High-mobility group protein B1
HO-1Heme oxygenase 1
IDFInsulin-degrading enzyme
MAPKMitogen-activated protein kinase
PPARγPeroxisome proliferator-activated receptor gamma
RhoARas homolog family member A
TGF-βTransforming growth factor beta
TMPRSS2Transmembrane protease, serine 2
TNFTumor necrosis factor
tPATissue-type plasminogen activator
UCP2Uncoupling protein 2

References

  1. WHO Coronavirus (COVID-19) Dashboard. Available online: https://covid19.who.int/ (accessed on 30 May 2022).
  2. Buscemi, S.; Corleo, D.; Randazzo, C. Risk Factors for COVID-19: Diabetes, Hypertension, and Obesity. Adv. Exp. Med. Biol. 2021, 1353, 115–129. [Google Scholar] [CrossRef] [PubMed]
  3. Hartmann-Boyce, J.; Rees, K.; Perring, J.C.; Kerneis, S.A.; Morris, E.M.; Goyder, C.; Otunla, A.A.; James, O.A.; Syam, N.R.; Seidu, S.; et al. Risks of and from SARS-CoV-2 Infection and COVID-19 in People with Diabetes: A Systematic Review of Reviews. Diabetes Care 2021, 44, 2790–2811. [Google Scholar] [CrossRef] [PubMed]
  4. Prattichizzo, F.; de Candia, P.; Nicolucci, A.; Ceriello, A. Elevated HbA1c levels in pre-COVID-19 infection increases the risk of mortality: A sistematic review and meta-analysis. Diabetes Metab. Res. Rev. 2022, 38, e3476. [Google Scholar] [CrossRef] [PubMed]
  5. Zhu, Z.; Mao, Y.; Chen, G. Predictive value of HbA1c for in-hospital adverse prognosis in COVID-19: A systematic review and meta-analysis. Prim. Care Diabetes 2021, 15, 910–917. [Google Scholar] [CrossRef]
  6. Govender, N.; Khaliq, O.P.; Moodley, J.; Naicker, T. Insulin resistance in COVID-19 and diabetes. Prim. Care Diabetes 2021, 15, 629–634. [Google Scholar] [CrossRef]
  7. Abramczyk, U.; Nowaczyński, M.; Słomczyński, A.; Wojnicz, P.; Zatyka, P.; Kuzan, A. Consequences of COVID-19 for the Pancreas. Int. J. Mol. Sci. 2022, 23, 864. [Google Scholar] [CrossRef]
  8. Feldman, E.L.; Savelieff, M.G.; Hayek, S.S.; Pennathur, S.; Kretzler, M.; Pop-Busui, R. COVID-19 and Diabetes: A Collision and Collusion of Two Diseases. Diabetes 2020, 69, 2549–2565. [Google Scholar] [CrossRef]
  9. National Institute for Health and Care Excellence (NICE). COVID-19 Rapid Guideline: Managing the Long-Term Effects of COVID-19; NICE: London, UK, 2020. [Google Scholar]
  10. Michelen, M.; Manoharan, L.; Elkheir, N.; Cheng, V.; Dagens, A.; Hastie, C.; O’Hara, M.; Suett, J.; Dahmash, D.; Bugaeva, P.; et al. Characterising long COVID: A living systematic review. BMJ Glob. Health 2021, 6, e005427. [Google Scholar] [CrossRef]
  11. Fernández-de-Las-Peñas, C.; Guijarro, C.; Torres-Macho, J.; Velasco-Arribas, M.; Plaza-Canteli, S.; Hernández-Barrera, V.; Arias-Navalón, J.A. Diabetes and the Risk of Long-term Post-COVID Symptoms. Diabetes 2021, 70, 2917–2921. [Google Scholar] [CrossRef]
  12. Raveendran, A.V.; Misra, A. Post COVID-19 Syndrome (“Long COVID”) and Diabetes: Challenges in Diagnosis and Management. Diabetes Metab. Syndr. 2021, 15, 102235. [Google Scholar] [CrossRef]
  13. Østergaard, L. SARS-CoV-2 related microvascular damage and symptoms during and after COVID-19: Consequences of capillary transit-time changes, tissue hypoxia and inflammation. Physiol. Rep. 2021, 9, e14726. [Google Scholar] [CrossRef]
  14. Mohiuddin Chowdhury, A.T.M.; Karim, M.R.; Ali, M.A.; Islam, J.; Li, Y.; He, S. Clinical Characteristics and the Long-Term Post-recovery Manifestations of the COVID-19 Patients-A Prospective Multicenter Cross-Sectional Study. Front. Med. 2021, 8, 663670. [Google Scholar] [CrossRef]
  15. Uhlén, M.; Fagerberg, L.; Hallström, B.M.; Lindskog, C.; Oksvold, P.; Mardinoglu, A.; Sivertsson, Å.; Kampf, C.; Sjöstedt, E.; Asplund, A.; et al. Tissue-based map of the human proteome. Science 2015, 347, 1260419. [Google Scholar] [CrossRef]
  16. Vargas-Rodriguez, J.R.; Garza-Veloz, I.; Flores-Morales, V.; Badillo-Almaraz, J.I.; Rocha-Pizaña, M.R.; Valdés-Aguayo, J.J.; Martinez-Fierro, M.L. Hyperglycemia and Angiotensin-Converting Enzyme 2 in Pulmonary Function in the Context of SARS-CoV-2 Infection. Front. Med. 2022, 8, 758414. [Google Scholar] [CrossRef]
  17. Gao, Q.; Zhang, W.; Li, T.; Yang, G.; Zhu, W.; Chen, N.; Jin, H. Interrelationship between 2019-nCov receptor DPP4 and diabetes mellitus targets based on protein interaction network. Sci. Rep. 2022, 12, 188. [Google Scholar] [CrossRef]
  18. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280. [Google Scholar] [CrossRef]
  19. Padmanabhan, P.; Desikan, R.; Dixit, N.M. Targeting TMPRSS2 and Cathepsin B/L together may be synergistic against SARS-CoV-2 infection. PLoS Comput. Biol. 2020, 16, e1008461. [Google Scholar] [CrossRef]
  20. Zhao, M.M.; Yang, W.L.; Yang, F.Y.; Zhang, L.; Huang, W.J.; Hou, W.; Fan, C.F.; Jin, R.H.; Feng, Y.M.; Wang, Y.C.; et al. Cathepsin L plays a key role in SARS-CoV-2 infection in humans and humanized mice and is a promising target for new drug development. Signal Transduct. Target. Ther. 2021, 6, 134. [Google Scholar] [CrossRef]
  21. Gordon, D.E.; Jang, G.M.; Bouhaddou, M.; Xu, J.; Obernier, K.; White, K.M.; O’Meara, M.J.; Rezelj, V.V.; Guo, J.Z.; Swaney, D.L.; et al. A SARS-CoV-2 protein interaction map reveals targets for drug repurposing. Nature 2020, 583, 459–468. [Google Scholar] [CrossRef]
  22. Farina, L. Network as a language for precision medicine. Ann. Dell’istituto Super. Sanità 2021, 57, 330–342. [Google Scholar]
  23. Ivanisenko, V.A.; Saik, O.V.; Ivanisenko, N.V.; Tiys, E.S.; Ivanisenko, T.V.; Demenkov, P.S.; Kolchanov, N.A. ANDSystem: An Associative Network Discovery System for automated literature mining in the field of biology. BMC Syst. Biol. 2015, 9 (Suppl. S2), S2. [Google Scholar] [CrossRef] [Green Version]
  24. Ivanisenko, V.A.; Demenkov, P.S.; Ivanisenko, T.V.; Mishchenko, E.L.; Saik, O.V. A new version of the ANDSystem tool for automatic extraction of knowledge from scientific publications with expanded functionality for reconstruction of associative gene networks by considering tissue-specific gene expression. BMC Bioinform. 2019, 20 (Suppl. S1), 34. [Google Scholar] [CrossRef]
  25. Saik, O.V.; Demenkov, P.S.; Ivanisenko, T.V.; Bragina, E.Y.; Freidin, M.B.; Goncharova, I.A.; Dosenko, V.E.; Zolotareva, O.I.; Hofestaedt, R.; Lavrik, I.N.; et al. Novel candidate genes important for asthma and hypertension comorbidity revealed from associative gene networks. BMC Med. Genom. 2018, 11 (Suppl. S1), 15. [Google Scholar] [CrossRef] [Green Version]
  26. Saik, O.V.; Nimaev, V.V.; Usmonov, D.B.; Demenkov, P.S.; Ivanisenko, T.V.; Lavrik, I.N.; Ivanisenko, V.A. Prioritization of genes involved in endothelial cell apoptosis by their implication in lymphedema using an analysis of associative gene networks with ANDSystem. BMC Med. Genom. 2019, 12 (Suppl. S2), 47. [Google Scholar] [CrossRef]
  27. Saik, O.V.; Klimontov, V.V. Bioinformatic Reconstruction and Analysis of Gene Networks Related to Glucose Variability in Diabetes and Its Complications. Int. J. Mol. Sci. 2020, 21, 8691. [Google Scholar] [CrossRef]
  28. Saik, O.V.; Klimontov, V.V. Hypoglycemia, Vascular Disease and Cognitive Dysfunction in Diabetes: Insights from Text Mining-Based Reconstruction and Bioinformatics Analysis of the Gene Networks. Int. J. Mol. Sci. 2021, 22, 12419. [Google Scholar] [CrossRef]
  29. Durón, C.; Pan, Y.; Gutmann, D.H.; Hardin, J.; Radunskaya, A. Variability of betweenness centrality and its effect on identifying essential genes. Bull. Math. Biol. 2019, 81, 3655–3673. [Google Scholar] [CrossRef]
  30. Klimontov, V.V.; Saik, O.V.; Korbut, A.I. Glucose variability: How does it work? Int. J. Mol. Sci. 2021, 22, 7783. [Google Scholar] [CrossRef]
  31. Friederich, M.; Olerud, J.; Fasching, A.; Liss, P.; Hansell, P.; Palm, F. Uncoupling protein-2 in diabetic kidneys: Increased protein expression correlates to increased non-transport related oxygen consumption. Adv. Exp. Med. Biol. 2008, 614, 37–43. [Google Scholar] [CrossRef]
  32. Noble, E.G.; Shen, G.X. Impact of exercise and metabolic disorders on heat shock proteins and vascular inflammation. Autoimmune Dis. 2012, 2012, 836519. [Google Scholar] [CrossRef] [Green Version]
  33. Zheng, Z.; Chen, H.; Ke, G.; Fan, Y.; Zou, H.; Sun, X.; Gu, Q.; Xu, X.; Ho, P.C. Protective Effect of Perindopril on Diabetic Retinopathy Is Associated with Decreased Vascular Endothelial Growth Factor–to–Pigment Epithelium–Derived Factor Ratio: Involvement of a Mitochondria–Reactive Oxygen Species Pathway. Diabetes 2009, 58, 954–964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Zhang, C.Y.; Baffy, G.; Perret, P.; Krauss, S.; Peroni, O.; Grujic, D.; Hagen, T.; Vidal-Puig, A.J.; Boss, O.; Kim, Y.B.; et al. Uncoupling protein-2 negatively regulates insulin secretion and is a major link between obesity, β cell dysfunction, and type 2 diabetes. Cell 2001, 105, 745–755. [Google Scholar] [CrossRef] [Green Version]
  35. Andrews, R.C.; Herlihy, O.; Livingstone, D.E.; Andrew, R.; Walker, B.R. Abnormal cortisol metabolism and tissue sensitivity to cortisol in patients with glucose intolerance. J. Clin. Endocrinol. Metab. 2002, 87, 5587–5593. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Li, L.; Jiang, X.G.; Hu, J.Y.; Yu, Z.Q.; Xu, J.Y.; Liu, F.; Zhao, G.C.; Zhang, L.; Gu, H.M.; Zhang, S.J.; et al. The association between interleukin-19 concentration and diabetic nephropathy. BMC Nephrol. 2017, 18, 65. [Google Scholar] [CrossRef] [Green Version]
  37. Ito, H.; Yamashita, H.; Nakashima, M.; Takaki, A.; Yukawa, C.; Matsumoto, S.; Omoto, T.; Shinozaki, M.; Nishio, S.; Abe, M.; et al. Current metabolic status affects urinary liver-type fatty-acid binding protein in normoalbuminuric patients with type 2 diabetes. J. Clin. Med. Res. 2017, 9, 366. [Google Scholar] [CrossRef] [Green Version]
  38. Chartrel, N.; Picot, M.; El Medhi, M.; Arabo, A.; Berrahmoune, H.; Alexandre, D.; Maucotel, J.; Anouar, Y.; Prévost, G. The neuropeptide 26RFa (QRFP) and its role in the regulation of energy homeostasis: A mini-review. Front. Neurosci. 2016, 10, 549. [Google Scholar] [CrossRef]
  39. Kim, J.E.; Kim, J.S.; Jo, M.J.; Cho, E.; Ahn, S.Y.; Kwon, Y.J.; Ko, G.J. The Roles and Associated Mechanisms of Adipokines in Development of Metabolic Syndrome. Molecules 2022, 27, 334. [Google Scholar] [CrossRef]
  40. Amin, M.M.; Asaad, G.F.; Abdel Salam, R.M.; El-Abhar, H.S.; Arbid, M.S. Novel CoQ10 antidiabetic mechanisms underlie its positive effect: Modulation of insulin and adiponectine receptors, Tyrosine kinase, PI3K, glucose transporters, sRAGE and visfatin in insulin resistant/diabetic rats. PLoS ONE 2014, 9, e89169. [Google Scholar] [CrossRef] [Green Version]
  41. Fujikawa, T.; Coppari, R. Hypothalamic-mediated control of glucose balance in the presence and absence of insulin. Aging 2014, 6, 92. [Google Scholar] [CrossRef] [Green Version]
  42. Qiu, C.; Sui, J.; Zhang, Q.; Wei, P.; Wang, P.; Fu, Q. Relationship of endothelial cell–specific molecule 1 level in stress hyperglycemia patients with acute ST-segment elevation myocardial infarction: A pilot study. Angiology 2016, 67, 829–834. [Google Scholar] [CrossRef]
  43. Rushakoff, R.A.; Goldfine, I.D.; Beccaria, L.J.; Mathur, A.; Brand, R.J.; Liddle, R.A. Reduced postprandial cholecystokinin (CCK) secretion in patients with noninsulin-dependent diabetes mellitus: Evidence for a role for CCK in regulating postprandial hyperglycemia. J. Clin. Endocrinol. Metab. 1993, 76, 489–493. [Google Scholar] [CrossRef]
  44. Lam, W.F.; Gielkens, H.A.; De Boer, S.Y.; Lamers, C.B.; Masclee, A.A. Influence of hyperglycemia on the satiating effect of CCK in humans. Physiol. Behav. 1998, 65, 505–511. [Google Scholar] [CrossRef]
  45. Huang, D.W.; Sherman, B.T.; Lempicki, R. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 2008, 4, 44–57. [Google Scholar] [CrossRef]
  46. Batlle, D.; Jose Soler, M.; Ye, M. ACE2 and diabetes: ACE of ACEs? Diabetes 2010, 59, 2994–2996. [Google Scholar] [CrossRef] [Green Version]
  47. Chhabra, K.H.; Chodavarapu, H.; Lazartigues, E. Angiotensin converting enzyme 2: A new important player in the regulation of glycemia. IUBMB Life 2013, 65, 731–738. [Google Scholar] [CrossRef] [Green Version]
  48. Hultstrom, M.; Fromell, K.; Larsson, A.; Quaggin, S.E.; Betsholtz, C.; Frithiof, R.; Lipcsey, M.; Jeansson, M. Elevated Angiopoietin-2 inhibits thrombomodulin-mediated anticoagulation in critically ill COVID-19 patients. medRxiv 2021. [Google Scholar] [CrossRef]
  49. Villa, E.; Critelli, R.; Lasagni, S.; Melegari, A.; Curatolo, A.; Celsa, C.; Romagnoli, D.; Melegari, G.; Pivetti, A.; Di Marco, L.; et al. Dynamic angiopoietin-2 assessment predicts survival and chronic course in hospitalized patients with COVID-19. Blood Adv. 2021, 5, 662–673. [Google Scholar] [CrossRef]
  50. Khalil, B.A.; Elemam, N.M.; Maghazachi, A.A. Chemokines and chemokine receptors during COVID-19 infection. Comput. Struct. Biotechnol. J. 2021, 19, 976–988. [Google Scholar] [CrossRef]
  51. Rayego-Mateos, S.; Morgado-Pascual, J.L.; Lavoz, C.; Rodrigues-Díez, R.R.; Márquez-Expósito, L.; Tejera-Muñoz, A.; Tejedor-Santamaría, L.; Rubio-Soto, I.; Marchant, V.; Ruiz-Ortega, M. CCN2 Binds to Tubular Epithelial Cells in the Kidney. Biomolecules 2022, 12, 252. [Google Scholar] [CrossRef]
  52. Tong, M.; Jiang, Y.; Xia, D.; Xiong, Y.; Zheng, Q.; Chen, F.; Zou, L.; Xiao, W.; Zhu, Y. Elevated expression of serum endothelial cell adhesion molecules in COVID-19 patients. J. Infect. Dis. 2020, 222, 894–898. [Google Scholar] [CrossRef]
  53. Kaur, S.; Hussain, S.; Kolhe, K.; Kumar, G.; Tripathi, D.M.; Tomar, A.; Kale, P.; Narayanan, A.; Bihari, C.; Bajpai, M.; et al. Elevated plasma ICAM1 levels predict 28-day mortality in cirrhotic patients with COVID-19 or bacterial sepsis. JHEP Rep. 2021, 3, 100303. [Google Scholar] [CrossRef]
  54. Garg, A.; Seeliger, B.; Derda, A.A.; Xiao, K.; Gietz, A.; Scherf, K.; Sonnenschein, K.; Pink, I.; Hoeper, M.M.; Welte, T.; et al. Circulating cardiovascular microRNAs in critically ill COVID-19 patients. Eur. J. Heart Fail. 2021, 23, 468–475. [Google Scholar] [CrossRef]
  55. Ueland, T.; Holter, J.C.; Holten, A.R.; Müller, K.E.; Lind, A.; Bekken, G.K.; Dudman, S.; Aukrust, P.; Dyrhol-Riise, A.M.; Heggelund, L. Distinct and early increase in circulating MMP-9 in COVID-19 patients with respiratory failure. J. Infect. 2020, 81, e41–e43. [Google Scholar] [CrossRef]
  56. Carolina, D.; Couto, A.E.; Campos, L.C.; Vasconcelos, T.F.; Michelon-Barbosa, J.; Corsi, C.A.; Mestriner, F.; Petroski-Moraes, B.C.; Garbellini-Diab, M.J.; Couto, D.M.; et al. MMP-2 and MMP-9 levels in plasma are altered and associated with mortality in COVID-19 patients. Biomed. Pharmacother. 2021, 142, 112067. [Google Scholar] [CrossRef]
  57. Jafarzadeh, A.; Nemati, M.; Jafarzadeh, S. Contribution of STAT3 to the pathogenesis of COVID-19. Microb. Pathog. 2021, 154, 104836. [Google Scholar] [CrossRef]
  58. Sivakorn, C.; Dechsanga, J.; Jamjumrus, L.; Boonnak, K.; Schultz, M.J.; Dondorp, A.M.; Phumratanaprapin, W.; Ratanarat, R.; Naorungroj, T.; Wattanawinitchai, P.; et al. High mobility group box 1 and interleukin 6 at intensive care unit admission as biomarkers in critically ill COVID-19 Patients. Am. J. Trop. Med. Hyg. 2021, 105, 73. [Google Scholar] [CrossRef]
  59. Chen, R.; Huang, Y.; Quan, J.; Liu, J.; Wang, H.; Billiar, T.R.; Lotze, M.T.; Zeh, H.J.; Kang, R.; Tang, D. HMGB1 as a potential biomarker and therapeutic target for severe COVID-19. Heliyon 2020, 6, e05672. [Google Scholar] [CrossRef]
  60. Ghoshal, K.; Bhattacharyya, M. Overview of platelet physiology: Its hemostatic and nonhemostatic role in disease pathogenesis. Sci. World J. 2014, 2014, 781857. [Google Scholar] [CrossRef] [Green Version]
  61. Nunes-Souza, V.; Alenina, N.; Qadri, F.; Penninger, J.M.; Santos, R.A.S.; Bader, M.; Rabelo, L.A. CD36/sirtuin 1 axis impairment contributes to hepatic steatosis in ACE2-deficient mice. Oxid. Med. Cell. Longev. 2016, 2016, 6487509. [Google Scholar] [CrossRef]
  62. Miller, R.; Wentzel, A.R.; Richards, G.A. COVID-19: NAD+ deficiency may predispose the aged, obese and type2 diabetics to mortality through its effect on SIRT1 activity. Med. Hypotheses 2020, 144, 110044. [Google Scholar] [CrossRef]
  63. Bordoni, V.; Tartaglia, E.; Sacchi, A.; Fimia, G.M.; Cimini, E.; Casetti, R.; Notari, S.; Grassi, G.; Marchioni, L.; Bibas, M.; et al. The unbalanced p53/SIRT1 axis may impact lymphocyte homeostasis in COVID-19 patients. Int. J. Infect. Dis. 2021, 105, 49–53. [Google Scholar] [CrossRef] [PubMed]
  64. Sriramula, S.; Cardinale, J.P.; Lazartigues, E.; Francis, J. ACE2 overexpression in the paraventricular nucleus attenuates angiotensin II-induced hypertension. Cardiovasc. Res. 2011, 92, 401–408. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Hashikawa-Hobara, N.; Hashikawa, N.; Inoue, Y.; Sanda, H.; Zamami, Y.; Takatori, S.; Kawasaki, H. Candesartan cilexetil improves angiotensin II type 2 receptor–mediated neurite outgrowth via the PI3K-Akt pathway in fructose-induced insulin-resistant rats. Diabetes 2012, 61, 925–932. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Sabater Molina, M.; Nicolás Rocamora, E.; Bendicho, A.I.; Vázquez, E.G.; Zorio, E.; Rodriguez, F.D.; Gil Ortuño, C.; Rodríguez, A.I.; Sánchez-López, A.J.; Jara Rubio, R.; et al. Polymorphisms in ACE, ACE2, AGTR1 genes and severity of COVID-19 disease. PLoS ONE 2022, 17, e0263140. [Google Scholar] [CrossRef] [PubMed]
  67. Kehoe, P.G.; Wong, S.; Al Mulhim, N.; Palmer, L.E.; Miners, J.S. Angiotensin-converting enzyme 2 is reduced in Alzheimer’s disease in association with increasing amyloid-β and tau pathology. Alzheimer’s Res. Ther. 2016, 8, 50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Ban, C.X.; Zhong, L.; Wang, T.; Zhu, M.J.; Wang, J.H.; Zhang, Z.L.; Wang, Z.; Su, N.; Liu, Y.Y.; Shi, Y.C.; et al. Enhanced diabetes susceptibility in community dwelling Han elders carrying the Apolipoprotein E 3/3 genotype. PLoS ONE 2016, 11, e0151336. [Google Scholar] [CrossRef]
  69. Kurki, S.N.; Kantonen, J.; Kaivola, K.; Hokkanen, L.; Mäyränpää, M.I.; Puttonen, H.; Martola, J.; Pöyhönen, M.; Kero, M.; Tuimala, J.; et al. APOE ε4 associates with increased risk of severe COVID-19, cerebral microhaemorrhages and post-COVID mental fatigue: A Finnish biobank, autopsy and clinical study. Acta Neuropathol. Commun. 2021, 9, 199. [Google Scholar] [CrossRef]
  70. Xiong, N.; Schiller, M.R.; Li, J.; Chen, X.; Lin, Z. Severe COVID-19 in Alzheimer’s disease: APOE4’s fault again? Alzheimer’s Res. Ther. 2021, 13, 111. [Google Scholar] [CrossRef]
  71. Velkoska, E.; Dean, R.G.; Burchill, L.; Levidiotis, V.; Burrell, L.M. Reduction in renal ACE2 expression in subtotal nephrectomy in rats is ameliorated with ACE inhibition. Clin. Sci. 2010, 118, 269–279. [Google Scholar] [CrossRef] [Green Version]
  72. Burrell, L.M.; Johnston, C.I.; Tikellis, C.; Cooper, M.E. ACE2, a new regulator of the renin–angiotensin system. Trends Endocrinol. Metab. 2004, 15, 166–169. [Google Scholar] [CrossRef]
  73. Kouhpayeh, H.R.; Tabasi, F.; Dehvari, M.; Naderi, M.; Bahari, G.; Khalili, T.; Clark, C.; Ghavami, S.; Taheri, M. Association between angiotensinogen (AGT), angiotensin-converting enzyme (ACE) and angiotensin-II receptor 1 (AGTR1) polymorphisms and COVID-19 infection in the southeast of Iran: A preliminary case-control study. Transl. Med. Commun. 2021, 6, 26. [Google Scholar] [CrossRef]
  74. Raizada, M.K.; Ferreira, A.J. ACE2: A new target for cardiovascular disease therapeutics. J. Cardiovasc. Pharmacol. 2007, 50, 112–119. [Google Scholar] [CrossRef]
  75. Oudit, G.Y.; Liu, G.C.; Zhong, J.; Basu, R.; Chow, F.L.; Zhou, J.; Loibner, H.; Janzek, E.; Schuster, M.; Penninger, J.M.; et al. Human recombinant ACE2 reduces the progression of diabetic nephropathy. Diabetes 2010, 59, 529–538. [Google Scholar] [CrossRef] [Green Version]
  76. Duan, Y.; Beli, E.; Li Calzi, S.; Quigley, J.L.; Miller, R.C.; Moldovan, L.; Feng, D.; Salazar, T.E.; Hazra, S.; Al-Sabah, J.; et al. Loss of angiotensin-converting enzyme 2 exacerbates diabetic retinopathy by promoting bone marrow dysfunction. Stem. Cells 2018, 36, 1430–1440. [Google Scholar] [CrossRef] [Green Version]
  77. Takeda, M.; Yamamoto, K.; Takemura, Y.; Takeshita, H.; Hongyo, K.; Kawai, T.; Hanasaki-Yamamoto, H.; Oguro, R.; Takami, Y.; Tatara, Y.; et al. Loss of ACE2 exaggerates high-calorie diet–induced insulin resistance by reduction of GLUT4 in mice. Diabetes 2013, 62, 223–233. [Google Scholar] [CrossRef] [Green Version]
  78. Cherney, D.Z.; Scholey, J.W.; Sochett, E.; Bradley, T.J.; Reich, H.N. The acute effect of clamped hyperglycemia on the urinary excretion of inflammatory cytokines/chemokines in uncomplicated type 1 diabetes: A pilot study. Diabetes Care 2011, 34, 177–180. [Google Scholar] [CrossRef] [Green Version]
  79. Conte, C.; Riant, E.; Toutain, C.; Pujol, F.; Arnal, J.F.; Lenfant, F.; Prats, A.C. FGF2 translationally induced by hypoxia is involved in negative and positive feedback loops with HIF-1α. PLoS ONE 2008, 3, e3078. [Google Scholar] [CrossRef]
  80. Hagiwara, S.; Iwasaka, H.; Hasegawa, A.; Koga, H.; Noguchi, T. Effects of hyperglycemia and insulin therapy on high mobility group box 1 in endotoxin-induced acute lung injury in a rat model. Crit. Care Med. 2008, 36, 2407–2413. [Google Scholar] [CrossRef]
  81. Yu, A.P.; Tam, B.T.; Yau, W.Y.; Chan, K.S.; Yu, S.S.; Chung, T.L.; Siu, P.M. Association of endothelin-1 and matrix metallopeptidase-9 with metabolic syndrome in middle-aged and older adults. Diabetol. Metab. Syndr. 2015, 7, 111. [Google Scholar] [CrossRef] [Green Version]
  82. Pala, L.; Rotella, C.M. The role of DPP4 activity in cardiovascular districts: In vivo and in vitro evidence. J. Diabetes Res. 2013, 2013, 590456. [Google Scholar] [CrossRef] [Green Version]
  83. Suda, M.; Shimizu, I.; Yoshida, Y.; Hayashi, Y.; Ikegami, R.; Katsuumi, G.; Wakasugi, T.; Yoshida, Y.; Okuda, S.; Soga, T.; et al. Inhibition of dipeptidyl peptidase-4 ameliorates cardiac ischemia and systolic dysfunction by up-regulating the FGF-2/EGR-1 pathway. PLoS ONE 2017, 12, e0182422. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Marchetti, C.; Di Carlo, A.; Facchiano, F.; Senatore, C.; De Cristofaro, R.; Luzi, A.; Federici, M.; Romani, M.; Napolitano, M.; Capogrossi, M.C.; et al. High mobility group box 1 is a novel substrate of dipeptidyl peptidase-IV. Diabetologia 2012, 55, 236–244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Arscott, W.T.; LaBauve, A.E.; May, V.; Wesley, U.V. Suppression of neuroblastoma growth by dipeptidyl peptidase IV: Relevance of chemokine regulation and caspase activation. Oncogene 2009, 28, 479–491. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Xu, Z.S.; Shu, T.; Kang, L.; Wu, D.; Zhou, X.; Liao, B.W.; Sun, X.L.; Zhou, X.; Wang, Y.Y. Temporal profiling of plasma cytokines, chemokines and growth factors from mild, severe and fatal COVID-19 patients. Signal Transduct. Target. Ther. 2020, 5, 100. [Google Scholar] [CrossRef]
  87. Fernández-Castañeda, A.; Lu, P.; Geraghty, A.C.; Song, E.; Lee, M.H.; Wood, J.; Yalcin, B.; Taylor, K.R.; Dutton, S.; Acosta-Alvarez, L.; et al. Mild respiratory SARS-CoV-2 infection can cause multi-lineage cellular dysregulation and myelin loss in the brain. bioRxiv 2022. [Google Scholar] [CrossRef]
  88. Forssmann, U.; Stoetzer, C.; Stephan, M.; Kruschinski, C.; Skripuletz, T.; Schade, J.; Schmiedl, A.; Pabst, R.; Wagner, L.; Hoffmann, T.; et al. Inhibition of CD26/dipeptidyl peptidase IV enhances CCL11/eotaxin-mediated recruitment of eosinophils in vivo. J. Immunol. 2008, 181, 1120–1127. [Google Scholar] [CrossRef] [Green Version]
  89. You, J.; Li, H.; Fan, P.; Yang, X.; Wei, Y.; Zheng, L.; Li, Z.; Yi, C. Inspiration for COVID-19 treatment: Network analysis and experimental validation of baicalin for cytokine storm. Front. Pharmacol. 2022, 13, 853496. [Google Scholar] [CrossRef]
  90. Gonzalez, Y.; Herrera, M.T.; Soldevila, G.; Garcia-Garcia, L.; Fabián, G.; Pérez-Armendariz, E.M.; Bobadilla, K.; Guzmán-Beltrán, S.; Sada, E.; Torres, M. High glucose concentrations induce TNF-α production through the down-regulation of CD33 in primary human monocytes. BMC Immunol. 2012, 13, 19. [Google Scholar] [CrossRef] [Green Version]
  91. Anoop, S.; Misra, A.; Bhatt, S.P.; Gulati, S.; Pandey, R.M.; Mahajan, H. High circulating plasma dipeptidyl peptidase-4 levels in non-obese Asian Indians with type 2 diabetes correlate with fasting insulin and LDL-C levels, triceps skinfolds, total intra-abdominal adipose tissue volume and presence of diabetes: A case–control study. BMJ Open Diabetes Res. Care 2017, 5, e000393. [Google Scholar] [CrossRef] [Green Version]
  92. Guo, Y.; Hu, K.; Li, Y.; Lu, C.; Ling, K.; Cai, C.; Wang, W.; Ye, D. Targeting TNF-α for COVID-19: Recent Advanced and Controversies. Front. Public Health 2022, 10, 833967. [Google Scholar] [CrossRef]
  93. Yang, G.; Jia, Z.; Aoyagi, T.; McClain, D.; Mortensen, R.M.; Yang, T. Systemic PPARγ deletion impairs circadian rhythms of behavior and metabolism. PLoS ONE 2012, 7, e38117. [Google Scholar] [CrossRef] [Green Version]
  94. Baumeier, C.; Schlüter, L.; Saussenthaler, S.; Laeger, T.; Rödiger, M.; Alaze, S.A.; Fritsche, L.; Häring, H.U.; Stefan, N.; Fritsche, A.; et al. Elevated hepatic DPP4 activity promotes insulin resistance and non-alcoholic fatty liver disease. Mol. Metab. 2017, 6, 1254–1263. [Google Scholar] [CrossRef]
  95. Desterke, C.; Turhan, A.G.; Bennaceur-Griscelli, A.; Griscelli, F. PPARγcistrome repression during activation of lung monocyte-macrophages in severe COVID-19. Iscience 2020, 23, 101611. [Google Scholar] [CrossRef]
  96. Barbosa-Desongles, A.; Hernández, C.; De Torres, I.; Munell, F.; Poupon, M.F.; Simó, R.; Selva, D.M. Diabetes protects from prostate cancer by downregulating androgen receptor: New insights from LNCaP cells and PAC120 mouse model. PLoS ONE 2013, 8, e74179. [Google Scholar] [CrossRef]
  97. Lin, H.P.; Lin, C.Y.; Liu, C.C.; Su, L.C.; Huo, C.; Kuo, Y.Y.; Tseng, J.C.; Hsu, J.M.; Chen, C.K.; Chuu, C.P. Caffeic acid phenethyl ester as a potential treatment for advanced prostate cancer targeting akt signaling. Int. J. Mol. Sci. 2013, 14, 5264–5283. [Google Scholar] [CrossRef] [Green Version]
  98. Mohamed, M.S.; Moulin, T.C.; Schiöth, H.B. Sex differences in COVID-19: The role of androgens in disease severity and progression. Endocrine 2021, 71, 3–8. [Google Scholar] [CrossRef]
  99. Arroba, A.I.; Mazzeo, A.; Cazzoni, D.; Beltramo, E.; Hernández, C.; Porta, M.; Simó, R.; Valverde, Á.M. Somatostatin protects photoreceptor cells against high glucose–induced apoptosis. Mol. Vis. 2016, 22, 1522. [Google Scholar]
  100. Temming, P.; Tröger, B.; Thonnissen, S.; Holterhus, P.M.; Schultz, C.; Härtel, C. The effect of hyperglycemia on neonatal immune responses in-vitro. J. Matern.-Fetal Neonatal Med. 2012, 25, 94–98. [Google Scholar] [CrossRef]
  101. Zeng, X.; Wang, H.; Xing, X.; Wang, Q.; Li, W. Dexmedetomidine protects against transient global cerebral ischemia/reperfusion induced oxidative stress and inflammation in diabetic rats. PLoS ONE 2016, 11, e0151620. [Google Scholar] [CrossRef] [Green Version]
  102. Ekinci, E.I.; Torkamani, N.; Ramchand, S.K.; Churilov, L.; Sikaris, K.A.; Lu, Z.X.; Houlihan, C.A. Higher maternal serum prolactin levels are associated with reduced glucose tolerance during pregnancy. J. Diabetes Investig. 2017, 8, 697–700. [Google Scholar] [CrossRef] [Green Version]
  103. Wu, Y.; Tang, L.; Chen, B. Oxidative stress: Implications for the development of diabetic retinopathy and antioxidant therapeutic perspectives. Oxidative Med. Cell. Longev. 2014, 2014, 752387. [Google Scholar] [CrossRef] [Green Version]
  104. Dai, H.; Liu, Q.; Liu, B. Research progress on mechanism of podocyte depletion in diabetic nephropathy. J. Diabetes Res. 2017, 2017, 2615286. [Google Scholar] [CrossRef]
  105. Kaur, H.; Chien, A.; Jialal, I. Hyperglycemia induces Toll like receptor 4 expression and activity in mouse mesangial cells: Relevance to diabetic nephropathy. Am. J. Physiol.-Ren. Physiol. 2012, 303, F1145–F1150. [Google Scholar] [CrossRef] [Green Version]
  106. Morey, M.; O’Gaora, P.; Pandit, A.; Hélary, C. Hyperglycemia acts in synergy with hypoxia to maintain the pro-inflammatory phenotype of macrophages. PLoS ONE 2019, 14, e0220577. [Google Scholar] [CrossRef] [Green Version]
  107. Rodriguez-Cerdeira, C.; Sanchez-Blanco, E.; Molares-Vila, A.; Alba, A. Unveiling new molecular factors useful for detection of pelvic inflammatory disease due to Chlamydia trachomatis infection. Int. Sch. Res. Not. 2012, 2012, 581725. [Google Scholar] [CrossRef] [Green Version]
  108. Mishiro, T.; Nakano, S.; Takahara, S.; Miki, M.; Nakamura, Y.; Yasuoka, S.; Nikawa, T.; Yasui, N. Relationship between cathepsin B and thrombin in rheumatoid arthritis. J. Rheumatol. 2004, 31, 1265–1273. [Google Scholar]
  109. Klemm, C.; Bruchhagen, C.; Van Krüchten, A.; Niemann, S.; Löffler, B.; Peters, G.; Ludwig, S.; Ehrhardt, C. Mitogen-activated protein kinases (MAPKs) regulate IL-6 over-production during concomitant influenza virus and Staphylococcus aureus infection. Sci. Rep. 2017, 7, 42473. [Google Scholar] [CrossRef] [Green Version]
  110. Steinfeld, S.; Maho, A.; Chaboteaux, C.; Daelemans, P.; Pochet, R.; Appelboom, T.; Kiss, R. Prolactin up-regulates cathepsin B and D expression in minor salivary glands of patients with Sjögren’s syndrome. Lab. Investig. 2000, 80, 1711–1720. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Yan, S.; Berquin, I.M.; Troen, B.R.; Sloane, B.F. Transcription of human cathepsin B is mediated by Sp1 and Ets family factors in glioma. DNA Cell Biol. 2000, 19, 79–91. [Google Scholar] [CrossRef] [PubMed]
  112. Koblansky, A.A.; Truax, A.D.; Liu, R.; Montgomery, S.A.; Ding, S.; Wilson, J.E.; Brickey, W.J.; Mühlbauer, M.; McFadden, R.M.T.; Hu, P.; et al. The innate immune receptor NLRX1 functions as a tumor suppressor by reducing colon tumorigenesis and key tumor-promoting signals. Cell Rep. 2016, 14, 2562–2575. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Barnes, T.C.; Cross, A.; Anderson, M.E.; Edwards, S.W.; Moots, R.J. Relative α1-anti-trypsin deficiency in systemic sclerosis. Rheumatology 2011, 50, 1373–1378. [Google Scholar] [CrossRef] [Green Version]
  114. Foghsgaard, L.; Wissing, D.; Mauch, D.; Lademann, U.; Bastholm, L.; Boes, M.; Elling, F.; Leist, M.; Jäättelä, M. Cathepsin B acts as a dominant execution protease in tumor cell apoptosis induced by tumor necrosis factor. J. Cell Biol. 2001, 153, 999–1010. [Google Scholar] [CrossRef] [Green Version]
  115. Li, S.; Zhang, Y.; Guan, Z.; Li, H.; Ye, M.; Chen, X.; Shen, J.; Zhou, Y.; Shi, Z.L.; Zhou, P.; et al. SARS-CoV-2 triggers inflammatory responses and cell death through caspase-8 activation. Signal Transduct. Target. Ther. 2020, 5, 235. [Google Scholar] [CrossRef]
  116. Lin, C.F.; Tsai, C.C.; Huang, W.C.; Wang, Y.C.; Tseng, P.C.; Tsai, T.T.; Chen, C.L. Glycogen synthase kinase-3β and caspase-2 mediate ceramide-and etoposide-induced apoptosis by regulating the lysosomal-mitochondrial axis. PLoS ONE 2016, 11, e0145460. [Google Scholar] [CrossRef]
  117. Park, J.H.; Lee, H.K. Re-analysis of single cell transcriptome reveals that the NR3C1-CXCL8-neutrophil axis determines the severity of COVID-19. Front. Immunol. 2020, 11, 2145. [Google Scholar] [CrossRef]
  118. Santa Cruz, A.; Mendes-Frias, A.; Oliveira, A.I.; Dias, L.; Matos, A.R.; Carvalho, A.; Capela, C.; Pedrosa, J.; Castro, A.G.; Silvestre, R. Interleukin-6 is a biomarker for the development of fatal severe acute respiratory syndrome coronavirus 2 pneumonia. Front. Immunol. 2021, 12, 263. [Google Scholar] [CrossRef]
  119. Al-Kuraishy, H.M.; Al-Gareeb, A.I.; Butnariu, M.; Batiha, G.E.S. The crucial role of prolactin-lactogenic hormone in COVID-19. Mol. Cell. Biochem. 2022, 477, 1381–1392. [Google Scholar] [CrossRef]
  120. Yoshimura, K.; Tsuchida, T.; Kawamoto, K. Expression of cathepsin B and cystatin C in the human adenohypophysis and in pituitary adenomas. Oncol. Rep. 2000, 7, 27–58. [Google Scholar] [CrossRef]
  121. Auriemma, R.S.; De Alcubierre, D.; Pirchio, R.; Pivonello, R.; Colao, A. Glucose abnormalities associated to prolactin secreting pituitary adenomas. Front. Endocrinol. 2019, 10, 327. [Google Scholar] [CrossRef] [Green Version]
  122. Khokhar, M.; Tomo, S.; Purohit, P. MicroRNAs based regulation of cytokine regulating immune expressed genes and their transcription factors in COVID-19. Meta Gene 2022, 31, 100990. [Google Scholar] [CrossRef]
  123. Mukherjee, S. Toll-like receptor 4 in COVID-19: Friend or foe? Future Virol. 2022, 17, 415–417. [Google Scholar] [CrossRef]
  124. Minuzzi, L.G.; Seelaender, M.; Silva, B.S.D.A.; Cunha, E.D.B.B.; Deus, M.D.C.; Vasconcellos, F.T.F.; Marqueze, L.F.B.; Gadotti, A.C.; Baena, C.P.; Pereira, T.; et al. COVID-19 outcome relates with circulating BDNF, according to patient adiposity and age. Front. Nutr. 2021, 8, 784429. [Google Scholar] [CrossRef]
  125. Ferreira, A.C.; Soares, V.C.; de Azevedo-Quintanilha, I.G.; Dias, S.D.S.G.; Fintelman-Rodrigues, N.; Sacramento, C.Q.; Mattos, M.; de Freitas, C.S.; Temerozo, J.R.; Teixeira, L.; et al. SARS-CoV-2 engages inflammasome and pyroptosis in human primary monocytes. Cell Death Discov. 2021, 7, 43. [Google Scholar] [CrossRef]
  126. Plassmeyer, M.; Alpan, O.; Corley, M.J.; Premeaux, T.A.; Lillard, K.; Coatney, P.; Vaziri, T.; Michalsky, S.; Pang, A.P.; Bukhari, Z.; et al. Caspases and therapeutic potential of caspase inhibitors in moderate–severe SARS-CoV-2 infection and long COVID. Allergy 2022, 77, 118–129. [Google Scholar] [CrossRef]
  127. Gulhan, P.Y.; Eroz, R.; Ataoglu, O.; Ince, N.; Davran, F.; Öztürk, C.E.; Gamsızkan, Z.; Balbay, O.A. The evaluation of both the expression and serum protein levels of Caspase-3 gene in patients with different degrees of SARS-CoV-2 infection. J. Med. Virol. 2022, 94, 897–905. [Google Scholar] [CrossRef]
  128. Tjan, L.H.; Furukawa, K.; Nagano, T.; Kiriu, T.; Nishimura, M.; Arii, J.; Hino, Y.; Iwata, S.; Nishimura, Y.; Mori, Y. Early differences in cytokine production by severity of coronavirus disease 2019. J. Infect. Dis. 2021, 223, 1145–1149. [Google Scholar] [CrossRef]
  129. Islam, H.; Chamberlain, T.C.; Mui, A.L.; Little, J.P. Elevated interleukin-10 levels in COVID-19: Potentiation of pro-inflammatory responses or impaired anti-inflammatory action? Front. Immunol. 2021, 12, 677008. [Google Scholar] [CrossRef]
  130. Ferreira-Gomes, M.; Kruglov, A.; Durek, P.; Heinrich, F.; Tizian, C.; Heinz, G.A.; Pascual-Reguant, A.; Du, W.; Mothes, R.; Fan, C.; et al. SARS-CoV-2 in severe COVID-19 induces a TGF-β-dominated chronic immune response that does not target itself. Nat. Commun. 2021, 12, 1961. [Google Scholar] [CrossRef]
  131. Zuniga, M.; Gomes, C.; Carsons, S.E.; Bender, M.T.; Cotzia, P.; Miao, Q.R.; Lee, D.C.; Rodriguez, A. Autoimmunity to Annexin A2 predicts mortality among hospitalised COVID-19 patients. Eur. Respir. J. 2021, 58, 2100918. [Google Scholar] [CrossRef]
  132. Patil, P.; Shetty, P.; Kuriakose, N.; Gollapalli, P.; Shetty, S.; Bhandary, R.; Vishwanatha, J.K.; Ghate, S.D. Molecular Insights on the Possible Role of Annexin A2 in COVID-19 Pathogenesis and Post-Infection Complications. Int. J. Mol. Sci. 2021, 22, 11028. [Google Scholar] [CrossRef]
  133. Klein, R.; Soung, A.; Sissoko, C.; Nordvig, A.; Canoll, P.; Mariani, M.; Jiang, X.; Bricker, T.; Goldman, J.; Rosoklija, G.; et al. COVID-19 induces neuroinflammation and loss of hippocampal neurogenesis. Res. Sq. 2021. [Google Scholar] [CrossRef]
  134. Aneva, I.Y.; Habtemariam, S.; Banach, M.; Sil, P.C.; Sarkar, K.; Sahebnasagh, A.; Kamal, M.A.; Khayatkashani, M.; Kashani, H.R.K. Can We Use mTOR Inhibitors for COVID-19 Therapy? Comb. Chem. High Throughput Screen. 2021. [Google Scholar] [CrossRef] [PubMed]
  135. Zeng, J.; Xie, X.; Feng, X.L.; Xu, L.; Han, J.B.; Yu, D.; Zou, Q.C.; Liu, Q.; Li, X.; Ma, G.; et al. Specific inhibition of the NLRP3 inflammasome suppresses immune overactivation and alleviates COVID-19 like pathology in mice. eBioMedicine 2022, 75, 103803. [Google Scholar] [CrossRef] [PubMed]
  136. Sarma, A.; Christenson, S.A.; Byrne, A.; Mick, E.; Pisco, A.O.; DeVoe, C.; Deiss, T.; Ghale, R.; Zha, B.S.; Tsitsiklis, A.; et al. Tracheal aspirate RNA sequencing identifies distinct immunological features of COVID-19 ARDS. Nat. Commun. 2021, 12, 5152. [Google Scholar] [CrossRef]
  137. Vaz de Paula, C.B.; de Azevedo, M.L.V.; Nagashima, S.; Martins, A.P.C.; Malaquias, M.A.S.; Miggiolaro, A.F.R.D.S.; da Silva Motta Júnior, J.; Avelino, G.; do Carmo, L.A.P.; Carstens, L.B.; et al. IL-4/IL-13 remodeling pathway of COVID-19 lung injury. Sci. Rep. 2020, 10, 18689. [Google Scholar] [CrossRef]
  138. Leng, L.; Cao, R.; Ma, J.; Mou, D.; Zhu, Y.; Li, W.; Lv, L.; Gao, D.; Zhang, S.; Gong, F.; et al. Pathological features of COVID-19-associated lung injury: A preliminary proteomics report based on clinical samples. Signal Transduct. Target. Ther. 2020, 5, 240. [Google Scholar] [CrossRef]
  139. Ziff, O.J.; Ashton, N.J.; Mehta, P.R.; Brown, R.; Athauda, D.; Heaney, J.; Heslegrave, A.J.; Benedet, A.L.; Blennow, K.; Checkley, A.M.; et al. Amyloid processing in COVID-19-associated neurological syndromes. J. Neurochem. 2022, 161, 146–157. [Google Scholar] [CrossRef]
  140. Kohli, A.; Sauerhering, L.; Fehling, S.K.; Klann, K.; Geiger, H.; Becker, S.; Koch, B.; Baer, P.C.; Strecker, T.; Münch, C. Proteomic landscape of SARS-CoV-2–and MERS-CoV–infected primary human renal epithelial cells. Life Sci. Alliance 2022, 5, e202201371. [Google Scholar] [CrossRef]
  141. Ackermann, M.; Mentzer, S.J.; Kolb, M.; Jonigk, D. Inflammation and intussusceptive angiogenesis in COVID-19: Everything in and out of flow. Eur. Respir. J. 2020, 56, 2003147. [Google Scholar] [CrossRef]
  142. Ren, X.; Wen, W.; Fan, X.; Hou, W.; Su, B.; Cai, P.; Li, J.; Liu, Y.; Tang, F.; Zhang, F.; et al. COVID-19 immune features revealed by a large-scale single-cell transcriptome atlas. Cell 2021, 184, 1895–1913. [Google Scholar] [CrossRef]
  143. Harford, J.B.; Kim, S.S.; Pirollo, K.F.; Chang, E.H. TP53 Gene Therapy as a Potential Treatment for Patients with COVID-19. Viruses 2022, 14, 739. [Google Scholar] [CrossRef]
  144. Yu, X.; Xu, X.; Wu, T.; Huang, W.; Xu, C.; Xie, W.; Long, X. APOA1 Level is Negatively Correlated with the Severity of COVID-19. Int. J. Gen. Med. 2022, 15, 689. [Google Scholar] [CrossRef]
  145. Bell, L.C.; Meydan, C.; Kim, J.; Foox, J.; Butler, D.; Mason, C.E.; Shapira, S.D.; Noursadeghi, M.; Pollara, G. Transcriptional response modules characterize IL-1β and IL-6 activity in COVID-19. Iscience 2021, 24, 101896. [Google Scholar] [CrossRef]
  146. Perreau, M.; Suffiotti, M.; Marques-Vidal, P.; Wiedemann, A.; Levy, Y.; Laouénan, C.; Ghosn, J.; Fenwick, C.; Comte, D.; Roger, T.; et al. The cytokines HGF and CXCL13 predict the severity and the mortality in COVID-19 patients. Nat. Commun. 2021, 12, 4888. [Google Scholar] [CrossRef]
  147. Wu, P.; Chen, D.; Ding, W.; Wu, P.; Hou, H.; Bai, Y.; Zhou, Y.; Li, K.; Xiang, S.; Liu, P.; et al. The trans-omics landscape of COVID-19. Nat. Commun. 2021, 12, 4543. [Google Scholar] [CrossRef]
  148. Dai, J.; Zhang, Q.; Wan, C.; Liu, J.; Zhang, Q.; Yu, Y.; Wang, J. Significances of viable synergistic autophagy-associated cathepsin B and cathepsin D (CTSB/CTSD) as potential biomarkers for sudden cardiac death. BMC Cardiovasc. Disord. 2021, 21, 233. [Google Scholar] [CrossRef]
  149. Liu, C.; Cai, Z.; Hu, T.; Yao, Q.; Zhang, L. Cathepsin B aggravated doxorubicin-induced myocardial injury via NF-κBsignalling. Mol. Med. Rep. 2020, 22, 4848–4856. [Google Scholar] [CrossRef]
  150. Niu, R.; Wang, J.; Geng, C.; Li, Y.; Dong, L.; Liu, L.; Chang, Y.; Shen, J.; Nie, Z.; Zhang, Y.; et al. Tandem mass tag-based proteomic analysis reveals cathepsin-mediated anti-autophagic and pro-apoptotic effects under proliferative diabetic retinopathy. Aging 2021, 13, 973. [Google Scholar] [CrossRef]
  151. Mittal, K.; Mani, R.J.; Katare, D.P. Type 3 diabetes: Cross talk between differentially regulated proteins of type 2 diabetes mellitus and Alzheimer’s disease. Sci. Rep. 2016, 6, 25589. [Google Scholar] [CrossRef]
  152. Tanaka, J.; Qiang, L.; Banks, A.S.; Welch, C.L.; Matsumoto, M.; Kitamura, T.; Ido-Kitamura, Y.; DePinho, R.A.; Accili, D. Foxo1 links hyperglycemia to LDL oxidation and endothelial nitric oxide synthase dysfunction in vascular endothelial cells. Diabetes 2009, 58, 2344–2354. [Google Scholar] [CrossRef] [Green Version]
  153. Zhang, D.; Wan, A.; Chiu, A.P.L.; Wang, Y.; Wang, F.; Neumaier, K.; Lal, N.; Bround, M.J.; Johnson, J.D.; Vlodavsky, I.; et al. Hyperglycemia-induced secretion of endothelial heparanase stimulates a vascular endothelial growth factor autocrine network in cardiomyocytes that promotes recruitment of lipoprotein lipase. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2830–2838. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  154. Li, X.; Weng, H.; Xu, C.; Reece, E.A.; Yang, P. Oxidative stress–induced JNK1/2 activation triggers proapoptotic signaling and apoptosis that leads to diabetic embryopathy. Diabetes 2012, 61, 2084–2092. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Hayashi, Y.; Toyomasu, Y.; Saravanaperumal, S.A.; Bardsley, M.R.; Smestad, J.A.; Lorincz, A.; Eisenman, S.T.; Cipriani, G.; Holte, M.H.N.; Al Khazal, F.J.; et al. Hyperglycemia increases interstitial cells of Cajal via MAPK1 and MAPK3 signaling to ETV1 and KIT, leading to rapid gastric emptying. Gastroenterology 2017, 153, 521–535. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Tohda, C.; Tohda, M. Extracellular cathepsin L stimulates axonal growth in neurons. BMC Res. Notes 2017, 10, 613. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Gerber, A.; Welte, T.; Ansorge, S.; Bühling, F. Expression of cathepsins B and L in human lung epithelial cells is regulated by cytokines. Adv. Exp. Med. Biol. 2000, 477, 287–292. [Google Scholar] [CrossRef]
  158. Castets, P.; Frank, S.; Sinnreich, M.; Rüegg, M.A. “Get the balance right”: Pathological significance of autophagy perturbation in neuromuscular disorders. J. Neuromuscul. Dis. 2016, 3, 127–155. [Google Scholar] [CrossRef] [Green Version]
  159. Khamaysi, I.; Singh, P.; Nasser, S.; Awad, H.; Chowers, Y.; Sabo, E.; Hammond, E.; Gralnek, I.; Minkov, I.; Noseda, A.; et al. The role of heparanase in the pathogenesis of acute pancreatitis: A potential therapeutic target. Sci. Rep. 2017, 7, 715. [Google Scholar] [CrossRef] [Green Version]
  160. Xu, Q.F.; Zheng, Y.; Chen, J.; Xu, X.Y.; Gong, Z.J.; Huang, Y.F.; Lu, C.; Maibach, H.I.; Lai, W. Ultraviolet A enhances cathepsin L expression and activity via JNK pathway in human dermal fibroblasts. Chin. Med. J. 2016, 129, 2853–2860. [Google Scholar] [CrossRef]
  161. Fiebiger, E.; Maehr, R.; Villadangos, J.; Weber, E.; Erickson, A.; Bikoff, E.; Ploegh, H.L.; Lennon-Duménil, A.M. Invariant chain controls the activity of extracellular cathepsin L. J. Exp. Med. 2002, 196, 1263–1270. [Google Scholar] [CrossRef] [Green Version]
  162. Buijsers, B.; Yanginlar, C.; de Nooijer, A.; Grondman, I.; Maciej-Hulme, M.L.; Jonkman, I.; Janssen, N.A.; Rother, N.; de Graaf, M.; Pickkers, P.; et al. Increased plasma heparanase activity in COVID-19 patients. Front. Immunol. 2020, 11, 575047. [Google Scholar] [CrossRef]
  163. Goel, S.; Sharif-Askari, F.S.; Askari, N.S.S.; Madkhana, B.; Alwaa, A.M.; Mahboub, B.; Zakeri, A.M.; Ratemi, E.; Hamoudi, R.; Hamid, Q.; et al. SARS-CoV-2 Switches ‘on’MAPK and NFκB Signaling via the Reduction of Nuclear DUSP1 and DUSP5 Expression. Front. Pharmacol. 2021, 12, 631879. [Google Scholar] [CrossRef]
  164. Li, W.; Zhang, L.; Chen, X.; Jiang, Z.; Zong, L.; Ma, Q. Hyperglycemia promotes the epithelial-mesenchymal transition of pancreatic cancer via hydrogen peroxide. Oxidative Med. Cell. Longev. 2016, 2016, 5190314. [Google Scholar] [CrossRef] [Green Version]
  165. Yu, S.; Yu, Y.; Zhang, W.; Yuan, W.; Zhao, N.; Li, Q.; Cui, Y.; Wang, Y.; Li, W.; Sun, Y.; et al. FOXO3a promotes gastric cancer cell migration and invasion through the induction of cathepsin L. Oncotarget 2016, 7, 34773. [Google Scholar] [CrossRef] [Green Version]
  166. Osman, I.O.; Garrec, C.; de Souza, G.A.P.; Zarubica, A.; Belhaouari, D.B.; Baudoin, J.P.; Lepidi, H.; Mege, J.L.; Malissen, B.; Scola, B.L.; et al. Control of CDH1/E-Cadherin Gene Expression and Release of a Soluble Form of E-Cadherin in SARS-CoV-2 Infected Caco-2 Intestinal Cells: Physiopathological Consequences for the Intestinal Forms of COVID-19. Front. Cell. Infect. Microbiol. 2022, 12, 798767. [Google Scholar] [CrossRef]
  167. Garsen, M.; Rops, A.L.; Dijkman, H.; Willemsen, B.; van Kuppevelt, T.H.; Russel, F.G.; Rabelink, T.J.; Berden, J.H.; Reinheckel, T.; van der Vlag, J. Cathepsin L is crucial for the development of early experimental diabetic nephropathy. Kidney Int. 2016, 90, 1012–1022. [Google Scholar] [CrossRef]
  168. Brings, S.; Fleming, T.; Herzig, S.; Nawroth, P.P.; Kopf, S. Urinary cathepsin L is predictive of changes in albuminuria and correlates with glucosepane in patients with type 2 diabetes in a closed-cohort study. J. Diabetes Its Complicat. 2020, 34, 107648. [Google Scholar] [CrossRef]
  169. Piwkowska, A.; Zdrojewski, Ł.; Heleniak, Z.; Dębska-Ślizień, A. Novel Markers in Diabetic Kidney Disease—Current State and Perspectives. Diagnostics 2022, 12, 1205. [Google Scholar] [CrossRef]
  170. Safran, M.; Rosen, N.; Twik, M.; BarShir, R.; Stein, T.I.; Dahary, D.; Fishilevich, S.; Lancet, D. The GeneCards Suite Chapter. In Practical Guide to Life Science Databases; Springer: Berlin/Heidelberg, Germany, 2022; pp. 27–56. [Google Scholar] [CrossRef]
  171. Lawyer, G. Understanding the influence of all nodes in a network. Sci. Rep. 2015, 5, srep08665. [Google Scholar] [CrossRef] [Green Version]
  172. Tay, M.Z.; Poh, C.M.; Rénia, L.; MacAry, P.A.; Ng, L.F. The trinity of COVID-19: Immunity, inflammation and intervention. Nat. Rev. Immunol. 2020, 20, 363–374. [Google Scholar] [CrossRef]
  173. Zhang, Q.; Xiang, R.; Huo, S.; Zhou, Y.; Jiang, S.; Wang, Q.; Yu, F. Molecular mechanism of interaction between SARS-CoV-2 and host cells and interventional therapy. Signal Transduct. Target. Ther. 2021, 6, 233. [Google Scholar] [CrossRef]
  174. Kowluru, R.A.; Shan, Y.; Mishra, M. Dynamic DNA methylation of matrix metalloproteinase-9 in the development of diabetic retinopathy. Lab. Investig. 2016, 96, 1040–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  175. Mishra, M.; Kowluru, R.A. The Role of DNA Methylation in the Metabolic Memory Phenomenon Associated with the Continued Progression of Diabetic Retinopathy. Investig. Ophthalmol. Vis Sci. 2016, 57, 5748–5757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  176. Zhao, J.; Yang, S.; Shu, B.; Chen, L.; Yang, R.; Xu, Y.; Xie, J.; Liu, X.; Qi, S. Transient High Glucose Causes Persistent Vascular Dysfunction and Delayed Wound Healing by the DNMT1-Mediated Ang-1/NF-κB Pathway. J. Investig. Dermatol. 2021, 141, 1573–1584. [Google Scholar] [CrossRef] [PubMed]
  177. Muhammad, J.S.; Saheb Sharif-Askari, N.; Cui, Z.G.; Hamad, M.; Halwani, R. SARS-CoV-2 Infection-Induced Promoter Hypomethylation as an Epigenetic Modulator of Heat Shock Protein A1L (HSPA1L) Gene. Front. Genet. 2021, 12, 622271. [Google Scholar] [CrossRef]
  178. Romere, C.; Duerrschmid, C.; Bournat, J.; Constable, P.; Jain, M.; Xia, F.; Saha, P.K.; Del Solar, M.; Zhu, B.; York, B.; et al. Asprosin, a Fasting-Induced Glucogenic Protein Hormone. Cell 2016, 165, 566–579. [Google Scholar] [CrossRef] [Green Version]
  179. Gaikwad, A.B.; Gupta, J.; Tikoo, K. Epigenetic changes and alteration of Fbn1 and Col3A1 gene expression under hyperglycaemic and hyperinsulinaemic conditions. Biochem. J. 2010, 432, 333–341. [Google Scholar] [CrossRef] [Green Version]
  180. Seyhanli, E.S.; Koyuncu, I.; Yasak, I.H.; Demir, H.A.; Temiz, E. Asprosin and Oxidative Stress Level in COVID-19 Patients. Clin. Lab. 2022, 68, 1. [Google Scholar] [CrossRef]
  181. Eddy, A.C.; Trask, A.J. Growth differentiation factor-15 and its role in diabetes and cardiovascular disease. Cytokine Growth Factor Rev. 2021, 57, 11–18. [Google Scholar] [CrossRef]
  182. Teng, X.; Zhang, J.; Shi, Y.; Liu, Y.; Yang, Y.; He, J.; Luo, S.; Huang, Y.; Liu, Y.; Liu, D.; et al. Comprehensive Profiling of Inflammatory Factors Revealed That Growth Differentiation Factor-15 Is an Indicator of Disease Severity in COVID-19 Patients. Front. Immunol. 2021, 12, 662465. [Google Scholar] [CrossRef]
  183. Ebihara, T.; Matsumoto, H.; Matsubara, T.; Togami, Y.; Nakao, S.; Matsuura, H.; Kojima, T.; Sugihara, F.; Okuzaki, D.; Hirata, H.; et al. Cytokine Elevation in Severe COVID-19 From Longitudinal Proteomics Analysis: Comparison with Sepsis. Front. Immunol. 2022, 12, 798338. [Google Scholar] [CrossRef]
  184. Yang, W.H.; Park, S.Y.; Ji, S.; Kang, J.G.; Kim, J.E.; Song, H.; Mook-Jung, I.; Choe, K.M.; Cho, J.W. O-GlcNAcylation regulates hyperglycemia-induced GPX1 activation. Biochem. Biophys. Res. Commun. 2010, 391, 756–761. [Google Scholar] [CrossRef]
  185. Sultan, C.S.; Saackel, A.; Stank, A.; Fleming, T.; Fedorova, M.; Hoffmann, R.; Wade, R.C.; Hecker, M.; Wagner, A.H. Impact of carbonylation on glutathione peroxidase-1 activity in human hyperglycemic endothelial cells. Redox Biol. 2018, 16, 113–122. [Google Scholar] [CrossRef]
  186. Huang, J.Q.; Zhou, J.C.; Wu, Y.Y.; Ren, F.Z.; Lei, X.G. Role of glutathione peroxidase 1 in glucose and lipid metabolism-related diseases. Free Radic. Biol. Med. 2018, 127, 108–115. [Google Scholar] [CrossRef]
  187. Maamoun, H.; Benameur, T.; Pintus, G.; Munusamy, S.; Agouni, A. Crosstalk Between Oxidative Stress and Endoplasmic Reticulum (ER) Stress in Endothelial Dysfunction and Aberrant Angiogenesis Associated With Diabetes: A Focus on the Protective Roles of Heme Oxygenase (HO)-1. Front Physiol. 2019, 10, 70. [Google Scholar] [CrossRef] [Green Version]
  188. Toro, A.; Ruiz, M.S.; Lage-Vickers, S.; Sanchis, P.; Sabater, A.; Pascual, G.; Seniuk, R.; Cascardo, F.; Ledesma-Bazan, S.; Vilicich, F.; et al. A Journey into the Clinical Relevance of Heme Oxygenase 1 for Human Inflammatory Disease and Viral Clearance: Why Does It Matter on the COVID-19 Scene? Antioxidants 2022, 11, 276. [Google Scholar] [CrossRef]
  189. Singh, R.D.; Barry, M.A.; Croatt, A.J.; Ackerman, A.W.; Grande, J.P.; Diaz, R.M.; Vile, R.G.; Agarwal, A.; Nath, K.A. The spike protein of SARS-CoV-2 induces heme oxygenase-1: Pathophysiologic implications. Biochim. Biophys. Acta Mol. Basis Dis. 2022, 1868, 166322. [Google Scholar] [CrossRef]
  190. Abraham, N.G.; Kushida, T.; McClung, J.; Weiss, M.; Quan, S.; Lafaro, R.; Darzynkiewicz, Z.; Wolin, M. Heme oxygenase-1 attenuates glucose-mediated cell growth arrest and apoptosis in human microvessel endothelial cells. Circ. Res. 2003, 93, 507–514. [Google Scholar] [CrossRef] [Green Version]
  191. Maamoun, H.; Zachariah, M.; McVey, J.H.; Green, F.R.; Agouni, A. Heme oxygenase (HO)-1 induction prevents Endoplasmic Reticulum stress-mediated endothelial cell death and impaired angiogenic capacity. Biochem. Pharmacol. 2017, 127, 46–59. [Google Scholar] [CrossRef]
  192. Tiwari, S.; Ndisang, J.F. The heme oxygenase system and type-1 diabetes. Curr. Pharm. Des. 2014, 20, 1328–1337. [Google Scholar] [CrossRef]
  193. Al-Hamodi, Z.; Saif-Ali, R.; Ismail, I.S.; Ahmed, K.A.; Muniandy, S. Effect of plasminogen activator inhibitor-1 and tissue plasminogen activator polymorphisms on susceptibility to type 2 diabetes in Malaysian subjects. J. Biomed. Biotechnol. 2012, 2012, 234937. [Google Scholar] [CrossRef] [Green Version]
  194. Venkat, P.; Chopp, M.; Chen, J. Blood–brain barrier disruption, vascular impairment, and ischemia/reperfusion damage in diabetic stroke. J. Am. Heart Assoc. 2017, 6, e005819. [Google Scholar] [CrossRef]
  195. Kelliher, S.; Weiss, L.; Cullivan, S.; O’Rourke, E.; Murphy, C.A.; Toolan, S.; Lennon, Á.; Szklanna, P.B.; Comer, S.P.; Macleod, H.; et al. Non-severe COVID-19 is associated with endothelial damage and hypercoagulability despite pharmacological thromboprophylaxis. J. Thromb. Haemost. 2022, 20, 1008–1014. [Google Scholar] [CrossRef]
  196. Liu, N.; Long, H.; Sun, J.; Li, H.; He, Y.; Wang, Q.; Pan, K.; Tong, Y.; Wang, B.; Wu, Q.; et al. New laboratory evidence for the association between endothelial dysfunction and COVID-19 disease progression. J. Med. Virol. 2022, 94, 3112–3120. [Google Scholar] [CrossRef]
  197. Rubina, K.; Shmakova, A.; Shabanov, A.; Andreev, Y.; Borovkova, N.; Kulabukhov, V.; Evseev, A.; Popugaev, K.; Petrikov, S.; Semina, E. Novel prognostic determinants of COVID-19-related mortality: A pilot study on severely-ill patients in Russia. PLoS ONE 2022, 17, e0264072. [Google Scholar] [CrossRef]
  198. Tundo, G.R.; Sbardella, D.; Ciaccio, C.; Grasso, G.; Gioia, M.; Coletta, A.; Polticelli, F.; Di Pierro, D.; Milardi, D.; Van Endert, P.; et al. Multiple functions of insulin-degrading enzyme: A metabolic crosslight? Crit. Rev. Biochem. Mol. Biol. 2017, 52, 554–582. [Google Scholar] [CrossRef]
  199. González-Casimiro, C.M.; Merino, B.; Casanueva-Álvarez, E.; Postigo-Casado, T.; Cámara-Torres, P.; Fernández-Díaz, C.M.; Leissring, M.A.; Cózar-Castellano, I.; Perdomo, G. Modulation of Insulin Sensitivity by Insulin-Degrading Enzyme. Biomedicines 2021, 9, 86. [Google Scholar] [CrossRef]
  200. Flentje, A.; Kalsi, R.; Monahan, T.S. Small GTPases and Their Role in Vascular Disease. Int. J. Mol. Sci. 2019, 20, 917. [Google Scholar] [CrossRef] [Green Version]
  201. Huang, Z.; Peng, Y.; Yu, H.; Yu, X.; Zhou, J.; Xiao, J. RhoA protects the podocytes against high glucose-induced apoptosis through YAP and plays critical role in diabetic nephropathy. Biochem. Biophys. Res. Commun. 2018, 504, 949–956. [Google Scholar] [CrossRef]
  202. Mahavadi, S.; Sriwai, W.; Manion, O.; Grider, J.R.; Murthy, K.S. Diabetes-induced oxidative stress mediates upregulation of RhoA/Rho kinase pathway and hypercontractility of gastric smooth muscle. PLoS ONE 2017, 12, e0178574. [Google Scholar] [CrossRef]
  203. Hasankhani, A.; Bahrami, A.; Sheybani, N.; Aria, B.; Hemati, B.; Fatehi, F.; Farahani, H.G.M.; Javanmard, G.; Rezaee, M.; Kastelic, J.P.; et al. Differential Co-Expression Network Analysis Reveals Key Hub-High Traffic Genes as Potential Therapeutic Targets for COVID-19 Pandemic. Front. Immunol. 2021, 12, 789317. [Google Scholar] [CrossRef] [PubMed]
  204. Sardar, R.; Satish, D.; Gupta, D. Identification of Novel SARS-CoV-2 Drug Targets by Host MicroRNAs and Transcription Factors Co-regulatory Interaction Network Analysis. Front. Genet. 2020, 11, 571274. [Google Scholar] [CrossRef] [PubMed]
  205. Ryter, S.W. Heme Oxygenase-1: An Anti-Inflammatory Effector in Cardiovascular, Lung, and Related Metabolic Disorders. Antioxidants 2022, 11, 555. [Google Scholar] [CrossRef] [PubMed]
  206. Chen, Y.H.; Lin, S.J.; Lin, M.W.; Tsai, H.L.; Kuo, S.S.; Chen, J.W.; Charng, M.J.; Wu, T.C.; Chen, L.C.; Ding, Y.A.; et al. Microsatellite polymorphism in promoter of heme oxygenase-1 gene is associated with susceptibility to coronary artery disease in type 2 diabetic patients. Hum. Genet. 2002, 111, 1–8. [Google Scholar] [CrossRef]
  207. Wu, R.; Zhu, Z.; Zhou, D. VEGF, apelin and HO-1 in diabetic patients with retinopathy: A correlation analysis. BMC Ophthalmol. 2020, 20, 326. [Google Scholar] [CrossRef]
  208. Lenoir, O.; Gaillard, F.; Lazareth, H.; Robin, B.; Tharaux, P.L. Hmox1 deficiency sensitizes mice to peroxynitrite formation and diabetic glomerular microvascular injuries. J. Diabetes Res. 2017, 2017, 9603924. [Google Scholar] [CrossRef] [Green Version]
  209. Negi, G.; Nakkina, V.; Kamble, P.; Sharma, S.S. Heme oxygenase-1, a novel target for the treatment of diabetic complications: Focus on diabetic peripheral neuropathy. Pharmacol. Res. 2015, 102, 158–167. [Google Scholar] [CrossRef]
  210. Rossi, M.; Piagnerelli, M.; Van Meerhaeghe, A.; ZouaouiBoudjeltia, K. Heme oxygenase-1 (HO-1) cytoprotective pathway: A potential treatment strategy against coronavirus disease 2019 (COVID-19)-induced cytokine storm syndrome. Med. Hypotheses 2020, 144, 110242. [Google Scholar] [CrossRef]
  211. Kearney, K.; Tomlinson, D.; Smith, K.; Ajjan, R. Hypofibrinolysis in diabetes: A therapeutic target for the reduction of cardiovascular risk. Cardiovasc. Diabetol. 2017, 16, 34. [Google Scholar] [CrossRef] [Green Version]
  212. Hoteit, L.; Deeb, A.P.; Andraska, E.A.; Kaltenmeier, C.; Yazdani, H.O.; Tohme, S.; Neal, M.D.; Mota, R.I. The Pathobiological Basis for Thrombotic Complications in COVID-19: A Review of the Literature. Curr. Pathobiol. Rep. 2021, 9, 107–117. [Google Scholar] [CrossRef]
  213. Carlsson, A.C.; Nowak, C.; Lind, L.; Östgren, C.J.; Nyström, F.H.; Sundström, J.; Carrero, J.J.; Riserus, U.; Ingelsson, E.; Fall, T.; et al. Growth differentiation factor 15 (GDF-15) is a potential biomarker of both diabetic kidney disease and future cardiovascular events in cohorts of individuals with type 2 diabetes: A proteomics approach. Upsala J. Med. Sci. 2020, 125, 37–43. [Google Scholar] [CrossRef] [Green Version]
  214. Xiao, Q.A.; He, Q.; Zeng, J.; Xia, X. GDF-15, a future therapeutic target of glucolipid metabolic disorders and cardiovascular disease. Biomed. Pharmacother. 2022, 146, 112582. [Google Scholar] [CrossRef]
  215. Boraldi, F.; Lofaro, F.D.; Cossarizza, A.; Quaglino, D. The “Elastic Perspective” of SARS-CoV-2 Infection and the Role of Intrinsic and Extrinsic Factors. Int. J. Mol. Sci. 2022, 23, 1559. [Google Scholar] [CrossRef]
  216. Guo, X.; Xiang, Y.; Yang, H.; Yu, L.; Peng, X.; Guo, R. Association of the LOX-1 rs1050283 polymorphism with risk for atherosclerotic cerebral infarction and its effect on sLOX-1 and LOX-1 expression in a Chinese population. J. Atheroscler. Thromb. 2016, 24, 572–582. [Google Scholar] [CrossRef] [Green Version]
  217. Mehta, J.L.; Khaidakov, M.; Hermonat, P.L.; Mitra, S.; Wang, X.; Novelli, G.; Sawamura, T. LOX-1: A new target for therapy for cardiovascular diseases. Cardiovasc. Drugs Ther. 2011, 25, 495–500. [Google Scholar] [CrossRef]
  218. Chen, J.; Ren, J.; Loo, W.T.; Hao, L.; Wang, M. Lysyl oxidases expression and histopathological changes of the diabetic rat nephron. Mol. Med. Rep. 2018, 17, 2431–2441. [Google Scholar] [CrossRef] [Green Version]
  219. Yu, S.S.; Du, J.L. Selenoprotein S: A therapeutic target for diabetes and macroangiopathy? Cardiovasc. Diabetol. 2017, 16, 101. [Google Scholar] [CrossRef] [Green Version]
  220. Ye, Y.; Fu, F.; Li, X.; Yang, J.; Liu, H. Selenoprotein S is highly expressed in the blood vessels and prevents vascular smooth muscle cells from apoptosis. J. Cell. Biochem. 2016, 117, 106–117. [Google Scholar] [CrossRef]
  221. Waasdorp, M.; Duitman, J.; Florquin, S.; Spek, A.C. Protease activated receptor 2 in diabetic nephropathy: A double edged sword. Am. J. Transl. Res. 2017, 9, 4512. [Google Scholar]
  222. Subramaniam, S.; Ruf, W.; Bosmann, M. Advocacy of targeting protease-activated receptors in severe coronavirus disease 2019. Br. J. Pharmacol. 2022, 179, 2086–2099. [Google Scholar] [CrossRef]
  223. Noh, H.; Oh, E.Y.; Seo, J.Y.; Yu, M.R.; Kim, Y.O.; Ha, H.; Lee, H.B. Histone deacetylase-2 is a key regulator of diabetes-and transforming growth factor-β1-induced renal injury. Am. J. Physiol.-Ren. Physiol. 2009, 297, F729–F739. [Google Scholar] [CrossRef] [Green Version]
  224. Sun, J.; Wang, Y.; Cui, W.; Lou, Y.; Sun, G.; Zhang, D.; Miao, L. Role of epigenetic histone modifications in diabetic kidney disease involving renal fibrosis. J. Diabetes Res. 2017, 2017, 7242384. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  225. Lindenmeyer, M.T.; Rastaldi, M.P.; Ikehata, M.; Neusser, M.A.; Kretzler, M.; Cohen, C.D.; Schlöndorff, D. Proteinuria and hyperglycemia induce endoplasmic reticulum stress. J. Am. Soc. Nephrol. 2008, 19, 2225–2236. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  226. Li, B.; Zhang, H.Q.; Shi, Y.; Min, Y.B.; Lin, S.F.; Wu, K.L.; Hu, J.; Tang, S.B. Overexpression of nuclear transport factor 2 may protect against diabetic retinopathy. Mol. Vis. 2009, 15, 861. [Google Scholar]
  227. Kwak, H.J.; Yang, D.; Hwang, Y.; Jun, H.S.; Cheon, H.G. Baicalein protects rat insulinoma INS-1 cells from palmitate-induced lipotoxicity by inducing HO-1. PLoS ONE 2017, 12, e0176432. [Google Scholar] [CrossRef]
  228. Raffaele, M.; Li Volti, G.; Barbagallo, I.A.; Vanella, L. Therapeutic efficacy of stem cells transplantation in diabetes: Role of heme oxygenase. Front. Cell Dev. Biol. 2016, 4, 80. [Google Scholar] [CrossRef] [Green Version]
  229. Grant, P.J. The genetics of atherothrombotic disorders: A clinician’s view. J. Thromb. Haemost. 2003, 1, 1381–1390. [Google Scholar] [CrossRef] [Green Version]
  230. Al-Hamodi, Z.; Ismail, I.S.; Saif-Ali, R.; Ahmed, K.A.; Muniandy, S. Association of plasminogen activator inhibitor-1 and tissue plasminogen activator with type 2 diabetes and metabolic syndrome in Malaysian subjects. Cardiovasc. Diabetol. 2011, 10, 23. [Google Scholar] [CrossRef] [Green Version]
  231. Schernthaner-Reiter, M.H.; Kasses, D.; Tugendsam, C.; Riedl, M.; Peric, S.; Prager, G.; Krebs, M.; Promintzer-Schifferl, M.; Clodi, M.; Luger, A.; et al. Growth differentiation factor 15 increases following oral glucose ingestion: Effect of meal composition and obesity. Eur. J. Endocrinol. 2016, 175, 623–631. [Google Scholar] [CrossRef] [Green Version]
  232. You, D.; Nilsson, E.; Tenen, D.E.; Lyubetskaya, A.; Lo, J.C.; Jiang, R.; Deng, J.; Dawes, B.A.; Vaag, A.; Ling, C.; et al. Dnmt3a is an epigenetic mediator of adipose insulin resistance. Elife 2017, 6, e30766. [Google Scholar] [CrossRef]
  233. Badeanlou, L.; Furlan-Freguia, C.; Yang, G.; Ruf, W.; Samad, F. Tissue factor–protease-activated receptor 2 signaling promotes diet-induced obesity and adipose inflammation. Nat. Med. 2011, 17, 1490–1497. [Google Scholar] [CrossRef]
  234. Alleman, R.J.; Katunga, L.A.; Nelson, M.A.; Brown, D.A.; Anderson, E.J. The “Goldilocks Zone” from a redox perspective—Adaptive vs. deleterious responses to oxidative stress in striated muscle. Front. Physiol. 2014, 5, 358. [Google Scholar] [CrossRef] [Green Version]
  235. Wang, X.L.; Yang, T.B.; Wei, J.; Lei, G.H.; Zeng, C. Association between serum selenium level and type 2 diabetes mellitus: A non-linear dose–response meta-analysis of observational studies. Nutr. J. 2015, 15, 48. [Google Scholar] [CrossRef] [Green Version]
  236. Olsson, M.; Olsson, B.; Jacobson, P.; Thelle, D.S.; Björkegren, J.; Walley, A.; Froguel, P.; Carlsson, L.M.; Sjöholm, K. Expression of the selenoprotein S (SELS) gene in subcutaneous adipose tissue and SELS genotype are associated with metabolic risk factors. Metabolism 2011, 60, 114–120. [Google Scholar] [CrossRef]
  237. Costa-Junior, J.M.; Ferreira, S.M.; Protzek, A.O.; Santos, G.J.; Cappelli, A.P.; Silveira, L.R.; Zoppi, C.; de Oliveira, C.A.; Boschero, A.C.; Carneiro, E.M.; et al. Endurance training inhibits insulin clearance and IDE expression in Swiss mice. PLoS ONE 2015, 10, e0118809. [Google Scholar] [CrossRef] [Green Version]
  238. Wei, X.; Ke, B.; Zhao, Z.; Ye, X.; Gao, Z.; Ye, J. Regulation of insulin degrading enzyme activity by obesity-associated factors and pioglitazone in liver of diet-induced obese mice. PLoS ONE 2014, 9, e95399. [Google Scholar] [CrossRef] [Green Version]
  239. Sun, R.; Wu, Y.; Hou, W.; Sun, Z.; Wang, Y.; Wei, H.; Mo, W.; Yu, M. Bromodomain-containing protein 2 induces insulin resistance via the mTOR/Akt signaling pathway and an inflammatory response in adipose tissue. Cell. Signal. 2017, 30, 92–103. [Google Scholar] [CrossRef]
  240. Briggs, D.B.; Giron, R.M.; Malinowski, P.R.; Nuñez, M.; Tsao, T.S. Role of redox environment on the oligomerization of higher molecular weight adiponectin. BMC Biochem. 2011, 12, 24. [Google Scholar] [CrossRef] [Green Version]
  241. Huang-Doran, I.; Bicknell, L.S.; Finucane, F.M.; Rocha, N.; Porter, K.M.; Tung, Y.L.; Szekeres, F.; Krook, A.; Nolan, J.J.; O’Driscoll, M.; et al. Genetic defects in human pericentrin are associated with severe insulin resistance and diabetes. Diabetes 2011, 60, 925–935. [Google Scholar] [CrossRef] [Green Version]
  242. Vazirani, R.P.; Verma, A.; Sadacca, L.A.; Buckman, M.S.; Picatoste, B.; Beg, M.; Torsitano, C.; Bruno, J.H.; Patel, R.T.; Simonyte, K.; et al. Disruption of adipose Rab10-dependent insulin signaling causes hepatic insulin resistance. Diabetes 2016, 65, 1577–1589. [Google Scholar] [CrossRef] [Green Version]
  243. McCarthy, J.J.; Somji, A.; Weiss, L.A.; Steffy, B.; Vega, R.; Barrett-Connor, E.; Talavera, G.; Glynne, R. Polymorphisms of the scavenger receptor class B member 1 are associated with insulin resistance with evidence of gene by sex interaction. J. Clin. Endocrinol. Metab. 2009, 94, 1789–1796. [Google Scholar] [CrossRef] [Green Version]
  244. Vardarlı, A.T.; Harman, E.; Çetintaş, V.B.; Kayıkçıoğlu, M.; Vardarlı, E.; Zengi, A.; Küçükaslan, A.Ş.; Eroğlu, Z. Polymorphisms of lipid metabolism enzyme-coding genes in patients with diabetic dyslipidemia. Anatol. J. Cardiol. 2017, 17, 313. [Google Scholar] [CrossRef]
  245. Virtanen, P.; Gommers, R.; Oliphant, T.E.; Haberland, M.; Reddy, T.; Cournapeau, D.; Burovski, E.; Peterson, P.; Weckesser, W.; Bright, J.; et al. SciPy 1.0: Fundamental algorithms for scientific computing in Python. Nat. Methods 2020, 17, 261–272. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Molecular network associated with hyperglycemia.
Figure 1. Molecular network associated with hyperglycemia.
Ijms 23 07247 g001
Figure 2. Molecules in the hyperglycemia-associated network.
Figure 2. Molecules in the hyperglycemia-associated network.
Ijms 23 07247 g002
Figure 3. Types of associations between genes and HG in the hyperglycemia network.
Figure 3. Types of associations between genes and HG in the hyperglycemia network.
Ijms 23 07247 g003
Figure 4. Positive feedback loop that involves HMGB1 in ACE2-related network. The red arrows correspond to the up-regulation and the blue arrow corresponds to the down-regulation.
Figure 4. Positive feedback loop that involves HMGB1 in ACE2-related network. The red arrows correspond to the up-regulation and the blue arrow corresponds to the down-regulation.
Ijms 23 07247 g004
Figure 5. The feedback loop in DPP4-related network involves TNF and PPAR-γ. The red arrows correspond to the up-regulation, the blue arrow corresponds to the down-regulation and the grey arrow corresponds to the association.
Figure 5. The feedback loop in DPP4-related network involves TNF and PPAR-γ. The red arrows correspond to the up-regulation, the blue arrow corresponds to the down-regulation and the grey arrow corresponds to the association.
Ijms 23 07247 g005
Figure 6. The regulation loop in the TMPRSS2-related network involves AR. The red arrows correspond to the up-regulation and the blue arrow corresponds to the down-regulation.
Figure 6. The regulation loop in the TMPRSS2-related network involves AR. The red arrows correspond to the up-regulation and the blue arrow corresponds to the down-regulation.
Ijms 23 07247 g006
Figure 7. The regulation loop in the CTSB-related network involves CASP8, IL6, CXCL8, SP1, TLR4, TNF, STAT3, and PRL. The red arrows correspond to the up-regulation and the blue arrow corresponds to the down-regulation.
Figure 7. The regulation loop in the CTSB-related network involves CASP8, IL6, CXCL8, SP1, TLR4, TNF, STAT3, and PRL. The red arrows correspond to the up-regulation and the blue arrow corresponds to the down-regulation.
Ijms 23 07247 g007
Figure 8. The regulation loop in the CTSL-related network involves FGF2, IL6, FOXO1, HPSE, JUN, and MAPK1.
Figure 8. The regulation loop in the CTSL-related network involves FGF2, IL6, FOXO1, HPSE, JUN, and MAPK1.
Ijms 23 07247 g008
Figure 9. Gene network of human proteins targeted by SARS-CoV-2.
Figure 9. Gene network of human proteins targeted by SARS-CoV-2.
Ijms 23 07247 g009
Figure 10. Molecules in the network of human proteins targeted by SARS-CoV-2.
Figure 10. Molecules in the network of human proteins targeted by SARS-CoV-2.
Ijms 23 07247 g010
Figure 11. Interactions between hyperglycemia-associated network and network of SARS-CoV-2-targeted proteins contribute to severe COVID-19 in patients with diabetes.
Figure 11. Interactions between hyperglycemia-associated network and network of SARS-CoV-2-targeted proteins contribute to severe COVID-19 in patients with diabetes.
Ijms 23 07247 g011
Figure 12. Venn diagram of intersection of the lists of SARS-CoV-2-targeted proteins and gene/protein sets associated with cardiovascular disease, diabetic neuropathy, diabetic nephropathy, and diabetic retinopathy. Numbers show the gene/protein count.
Figure 12. Venn diagram of intersection of the lists of SARS-CoV-2-targeted proteins and gene/protein sets associated with cardiovascular disease, diabetic neuropathy, diabetic nephropathy, and diabetic retinopathy. Numbers show the gene/protein count.
Ijms 23 07247 g012
Figure 13. Flowchart of the study.
Figure 13. Flowchart of the study.
Ijms 23 07247 g013
Table 1. Top 10 GO biological processes overrepresented for hyperglycemia-associated genes found by DAVID (p-values with FDR correction < 0.05).
Table 1. Top 10 GO biological processes overrepresented for hyperglycemia-associated genes found by DAVID (p-values with FDR correction < 0.05).
Gene Ontology Biological ProcessGene Ontology IDGenesp-Values with FDR Correction
Glucose homeostasisGO:0042593ADIPOQ, ADIPOR1, ADRA2A, CEBPA, CNR1, FBN1, G6PC1, GCGR, GCK, HIF1A, HNF1A, IL6, INS, IRS1, LEP, LEPR, MTNR1B, NEUROD1, NGFR, PAX6, PDK4, PDX1, POMC, PPARG, PRKAA1, PRKAA2, RBP4, SIRT6, SLC2A4, STAT3, STK11, TCF7L27.92 × 10−23
Inflammatory responseGO:0006954AGER, AOC3, CALCA, CCL11, CCL2, CD40LG, CRH, CRP, CXCL12, CXCL8, CXCR4, CYBB, ECM1, F2R, FOS, FPR1, HMGB1, IL10, IL13, IL18, IL19, IL1A, IL1B, IL22, IL6, NFATC3, NFE2L2, NGFR, NLRP3, NOX4, PIK3CD, PIK3CG, PRKD1, PTGER2, PTGS2, RAC1, RELA, SELE, SELP, SPP1, TBXA2R, TGFB1, THBS1, TLR2, TLR4, TNF, TNFRSF11B6.69 × 10−17
Response to hypoxiaGO:0001666ADIPOQ, ADM, AGER, ANGPT2, ANGPTL4, CASP1, CASP3, CAT, CAV1, CCL2, CDKN1B, CREB1, CXCL12, CXCR4, DPP4, EGR1, EPO, HIF1A, HMOX1, HSPD1, LEP, MB, MMP2, NOS1, NOS2, NOX4, PLAT, PLAU, PPARA, PRKAA1, PRKCB, RYR2, SOD2, TGFB1, THBS1, TLR2, UCP2, VCAM1, VEGFA, VEGFB1.41 × 10−23
Positive regulation of angiogenesisGO:0045766ADM, ANGPT2, ANGPTL4, CCL11, CX3CL1, CXCL8, CYBB, DDAH1, ECM1, F3, FGF2, GATA4, HGF, HIF1A, HMOX1, IL1A, IL1B, KDR, NFE2L2, NOS3, PRKCB, PRKD1, PTGIS, SERPINE1, SIRT1, TBXA2R, THBS1, VEGFA9.5 × 10−17
Positive regulation of cell proliferationGO:0008284ADM, ADRA2A, AR, ATF3, AVP, AVPR1A, BCL2, CCK, CCN2, CCND2, CD47, CDK2, CDKN1B, CRH, CTF1, DPP4, EDN1, EGFR, EIF5A, EPO, ESM1, F2R, FABP1, FGF2, FGF21, FN1, GDNF, GHRH, HGF, IFNG, IGF1, IGF1R, IGF2, IL2, IL6, INS, IRS1, IRS2, KDR, LEP, MAPK1, MYC, NAMPT, NOTCH1, NRG1, NTN1, PDX1, PRKAA1, PTEN, REG1A, RELA, S100B, SIRT1, STAT3, TGFB1, THBS1, VEGFA, VIP2.86 × 10−21
Negative regulation of apoptotic processGO:0043066ALB, ANGPT1, ANGPTL4, AVP, BCL2, CASP3, CAT, CCND2, CD40LG, CD44, CDKN1A, CDKN1B, DDAH2, EGFR, FABP1, FOXO1, GAS6, GCG, GDNF, GLO1, GSK3B, HSPD1, IGF1, IGF1R, IL10, IL2, IL4, IL6, KDR, LEP, LTF, MAPK7, MMP9, MPZ, MYC, NGF, NGFR, NQO1, PAX4, PIK3R1, PRKAA1, PRKAA2, PRNP, PTEN, RELA, SIRT1, SNCA, SOCS3, SOD2, STAT3, THBS1, TP53, UCP2, VEGFA, VEGFB1.69 × 10−19
Positive regulation of protein kinase B signalingGO:0051897ANGPT1, CD28, EGFR, F3, FGF2, GAS6, GPX1, HPSE, IGF2, IL18, IL6, INS, LEP, MTOR, NOX4, NRG1, PIK3CG, RICTOR, TCF7L2, TGFB1, THBS1, TNF, TXN, VEGFB8.49 × 10−16
Positive regulation of transcription from RNA polymerase II promoterGO:0045944APP, AR, ARNTL, ATF3, CD28, CEBPA, CREB1, CREM, CTNNB1, DCN, DDIT3, EDN1, EGFR, EGR1, FGF2, FOS, FOXO1, FOXO3, GALR1, GATA4, GDNF, GSK3B, HGF, HIF1A, HMGA1, HMGB1, HNF1A, IFNG, IGF1, IL10, IL18, IL1A, IL1B, IL2, IL4, IL6, JUN, MAFA, MAFB, MAPK7, MEF2A, MEF2C, MEN1, MYC, NAMPT, NCK1, NEUROD1, NEUROG3, NFAT5, NFATC3, NFE2L2, NLRP3, NOS1, NOTCH1, NR1H2, NRG1, OGT, PARP1, PAX3, PAX6, PDX1, PIK3R1, POMC, PPARA, PPARG, PRKD1, PTH, RELA, SERPINE1, SIRT1, SIRT2, SP1, SREBF1, STAT3, TCF7L2, TGFB1, TLR2, TLR4, TNF, TP53, VEGFA2.25 × 10−19
AgingGO:0007568ADM, ADRB3, AGT, ARG1, CALCA, CAT, CCL2, CCN2, CNR1, COL3A1, CREB1, DCN, EPO, FGF2, FOS, FOXO3, IGFBP1, IL10, IL6, JUN, KL, NFE2L2, NQO1, PTEN, RELA, RETN, SERPINF1, SNCA, SOD1, SREBF1, STAT3, TGFB1, UCP2, VCAM12.47 × 10−18
Response to drugGO:0042493ABCA1, ABCC8, APOA1, ARG1, BCL2, BGLAP, CASP3, CAT, CCND1, CDH1, CDH3, CDKN1A, CDKN1B, CREB1, CRH, CTNNB1, CYBB, DUSP6, FOS, GATA4, GIP, HSPD1, ICAM1, IFNG, IL10, IL4, IL6, JUN, KCNJ11, LCN2, LPL, MYC, NEUROD1, PAX4, PDX1, PPARG, PTEN, PTGS2, PTH, RELA, SMPD1, SNCA, SOD1, SOD2, SORD, SREBF1, SST, STAT3, TBXA2R, TGFB1, THBS1, TIMP4, TNFRSF11B, TXNIP, VEGFB1.44 × 10−27
Table 2. Types of associations of the genes/proteins from hyperglycemia-related and ACE2-related networks with high glucose (HG) and ACE2.
Table 2. Types of associations of the genes/proteins from hyperglycemia-related and ACE2-related networks with high glucose (HG) and ACE2.
HGGene Expression is Upregulated by HGGene Expression is Downregulated by HGMolecules with Hyperglycemic ActivityMolecules with Antihyperglycemic EffectOther Relations
ACE2
Molecules are upregulated by ACE2BCL2, CCND1, MMP2, NOS1, NOS3, SOD1, UCP2BCL2, CDH1, NOS3IL1B, NOS2 NPHS1, SIRT6
Molecules are downregulated by ACE2ANGPT2, CCL2, CCN2, HMGB1, ICAM1, MIR21, MMP9, STAT3, VCAM1VEGFAAGTR1STAT3ACE, ICAM1
Molecules upregulating ACE2HMGB1 INS
Molecules downregulating ACE2EDN1SIRT1AGTR1INS, SIRT1ACE, ALB, APOE
Other relationsAGTCAT, IRS1GCG CALM1
Table 3. The most overrepresented GO biological processes that are common for the sets of genes linked with ACE2 and associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, and insulin resistance.
Table 3. The most overrepresented GO biological processes that are common for the sets of genes linked with ACE2 and associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, and insulin resistance.
Gene Ontology Biological ProcessGene Ontology IDp-Values with FDR Correction
HyperglycemiaCVDDiabetic NeuropathyDiabetic NephropathyDiabetic RetinopathyInsulin Resistance
Positive regulation of cell migrationGO:00303353.28 × 10−59.10 × 10−41.11 × 10−53.60 × 10−57.24 × 10−63.46 × 10−4
Negative regulation of gene expressionGO:00106296.51 × 10−72.68 × 10−70.01556.72 × 10−91.93 × 10−86.51 × 10−7
Positive regulation of vascular smooth muscle cell proliferationGO:19047071.28 × 10−43.12 × 10−40.01554.46 × 10−42.53 × 10−40.0213
Positive regulation of phosphatidylinositol 3-kinase signalingGO:00140683.28 × 10−50.01210.02771.62 × 10−48.03 × 10−58.51 × 10−4
Positive regulation of cell proliferationGO:00082841.45 × 10−41.68 × 10−40.04944.29 × 10−40.00325.49 × 10−5
Negative regulation of apoptotic processGO:00430661.32 × 10−40.00280.04896.47 × 10−50.01224.01 × 10−5
Response to hypoxiaGO:00016665.05 × 10−78.51 × 10−7 1.20 × 10−96.91 × 10−74.30 × 10−8
Response to lipopolysaccharideGO:00324963.28 × 10−53.43 × 10−7 4.43 × 10−93.81 × 10−71.82 × 10−8
Nitric oxide mediated signal transductionGO:00072636.11 × 10−61.63 × 10−7 2.08 × 10−53.63 × 10−41.85 × 10−6
Positive regulation of vascular endothelial cell proliferationGO:19055644.15 × 10−43.52 × 10−5 1.48 × 10−62.49 × 10−51.70 × 10−4
Table 4. Types of associations of the genes/proteins from hyperglycemia-related and DPP4-related networks with high glucose (HG) and DPP4.
Table 4. Types of associations of the genes/proteins from hyperglycemia-related and DPP4-related networks with high glucose (HG) and DPP4.
HGGene Expression Is Upregulated by HGGene Expression Is Downregulated by HGMolecules with Hyperglycemic ActivityMolecules with Antihyperglycemic EffectOther Relations
DPP4
Genes are upregulated by DPP4CD36, CD8A, CRP, IL6, MMP2, SPP1CD44, HIF1A, VEGFA PPARGPLAT
Genes are downregulated by DPP4CCL11, FGF2, HMGB1, MMP9, NPY, THBS1, VIPPPYCREB1, EGR1, GCG, GIPADIPOQ, EPO, GLP1R, INS, SERPINF1CXCL12, GHRH, NPHS1
Molecules that upregulate DPP4CCL11, EGFR, IFNG, TNF INSIL2, IL13
Molecules that downregulate DPP4NPY, TLR4 MYCPTH, TFPI
Other relationsCXCR4, FN1CDH1GCG, NOS2CAV1, KLHNF1A, LGALS3
Table 5. Most overrepresented GO biological processes that are common for the sets of genes linked with DPP4 and are associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, insulin resistance, and beta-cell dysfunction.
Table 5. Most overrepresented GO biological processes that are common for the sets of genes linked with DPP4 and are associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, insulin resistance, and beta-cell dysfunction.
Gene Ontology Biological ProcessGene Ontology IDp-Values with FDR Correction
HyperglycemiaCVDDiabetic NeuropathyDiabetic NephropathyDiabetic RetinopathyInsulin ResistanceBeta-Cell Dysfunction
Response to hypoxiaGO:00016663.68 × 10−132.00 × 10−71.50 × 10−58.14 × 10−131.93 × 10−93.71 × 10−10
Positive regulation of ERK1 and ERK2 cascadeGO:00703741.78 × 10−97.39 × 10−53.06 × 10−62.13 × 10−101.14 × 10−81.10 × 10−10
Positive regulation of smooth muscle cell proliferationGO:00486611.78 × 10−92.23 × 10−41.55 × 10−41.47 × 10−62.11 × 10−72.41 × 10−4
Response to activityGO:00148231.28 × 10−88.42 × 10−51.03 × 10−47.24 × 10−65.26 × 10−49.71 × 10−4
Positive regulation of interleukin-8 productionGO:00327573.34 × 10−59.36 × 10−60.00224.08 × 10−81.48 × 10−71.20 × 10−6
AgingGO:00075682.59 × 10−40.00161.94 × 10−45.18 × 10−55.14 × 10−61.87 × 10−4
Positive regulation of phosphatidylinositol 3-kinase signalingGO:00140680.00124.54 × 10−41.94 × 10−41.09 × 10−85.43 × 10−75.45 × 10−43.07 × 10−4
Negative regulation of lipid storageGO:00108888.41 × 10−55.31 × 10−60.00224.58 × 10−61.71 × 10−62.52 × 10−5
Cellular response to lipopolysaccharideGO:00712222.59 × 10−42.00 × 10−70.00227.19 × 10−75.14 × 10−66.64 × 10−8
Acute-phase responseGO:00069531.51 × 10−46.18 × 10−48.12 × 10−44.08 × 10−58.33 × 10−60.0039
Table 6. Types of associations of the genes/proteins from hyperglycemia-related and cathepsin B (CTSB)-related networks with high glucose (HG) and cathepsin B (STSB).
Table 6. Types of associations of the genes/proteins from hyperglycemia-related and cathepsin B (CTSB)-related networks with high glucose (HG) and cathepsin B (STSB).
HGGene Expression Is Upregulated by HGGene Expression Is Downregulated by HGMolecules with Hyperglycemic ActivityMolecules with Antihyperglycemic EffectOther Relations
CTSB
Genes are upregulated by cathepsin BBAX, BCL2, BDNF, CASP1, CASP3, CASP8, CCL2, CXCL8, DCX, IL18, MMP9, MTOR, NLRP3, PRL, PTENBCL2, CCK, VEGFAPRLIL4, IL18, MTORAPOE, HSPG2
Genes are downregulated by cathepsin BAPP, FN1BGLAP, SIRT1CDKN1BSIRT1
Molecules that upregulate CTSBCASP8, CXCL8, IL6, PRL, SP1, STAT3, TLR4, TNFCCK, NTN1PRLSMPD1, STAT3CXCL12, SNCA, SP1
Molecules that downregulate CTSBTGFB1VEGFA IL10
Other relationsANXA2, EGFR, HMGB1, MKI67, TP53APOA1, KDR, PLAUIL1BCAV1, HGFFOXO3
Table 7. Most overrepresented GO biological processes that are common for the sets of genes linked with CTSB and associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, insulin resistance, and beta-cell dysfunction.
Table 7. Most overrepresented GO biological processes that are common for the sets of genes linked with CTSB and associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, insulin resistance, and beta-cell dysfunction.
Gene Ontology Biological ProcessGene Ontology IDp-Values with FDR Correction
HyperglycemiaCVDDiabetic NeuropathyDiabetic NephropathyDiabetic RetinopathyInsulin ResistanceBeta-Cell Dysfunction
Positive regulation of cell proliferationGO:00082844.03 × 10−91.92 × 10−50.00112.85 × 10−106.20 × 10−91.92 × 10−111.03 × 10−4
Positive regulation of gene expressionGO:00106285.24 × 10−104.48 × 10−87.49 × 10−81.68 × 10−115.09 × 10−104.75 × 10−110.006
Positive regulation of protein phosphorylationGO:00019348.99 × 10−98.17 × 10−72.68*10−41.31 × 10−86.19 × 10−86.81 × 10−80.0062
Lipopolysaccharide-mediated signaling pathwayGO:00316632.08 × 10−64.81 × 10−66.83 × 10−42.57 × 10−55.29 × 10−63.06 × 10−70.0062
Negative regulation of apoptotic processGO:00430663.53 × 10−91.75 × 10−60.00113.36 × 10−132.88 × 10−74.87 × 10−110.006
Positive regulation of glial cell proliferationGO:00602525.50 × 10−62.71 × 10−70.00523.82 × 10−51.23 × 10−51.13 × 10−70.0031
Protein kinase B signalingGO:00434913.02 × 10−62.13 × 10−48.72 × 10−41.63 × 10−68.27 × 10−61.93 × 10−40.0074
Response to drugGO:00424931.38 × 10−91.20 × 10−60.00723.64 × 10−69.35 × 10−56.81 × 10−80.0017
Positive regulation of protein kinase B signalingGO:00518972.83 × 10−54.95 × 10−70.00852.60 × 10−81.51 × 10−96.81 × 10−80.0031
Positive regulation of interleukin-8 productionGO:00327572.09 × 10−56.03 × 10−90.0029.11 × 10−73.52 × 10−97.49 × 10−110.0139
Table 8. Types of associations of the genes/proteins from hyperglycemia-related and cathepsin L (CTSL)-related networks with high glucose (HG) and cathepsin L (STSL).
Table 8. Types of associations of the genes/proteins from hyperglycemia-related and cathepsin L (CTSL)-related networks with high glucose (HG) and cathepsin L (STSL).
HGGene Expression Is Upregulated by HGGene Expression Is Downregulated by HGMolecules with Hyperglycemic ActivityMolecules with Antihyperglycemic EffectOther Relations
CTSL
Genes are upregulated by cathepsin LBCL2, CXCL8, HPSEBCL2
Genes are downregulated by cathepsin LCDKN1A, LEPRCDH1IGFBP3 LEPR, TF
Molecules that upregulate CTSLFGF2, FOXO1, HPSE, IL6, JUN, MAPK1 INS, MYCFOS
Molecules that downregulate CTSLCDKN1A, TGFB1
Other relationsCCL2, F3, TP53PLAU POMC
Table 9. Most overrepresented GO biological processes that are common for the sets of genes linked with CTSL and associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, and insulin resistance.
Table 9. Most overrepresented GO biological processes that are common for the sets of genes linked with CTSL and associated with hyperglycemia, CVD, diabetic neuropathy, diabetic nephropathy, diabetic retinopathy, and insulin resistance.
Gene Ontology Biological ProcessGene Ontology IDp-Values with FDR Correction
HyperglycemiaCVDDiabetic NeuropathyDiabetic NephropathyDiabetic RetinopathyInsulin Resistance
Positive regulation of gene expressionGO:00106287.19 × 10−60.00120.0037.98 × 10−41.25 × 10−44.44 × 10−4
ChemotaxisGO:00069358.06 × 10−40.0033 0.00210.00270.0146
Positive regulation of protein phosphorylationGO:0001934 0.01670.01151.25 × 10−45.80 × 10−4
Positive regulation of MAPK cascadeGO:00434100.01650.00690.0120.0064.87 × 10−50.0086
Negative regulation of cell proliferationGO:00082855.96 × 10−50.04140.01236.86 × 10−4 0.0041
Positive regulation of protein kinase B signalingGO:00518978.49 × 10−40.023 0.01842.07 × 10−40.0163
Positive regulation of cell migrationGO:00303350.00580.02710.026 0.0027
Lipopolysaccharide-mediated signaling pathwayGO:00316630.01730.0128 0.0103 0.033
Positive regulation of protein import into nucleusGO:00423070.01950.0167 7.98 × 10−4 0.0438
Positive regulation of production of miRNAs involved in gene silencing by miRNAGO:19038000.00340.02710.00390.0228 0.048
Table 10. Overrepresented GO biological processes for SARS-CoV-2-targeted human proteins found by DAVID (p-values < 0.05 with FDR correction).
Table 10. Overrepresented GO biological processes for SARS-CoV-2-targeted human proteins found by DAVID (p-values < 0.05 with FDR correction).
Gene Ontology Biological ProcessGene Ontology IDGenesp-Values with FDR Correction
Viral processGO:0016032BRD4, CCDC86, CRTC3, CUL2, EIF4H, ELOC, MFGE8, NLRX1, NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, POLA1, RAE1, RALA, RBX1, RHOA0.0011
Intracellular transport of virusGO:0075733NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, RAE10.0079
Protein transportGO:0015031AKAP8, ARF6, CENPF, CHMP2A, ERC1, GORASP1, HOOK1, JAKMIP1, LMAN2, NUP210, PLEKHF2, PPT1, RAB14, RAB18, RAB2A, RAB5C, RAB7A, TIMM10B, TIMM8B, TIMM9, TMED5, WASHC4, YIF1A0.0013
Protein foldingGO:0006457BAG5, CSNK2A2, CSNK2B, CWC27, DNAJC19, ERO1B, ERP44, FKBP10, FKBP15, FKBP7, GNB1, GRPEL1, MOGS, PPIL3, QSOX2, SIL1, TBCA4.05 × 10−4
Regulation of glucose transportGO:0010827NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, RAE17.74 × 10−4
Mitotic nuclear envelope disassemblyGO:0007077NEK9, NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, RAE17.74 × 10−4
tRNA export from nucleusGO:0006409NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, RAE17.74 × 10−4
Regulation of cellular response to heatGO:1900034BAG5, HSBP1, NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, RAE10.0022
G2/M transition of mitotic cell cycleGO:0000086AKAP9, CDK5RAP2, CEP135, CEP250, CEP43, CIT, CNTRL, NINL, PCNT, PRKACA, PRKAR2B, RAB8A0.0087
Protein heterotrimerizationGO:0070208COL6A1, GNB1, NUP54, NUP58, NUP620.0142
mRNA export from nucleusGO:0006406NUP210, NUP214, NUP54, NUP58, NUP62, NUP88, NUP98, RAE1, SLU7, UPF10.0142
U4 snRNA 3’-end processingGO:0034475EXOSC2, EXOSC3, EXOSC5, EXOSC80.0387
Chaperone-mediated protein transportGO:0072321TIMM10, TIMM8B, TIMM9, TOR1A0.0387
Protein targeting to mitochondrionGO:0006626DNAJC19, TIMM10, TIMM10B, TIMM8B, TIMM9, TOMM700.0418
Nuclear-transcribed mRNA catabolic process, exonucleolytic, 3’-5’GO:0034427EXOSC2, EXOSC3, EXOSC5, EXOSC80.0497
Table 11. Most overrepresented GO biological processes for the sets of genes linked with SARS-CoV-2-targeted human proteins and associated with CVD, diabetic neuropathy, diabetic nephropathy, and diabetic retinopathy.
Table 11. Most overrepresented GO biological processes for the sets of genes linked with SARS-CoV-2-targeted human proteins and associated with CVD, diabetic neuropathy, diabetic nephropathy, and diabetic retinopathy.
Gene Ontology Biological ProcessGene Ontology IDp-Values with FDR Correction
CVDDiabetic NeuropathyDiabetic NephropathyDiabetic Retinopathy
Cytokine-mediated signaling pathwayGO:00192213.79 × 10−231.92 × 10−114.53 × 10−372.35 × 10−20
Response to hypoxiaGO:00016663.16 × 10−167.16 × 10−103.50 × 10−275.42 × 10−25
Positive regulation of gene expressionGO:00106281.25 × 10−181.71 × 10−91.59 × 10−302.56 × 10−24
Positive regulation of phosphatidylinositol 3-kinase signalingGO:00140684.48 × 10−171.89 × 10−82.91 × 10−247.80 × 10−22
Inflammatory responseGO:00069547.23 × 10−25 1.53 × 10−333.86 × 10−26
Positive regulation of cell proliferationGO:0008284 1.86 × 10−86.54 × 10−251.09 × 10−22
Positive regulation of smooth muscle cell proliferationGO:00486611.01 × 10−162.09 × 10−88.79 × 10−23
Positive regulation of protein kinase B signalingGO:00518972.57 × 10−16 4.87 × 10−23
AgingGO:00075681.14 × 10−169.23 × 10−11
Positive regulation of ERK1 and ERK2 cascadeGO:0070374 6.15 × 10−96.70 × 10−24
Negative regulation of apoptotic processGO:0043066 4.70 × 10−28
Positive regulation of angiogenesisGO:0045766 1.47 × 10−25
Cellular response to lipopolysaccharideGO:00712223.00 × 10−19
Positive regulation of protein phosphorylationGO:0001934 4.30 × 10−19
Positive regulation of cell migrationGO:0030335 5.16 × 10−19
Response to xenobiotic stimulusGO:0009410 3.10 × 10−17
Regulation of blood pressureGO:00082173.16 × 10−16
Positive regulation of peptidyl-tyrosine phosphorylationGO:0050731 1.71 × 10−9
Positive regulation of apoptotic processGO:0043065 6.15 × 10−9
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Saik, O.V.; Klimontov, V.V. Gene Networks of Hyperglycemia, Diabetic Complications, and Human Proteins Targeted by SARS-CoV-2: What Is the Molecular Basis for Comorbidity? Int. J. Mol. Sci. 2022, 23, 7247. https://doi.org/10.3390/ijms23137247

AMA Style

Saik OV, Klimontov VV. Gene Networks of Hyperglycemia, Diabetic Complications, and Human Proteins Targeted by SARS-CoV-2: What Is the Molecular Basis for Comorbidity? International Journal of Molecular Sciences. 2022; 23(13):7247. https://doi.org/10.3390/ijms23137247

Chicago/Turabian Style

Saik, Olga V., and Vadim V. Klimontov. 2022. "Gene Networks of Hyperglycemia, Diabetic Complications, and Human Proteins Targeted by SARS-CoV-2: What Is the Molecular Basis for Comorbidity?" International Journal of Molecular Sciences 23, no. 13: 7247. https://doi.org/10.3390/ijms23137247

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop