Skip to main content
Erschienen in: Journal of Translational Medicine 1/2024

Open Access 01.12.2024 | Research

Deciphering the omicron variant: integrated omics analysis reveals critical biomarkers and pathophysiological pathways

verfasst von: Qianyue Yang, Zhiwei Lin, Mingshan Xue, Yueting Jiang, Libing Chen, Jiahong Chen, Yuhong Liao, Jiali Lv, Baojun Guo, Peiyan Zheng, Huimin Huang, Baoqing Sun

Erschienen in: Journal of Translational Medicine | Ausgabe 1/2024

Abstract

Background

The rapid emergence and global dissemination of the Omicron variant of SARS-CoV-2 have posed formidable challenges in public health. This scenario underscores the urgent need for an enhanced understanding of Omicron's pathophysiological mechanisms to guide clinical management and shape public health strategies. Our study is aimed at deciphering the intricate molecular mechanisms underlying Omicron infections, particularly focusing on the identification of specific biomarkers.

Methods

This investigation employed a robust and systematic approach, initially encompassing 15 Omicron-infected patients and an equal number of healthy controls, followed by a validation cohort of 20 individuals per group. The study's methodological framework included a comprehensive multi-omics analysis that integrated proteomics and metabolomics, augmented by extensive bioinformatics. Proteomic exploration was conducted via an advanced Ultra-High-Performance Liquid Chromatography (UHPLC) system linked with mass spectrometry. Concurrently, metabolomic profiling was executed using an Ultra-Performance Liquid Chromatography (UPLC) system. The bioinformatics component, fundamental to this research, entailed an exhaustive analysis of protein–protein interactions, pathway enrichment, and metabolic network dynamics, utilizing state-of-the-art tools such as the STRING database and Cytoscape software, ensuring a holistic interpretation of the data.

Results

Our proteomic inquiry identified eight notably dysregulated proteins (THBS1, ACTN1, ACTC1, POTEF, ACTB, TPM4, VCL, ICAM1) in individuals infected with the Omicron variant. These proteins play critical roles in essential physiological processes, especially within the coagulation cascade and hemostatic mechanisms, suggesting their significant involvement in the pathogenesis of Omicron infection. Complementing these proteomic insights, metabolomic analysis discerned 146 differentially expressed metabolites, intricately associated with pivotal metabolic pathways such as tryptophan metabolism, retinol metabolism, and steroid hormone biosynthesis. This comprehensive metabolic profiling sheds light on the systemic implications of Omicron infection, underscoring profound alterations in metabolic equilibrium.

Conclusions

This study substantially enriches our comprehension of the physiological ramifications induced by the Omicron variant, with a particular emphasis on the pivotal roles of coagulation and platelet pathways in disease pathogenesis. The discovery of these specific biomarkers illuminates their potential as critical targets for diagnostic and therapeutic strategies, providing invaluable insights for the development of tailored treatments and enhancing patient care in the dynamic context of the ongoing pandemic.
Hinweise

Supplementary Information

The online version contains supplementary material available at https://​doi.​org/​10.​1186/​s12967-024-05022-z.
Qianyue Yang, Zhiwei Lin, and Mingshan Xue have contributed equally to this work.

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Abkürzungen
SARS-CoV-2
Severe acute respiratory syndrome coronavirus 2
VOC
Variant of concern
PLT
Platelets
WHO
The World Health Organization
ACE2
Angiotensin-converting enzyme 2
TMPRSS2
Transmembrane serine protease 2
COVID-19
Corona Virus Disease 2019
UHPLC
Ultra-high-performance liquid chromatography
UPLC
Ultra-performance liquid chromatography
DEPs
Differentially expressed proteins
DEMs
Differentially expressed metabolites
Log2FC
Log2FoldChange
GO
Gene ontology
KEGG
Kyoto encyclopedia of genes and genomes
PPI
Protein–protein interaction networks
OPLS-DA
Orthogonal partial least squares-discriminant analysis
VIP
Variable importance in projection
FDR
False discovery rate
IQR
Interquartile range
ROC
Receiver operating characteristic
AUC
The area under the curve
WBC
White blood cell
NEU
Neutrophil
LYM
Lymphocyte
MONO
Monocyte
EOS
Eosinophils
BASO
Basophil
PT
Prothrombin time
INR
International normalized ratio
PTTA
Prothrombin activity
FIB
Fibrinogen
APTT
Activated partial thromboplastin time
TT
Thrombin
DD
D-Dimer
CRP
C-reactive protein
SAA
Serumamyloid A
PCT
Procalcitonin
BP
Biological process
CC
Cellular component
MF
Molecular function
PCA
Principal component analysis
95% CI
95% Confidence interval
I3A
Indole-3-acetate
THBS1
Thrombospondin 1
ACTN1
Alpha-actinin 1
ACTC1
Actin alpha cardiac muscle 1
POTEF
POTE ankyrin domain family member F
ACTB
Actin beta
TPM4
Tropomyosin 4
VCL
Vinculin
ICAM1
Intercellular adhesion molecule 1
PARK7
Parkinsonism associated deglycase
YWHAB
Tyrosine 3-Monooxygenase/Tryptophan 5-Monooxygenase Activation Protein Beta

Introduction

In November 2021, amidst the ongoing global pandemic of the novel coronavirus (SARS-CoV-2), South African scientists identified a new variant of concern (VOC), Omicron, which was promptly classified as such by the World Health Organization (WHO) on November 26, 2021 [1]. Despite increasing vaccination rates, Omicron and its subvariants continue to spread rapidly worldwide [2]. Compared to the early wild-type virus, Omicron exhibits a higher transmissibility and immune evasion capability, posing a significant public health challenge [3]. Studies have shown that both Omicron and the Delta variant can evade host immune responses through single-point mutations on the spike protein [4], with Omicron having a stronger binding affinity to receptors, thus enhancing its potential for immune evasion [5]. Although Omicron may be less pathogenic, it still poses a lethal threat to immunocompromised individuals or patients undergoing immunosuppressive therapy. The pathogenesis of Omicron is increasingly evidenced to be closely associated with cytokine storms [6], systemic inflammatory responses [6], and platelet count abnormalities [7]. The mechanisms related to platelet counts remain inadequately explained in the context of SARS-CoV-2 infection.
Platelets (PLT), key cellular components in maintaining vascular integrity and initiating hemostasis, are produced by megakaryocytes in the bone marrow and circulate in the blood as cell fragments [8]. They play a crucial role in the development, progression, and ultimate outcome of many diseases, intimately associated with platelet-mediated hemostasis and thrombosis, which may trigger adverse inflammatory responses [9]. Platelets can release serotonin and participate in immune responses by promoting T-cell activation, thus modulating immune reactions [10]. Their inherent adhesiveness allows them to interact with a variety of cells from both the innate and adaptive immune systems [11]. Studies, including that by Sevilya et al., have shown a direct link between platelet activation and SARS-CoV-2 infection [12], and it has been suggested that the expression of the angiotensin-converting enzyme 2 (ACE2) receptor and the transmembrane serine protease transmembrane serine protease 2 (TMPRSS2) on platelets and megakaryocytes play a role in mediating viral infection [13, 14]. Moreover, the functional heterogeneity of platelet aggregation is thought to play a role in the pathogenesis of Omicron infection, contributing to the regulation of immune function within the hematological ecosystem [15].
While current research on COVID-19 pathogenesis and the coagulation/hemostasis pathways are ongoing, most studies still focus on the perspective of biomarkers [16, 17], and there are few studies on Omicron patients. This study aims to delve into the molecular interactions among key differential proteins of the Omicron variant, to identify potential targets for intervention, and to further analyze the metabolomic characteristics in the peripheral blood of Omicron patients. The urgency of this research stems from the necessity to understand the intricate interplay between the virus's pathogenesis and the host's immune response. Given the global impact of Omicron, elucidating these mechanisms is not only of scientific interest but is imperative for the development of targeted therapies and for informing public health strategies.

Methodology

Biospecimen collection and clinical data acquisition

The study systematically collected biospecimens and clinical data across two distinct phases: screening and validation, ensuring a robust analytical framework. In the screening phase, 15 patients diagnosed with Omicron variant infections via nucleic acid testing were recruited from the First Affiliated Hospital of Guangzhou Medical University. Diagnoses were established according to the latest COVID-19 Omicron diagnostic and treatment guidelines released by the World Health Organization, coupled with clinical symptoms and pharyngeal swab nucleic acid test results. Additionally, 15 healthy volunteers, matched by age and gender, were selected as controls. Comprehensive demographic, clinical, and laboratory data were compiled for both groups, followed by serum collection for in-depth proteomic and metabolomic analyses. The validation phase further substantiated our initial findings through an expanded cohort of 20 Omicron patients and 20 controls, employing identical proteomics methodologies and Receiver operating characteristic (ROC) analyses to ensure the reproducibility and validity of our screening phase outcomes. Informed consent was obtained from all participants, and the study protocol was approved by the Ethics Committee of the First Affiliated Hospital of Guangzhou Medical University (Ethical Approval Numbers: 202001134 and 202115202). A schematic representation of the study design can be found in Fig. 1.

Proteomic analysis

Upon thawing, serum samples were treated with methanol/acetonitrile (1:1, volume ratio, Sigma-Aldrich, St. Louis, MO, USA) to achieve a total volume ratio of 2:2:1 (excluding water). The samples were vortexed on ice for 30 s, subjected to high-intensity ultrasonication for 10 min, and then left to stand at -20 °C for one hour. Subsequently, the samples were centrifuged at 4 °C (12,000g, 15 min), and the supernatant was collected and dried at room temperature. The dried samples were reconstituted in acetonitrile/water (1:1, volume ratio, Sigma-Aldrich, St. Louis, MO, USA), vortexed and ultrasonicated again, and centrifuged to remove any precipitate. The supernatant was stored at −80 °C until analysis. The samples were resolved in solvent A (0.1% formic acid in 2% acetonitrile/water, Sigma-Aldrich, St. Louis, MO, USA) and separated using a homemade reverse-phase liquid chromatography column (25 cm length, 75/100 μm inner diameter). Chromatographic separation was carried out on a nanoElute Ultra-High-Performance Liquid Chromatography (UHPLC) system (Bruker Daltonics, Billerica, MA, USA) at a flow rate of 450 nL/min, followed by electrospray ionization through a capillary source and mass spectrometric analysis using a timsTOF Pro (Bruker Daltonics, Billerica, MA, USA) in PASEF mode with a dynamic exclusion time of 30 s.

Metabolomic analysis

Metabolite separation was conducted using a Waters ACQUITY UltraPerformance Liquid Chromatography (Waters ACQUITY UPLC) System (Waters Corporation, Milford, MA, USA) equipped with a BEH C18 column. Chromatographic conditions included a 10 µL injection volume, a flow rate of 400 µL/min, and a column temperature maintained at 40 °C. Metabolomics data were processed using MassLynx (Waters Corporation, Milford, MA, USA), which entailed peak extraction, alignment, and retention time correction, ensuring that the mass accuracy was controlled within 20 ppm to guarantee data accuracy.

Bioinformatics analysis

Proteomics data were analyzed using the limma package in R software (version 3.6.3) to identify differentially expressed proteins (DEPs) with p < 0.05 and Log2FC > 1 [18, 19]. Heatmaps, volcano plots, and boxplots were generated using the "heatmap" and "ggplot2" packages in R (version 3.3.3). Proteins were annotated using the UniProt-GOA database (http://​www.​ebi.​ac.​uk/​GOA), and enriched pathways were determined using Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis. Protein–protein interaction networks (PPI) for the DEPs were constructed using the STRING database (https://​string-db.​org/​), and further analysis of the PPI was carried out in Cytoscape software using cytohubba and MCODE plugins to identify and analyze key hub proteins, which were then subjected to GO/KEGG enrichment analysis.
Metabolomics data were normalized and integrated using support vector regression and analyzed for significant differences using orthogonal partial least squares-discriminant analysis (OPLS-DA). Variable importance in projection (VIP) values were calculated for each metabolite, with a VIP > 1 considered significant (P < 0.05) [20]. Pathway enrichment analysis for these differentially expressed metabolites was performed using GO/KEGG databases to identify significantly enriched pathways.

Prediction of potential therapeutic agents

The DSigDB database (http://​tanlab.​ucdenver.​edu/​DSigDB) was utilized to predict potential therapeutic agents for COVID-19 based on protein–drug interaction data. The thresholds set for selection were False Discovery Rate (FDR) < 0.05 and composite score > 5000.

Statistical analysis

Statistical analyses were conducted using SPSS software version 27.0, with continuous variables expressed as median and interquartile range (IQR). The Mann–Whitney U test (for two groups) and independent samples t-test were employed to assess differences in continuous variables. A P < 0.05 was considered statistically significant. Receiver operating characteristic (ROC) curves were plotted using Graphpad Prism software (version 9.5.1) to determine the sensitivity and specificity of hub genes. The area under the ROC curve (AUC) was measured, with an AUC > 0.7000 indicating diagnostic significance for the differentially expressed proteins.

Results

Clinical characteristics of the screening cohort

In terms of clinical parameters, compared to the healthy control group, the Omicron patient group exhibited elevated levels of Neutrophil percentage (NEU%), Monocyte count and percentage (MONO% and MONO), Basophils percentage (BASO%), International Normalized Ratio (INR), Fibrinogen (FIB), D-Dimer (D-D), C-reactive protein (CRP), Serum Amyloid A (SAA), and Procalcitonin (PCT) (P < 0.05). Conversely, there was a significant decrease in Lymphocyte count and percentage (LYM% and LYM), Eosinophils count and percentage (EOS and EOS%), Basophil count (BASO), and Prothrombin Time Activity (PTTA) (P < 0.05). These differences were statistically significant (Table 1).
Table 1
Basic information of healthy controls and omicron patients
 
Healthy controls
Omicron
P
N
15
15
 
Age
49.00 (37.00–61.00)
45.00 (29.00–57.00)
0.578
WBC, 109/L
5.90 (4.80–6.60)
5.33 (4.12–7.17)
0.817
NEU %
58.20 (52.20–62.30)
67.90 (59.20–76.20)
0.024
LYM %
29.80 (27.80–37.60)
18.80 (13.00–30.10)
0.002
MONO %
6.60 (6.40–8.20)
10.10 (8.50–13.70)
0.001
EOS %
3.20 (1.50–4.00)
0.40 (0.30–1.10)
 < 0.001
BASO %
0.13 (0.12–0.23)
0.42 (0.41–0.66)
 < 0.001
NEU, 109/L
3.40 (3.00–3.70)
3.71 (2.59–5.03)
0.380
LYM, 109/L
1.80 (1.60–2.20)
1.30 (0.69–1.39)
 < 0.001
MONO,109/L
0.42 (0.40–0.51)
0.57 (0.42–0.87)
0.040
EOS, 109/L
0.19 (0.16–0.22)
0.02 (0.01–0.06)
 < 0.001
BASO, 10^9/L
0.07 (0.05–0.08)
0.03 (0.02–0.04)
 < 0.001
PT, S
12.80 (12.50–13.50)
12.80 (11.30–15.30)
0.844
INR
0.60 (0.40–0.90)
0.98 (0.85–1.35)
 < 0.001
PTTA, %
96.00 (87.00–106.00)
82.00 (73.00–89.00)
0.004
FIB, g/L
3.22 (2.45–3.26)
3.48 (3.31–3.65)
0.003
APTT, S
33.10 (32.10–36.20)
36.40 (32.20–41.20)
0.171
TT, S
16.20 (15.50–17.80)
17.80 (16.20–18.20)
0.224
D-Dimer, μg/L
333.00 (258.00–364.00)
486.00 (452.00–852.00)
 < 0.001
CRP, mg/dL
2.50 (1.70–3.00)
5.30 (2.82–9.44)
0.005
SAA, mg/L
6.30 (2.90–8.40)
43.60 (29.40–67.60)
 < 0.001
PCT, ng/mL
0.02 (0.01–0.04)
0.09 (0.05–0.14)
 < 0.001
WBC White blood cell, NEU Neutrophil, LYM Lymphocyte, MONO Monocyte, EOS Eosinophils, BASO Basophil, PT Prothrombin time, INR International normalized ratio, PTTA Prothrombin activity, FIB Fibrinogen, APTT Activated partial thromboplastin time, TT Thrombin, DD D-Dimer, CRP C-reactive protein, SAA Serumamyloid A, PCT Procalcitonin

Proteomic profile and functional alterations associated with omicron

Using the UHPLC system, a total of 746 proteins were identified in the serum samples of both groups. Based on the criteria of | log2(FC) |> 1 and p-value < 0.05, a total of 27 differentially expressed proteins (DEPs) were identified, which included 15 upregulated proteins and 12 downregulated proteins, as shown in Fig. 2A (Additional file 1: Table S1). Principal Component Analysis (PCA), conducted using the normalized expression matrix, confirmed the variability between the proteomes of the samples (Fig. 2B). The heatmap of the aforementioned differentially expressed proteins is depicted in Fig. 2C.
GO and KEGG enrichment analyses were conducted on the differentially expressed proteins (Additional file 1: Table S2). These proteins were significantly enriched in GO pathways related to hemostasis, coagulation, and blood clotting, and were significantly enriched in the KEGG pathways related to Shigella and focal adhesion kinase pathways (Figs. 2D–H).

Selection and functional enrichment analysis of omicron hub proteins

Protein–protein interaction networks were constructed using the STRING database (Fig. 3A), and core hub proteins for both groups were identified through the MCC algorithm of the cytohubba plugin in Cytoscape software (Fig. 3C), leading to the selection of 10 hub proteins: THBS1, ACTN1, ACTC1, POTEF, ACTB, TPM4, VCL, ICAM1, PARK7, and YWHAB. Further analysis confirmed 8 of these hub proteins by MCODE: THBS1, ACTN1, ACTC1, POTEF, ACTB, TPM4, VCL, and ICAM1 (Fig. 3B). Venn diagrams from both methods confirmed the 8 common differentially expressed hub proteins (Fig. 3D). Heatmap analysis revealed an upregulated trend in Omicron patients compared to the healthy control group for all hub proteins except THBS1 (Fig. 3E).
Enrichment analysis of the 8 differentially expressed hub proteins was performed using GO/KEGG pathways (Additional file 1: Table S3). The results showed that the most enriched GO pathways included hemostasis, coagulation, and platelet aggregation, while the most enriched KEGG pathways were focal adhesion kinase, leukocyte transendothelial migration, Shigella, and actin cytoskeleton among others (Figs. 3F, G).

Clinical characteristics of the validation cohort

In the validation cohort, significant differences were observed when comparing Omicron patients (n = 20) to the healthy control group (n = 20). There was a notable increase in Neutrophil percentage (NEU%), Monocyte percentage (MONO%), Prothrombin time (PT), Fibrinogen (FIB), Activated partial thromboplastin time (APTT), D-Dimer (D-D), C-reactive protein (CRP), Serum amyloid (SAA), and Procalcitonin (PCT) levels (P < 0.05) in the Omicron patients. Conversely, a significant decrease was found in Lymphocyte percentage (LYM% and LYM), Eosinophils (EOS), Basophil percentage (BASO%), and Prothrombin Activity (PTTA) (P < 0.05). These differences were statistically significant (Table 2).
Table 2
Basic information of participants in the second batch verification queue
 
Healthy controls
Omicron
P
N
20
20
 
Age
54.00 (36.75–58.75)
53.00 (46.00–62.00)
0.580
WBC, 109/L
6.64 (5.35–9.27)
5.80 (4.62–7.85)
0.229
NEU %
53.88 (44.30–61.44)
92.66 (73.34–105.28)
 < 0.001
LYM %
29.37 (26.30–34.81)
16.71 (13.34–28.89)
 < 0.001
MONO %
6.98 (5.34–8.29)
12.33 (10.61–14.60)
 < 0.001
EOS %
1.45 (0.88–3.38)
1.65 (0.93–2.10)
0.839
BASO %
0.86 (0.38–1.13)
0.17 (0.10–0.22)
 < 0.001
NEU, 109/L
5.00 (3.67–6.05)
5.28 (2.65–5.53)
0.655
LYM, 109/L
2.40 (1.80–3.15)
1.50 (0.80–2.30)
0.004
MONO, 109/L
0.44 (0.26–0.65)
0.42 (0.37–0.66)
0.960
EOS, 109/L
0.22 (0.16–0.37)
0.15 (0.08–0.21)
0.004
BASO, 109/L
0.02 (0.01–0.03)
0.03 (0.02–0.03)
0.194
PT, S
12.45 (11.73–13.40)
14.00 (12.85–14.58)
0.003
INR
0.75 (0.60–0.90)
0.90 (0.80–1.18)
0.060
PTTA, %
97.00 (92.43–109.00)
83.20 (76.25–94.10)
 < 0.001
FIB, g/L
2.90 (2.53–3.45)
4.74 (3.53–5.52)
 < 0.001
APTT, S
35.70 (31.50–40.15)
41.40 (31.85–47.18)
0.048
TT, S
16.15 (15.23–18.78)
16.55 (15.20–18.33)
0.745
D-Dimer, μg/L
297.00 (211.00–364.75)
510.00 (439.50–603.50)
 < 0.001
CRP, mg/dL
2.75 (1.65–4.13)
8.50 (5.80–10.40)
 < 0.001
SAA, mg/L
5.50 (3.10–8.38)
35.75 (30.48–44.88)
 < 0.001
PCT, ng/mL
0.02 (0.01–0.05)
0.09 (0.06–0.15)
 < 0.001
WBC White blood cell, NEU Neutrophil, LYM Lymphocyte, MONO Monocyte, EOS Eosinophils, BASO Basophil, PT Prothrombin time, INR International normalized ratio, PTTA Prothrombin activity, FIB Fibrinogen, APTT Activated partial thromboplastin time, TT Thrombin, DD D-Dimer, CRP C-reactive protein, SAA Serumamyloid A, PCT Procalcitonin

Validation of differential hub protein expression and diagnostic value

The validation phase confirmed that the eight differentially expressed hub proteins significantly differed between the healthy control group and Omicron patients (P < 0.05) (Fig. 4A–H), also demonstrating diagnostic significance (Fig. 4I–P). Specifically, the area under the curve (AUC) for THBS1 was 0.7125 (95% CI 0.5527–0.8723), for ACTN1 it was 0.8450 (95% CI 0.7247–0.9653), for ACTC1 it was 0.8025 (95% CI 0.6672–0.9378), for POTEF 0.8025 (95% CI 0.6667–0.9383), for ACTB 0.7300 (95% CI 0.5733–0.8867), for TPM4 0.8250 (95% CI 0.6924–0.9576), for VCL 0.8775 (95% CI 0.7743–0.9807), and for ICAM1 0.7605 (95% CI 0.6113–0.9187).

Omicron-associated metabolomic features and functional changes

Through the application of 4D non-targeted metabolomics techniques, we identified 146 differentially expressed metabolites (DEMs) out of 357 metabolites (Additional file 1: Table S4). Quality control was performed based on the distribution of the total metabolites by group. Orthogonal partial least squares-discriminant analysis (OPLS-DA) demonstrated a clear separation between the healthy control group and the Omicron group, indicating distinctive metabolomic profiles (Fig. 5A). A volcano plot revealed the expression differences among all metabolites, with 27 being upregulated and 57 downregulated (Fig. 5B). A heatmap displayed the top 50 differential metabolites between the two groups, underscoring the metabolic disparities (Fig. 5C). These differentially expressed metabolites were primarily enriched in pathways related to tryptophan metabolism, retinol metabolism, and steroid hormone biosynthesis, among others (Fig. 5D, E).

Target drug prediction

The DSigDB database was used to predict potential target drugs associated with eight differentially expressed hub proteins that may treat Omicron infection by modulating the hub proteins. A total of 714 target drugs were finally predicted; the composite scores and corresponding target genes are listed in Additional file 1: Table S5. The top 10 predicted target drugs according to the composite scores are shown in Fig. 6.

Discussion

Amid the current global health crisis, the rapid transmission of the Omicron variant has had profound impacts on economies and public health systems worldwide. Omicron, with its enhanced ability to evade the immune system, has shown significant transmissibility compared to previous SARS-CoV-2 strains [21]. Although our understanding of the host response dynamics to this new variant is limited, identifying biomarkers that can predict disease progression or assist in diagnosis is vital to meet the challenges posed by Omicron. Our study integrates clinical diagnostics with multi-omics analyses to compare biomarkers between individuals infected with Omicron and healthy subjects, identifying eight differentially expressed hub proteins—THBS1, ACTN1, ACTC1, POTEF, ACTB, TPM4, VCL, and ICAM1.
Significant mutations in the spike protein of Omicron enhance its ability to evade immune responses [22], which may be associated with severe clinical outcomes such as cerebral embolism, deep vein thrombosis, and respiratory failure [23]. These findings resonate with studies positioning antithrombin as a broad-spectrum inhibitor of coronavirus infection, suggesting that the coagulation pathway may be a key element in the course of Omicron infection [24]. However, the specific relationship between Omicron and the coagulation pathway has not yet been elucidated. Therefore, the relationship between the coagulation pathway and Omicron warrants deeper investigation. This study's analysis of coagulation markers, including Activated Partial Thromboplastin Time (APTT), Fibrinogen (FIB), and D-Dimer, in patients infected with the Omicron variant reveals significant alterations when compared to healthy individuals. These changes underscore a pronounced association with hemostasis, coagulation, and blood clotting pathways, mirroring the conclusions drawn in earlier research [25]. Further substantiating these findings, our GO and KEGG pathway analyses on eight central hub proteins identified a pronounced enrichment in biological pathways directly tied to hemostasis, coagulation, and blood clotting. This enrichment not only corroborates the observed changes in coagulation markers but also suggests that Omicron infection might precipitate distinct alterations in the biological processes integral to these pathways.
The interplay between inflammation and coagulation is well recognized, with inflammation able to trigger coagulation mechanisms, and vice versa [26]. In the context of COVID-19, the activation of coagulation and hemostasis pathways is considered to be mediated by the inflammatory system's tissue factors [27]. Even though Omicron exhibits biological characteristics different from previous variants, severe infections may still induce platelet activation and partial desensitization, thereby affecting the coagulation pathway [28], and the coagulation pathway plays a significant role in regulating pulmonary fibrosis, hemostatic disorders, and surfactant damage [29]. VCL, a protein located at the cytoplasmic face of cellular matrix and intercellular adhesions, is involved in linking integrins to the F-actin cytoskeleton and can act as a mechanical sensor transmitting forces to maintain cellular shape, serving as a dynamic regulator of cell adhesion [30, 31]. The high expression of VCL can enhance cellular adhesiveness, and its regulatory role in adhesion is crucial for biological processes such as leukocyte transmigration through epithelial or endothelial layers, thereby mediating the migration of leukocytes in the inflammatory response of COVID-19 [32]. ACTB protein, also known as β-actin, plays a vital role in various cellular processes such as cell division, migration, invasion, vesicle transport, and cellular structural regulation[33]. ACTB can modulate endothelial nitric oxide synthase activity, altering NO production and thus causing endothelial dysfunction, activating coagulation pathways and inflammatory responses [34]. THBS1 is a matrix glycoprotein released by activated platelets, involved in angiogenesis, inflammatory responses, platelet aggregation, cell apoptosis, and fibrosis [35]. A decline in THBS1 expression is associated with oxidative stress damage [36], and THBS1 is involved in the fibrin clot response to injury [37]. Furthermore, silencing of THBS1 can inhibit the activation of the NLRP3 inflammasome, reducing the levels of inflammatory cytokines, thereby reducing pneumonia caused by cytokine storms [38]. POTEF, as a member of the POTE ankyrin domain family, can regulate TLR signaling which is critical in innate immunity and is expressed in immune cells playing a significant role during cell invasion [39], and it can inhibit apoptosis and promote cell growth [40]. However, the role of POTEF in pneumonia caused by Omicron is not clear, and our findings suggest that POTEF is involved in tissue coagulation processes, participating in disease inflammation and cell regulatory responses.
ACTN1 is a cytoskeletal actin-binding protein [41] that, in addition to mediating sarcomere function, performs important non-muscle functions, such as the regulation of cytokinesis, cell adhesion, and migration [42]. Our study found that high expression of ACTN1 is associated with cell adhesion and is a significant factor in the development and progression of Omicron. ACTC1 is a cardiac-related protein expressed in regenerating muscle fibers in diseased mature skeletal muscle [43], and our study shows that in the Omicron group, ACTC1 expression is elevated through the actin filament organization pathway. TPM4 is a key actin-binding protein [44], and studies have shown that TPM4 expression is strongly increased in the later stages of megakaryocyte formation and that TPM4 has a functional role in thrombus formation in mammals [45, 46]. This is consistent with our findings, which indicate that TPM4 can act as a promoter in pathways such as focal adhesion, mediating the occurrence, development, and outcome of the disease. Studies suggest that focal adhesion is associated with mechanisms of cell migration [47], which can regulate mechanotransduction, cell migration, cell cycle progression, proliferation differentiation, growth, and repair [48], and our study finds focal adhesion to be related to the pathological process of the disease. Additionally, ICAM-1 is a cell surface glycoprotein and adhesion receptor that, besides being expressed on vascular endothelial cells, also functions on epithelial and immune cells, affecting inflammatory stimulation [49]. Studies indicate that ICAM-1 is associated with leukocyte-endothelial cell interactions and solute permeability changes [50]. Our study suggests that ICAM-1 has a significant association with the inflammatory exudation and endothelial barrier establishment caused by Omicron.
In addition, the tryptophan pathway can be affected by systemic inflammatory signaling factors, promoting immune suppression and evasion of immune surveillance in inflammation [51], thereby helping to suppress inflammation caused by Omicron. L-tryptophan is an essential amino acid that produces a variety of signal molecules through complex metabolic pathways [52, 53]. Studies also suggest that the L-tryptophan pathway can achieve immune suppression by depleting tryptophan, depriving proliferating T cells of essential amino acids [52, 53]. Indole-3-acetate (I3A), a metabolite depleted in the context of the microbiome and high-fat diets, has been proven to reduce the production of pro-inflammatory cytokines and inhibit cell migration towards chemokines, achieving the effect of alleviating inflammation [54]. These is consistent with our findings.
This study highlights the therapeutic potential of targeting eight differentially expressed proteins in treating Omicron infection, with Domperidone and Cytochalasin D being particularly noteworthy. Domperidone's antiviral properties, as demonstrated by stimulating prolactin secretion, suggest a dual role in enhancing both innate and adaptive immune responses [55], aligning with our findings and highlighting the necessity for further research on its clinical application parameters. Similarly, Cytochalasin D's inhibition of actin polymerization and its capacity to reverse PLT-induced suppression of cell apoptosis present a novel therapeutic pathway worth exploring [56].
The study acknowledges several limitations, such as the complexity of interpreting metabolomic data, which necessitates advanced bioinformatics and further biological validation. While eight potential biomarkers were identified, their clinical significance requires validation in a more diverse population. The absence of direct mechanistic evidence, focusing instead on associations, calls for further research to clarify how these changes affect disease progression. Despite these challenges, the study offers a comprehensive view of Omicron's pathophysiology, contributing valuable insights into coagulation and inflammation and underscoring the need for continued research to develop targeted clinical and public health strategies.

Conclusion

In conclusion, our study highlights the significant impact of the Omicron variant on clinical and molecular levels. The identified hub proteins, enriched in coagulation and inflammatory pathways, underscore the intricate interplay between the inflammatory response and coagulation system in the context of Omicron infection. The biomarkers discovered offer a valuable resource for potential diagnostic and prognostic applications. While Omicron's unique biological characteristics necessitate further investigation, our findings contribute to the understanding of the pathophysiological mechanisms behind its clinical manifestations. However, the limitations of our study, include a call for further research with larger cohorts to validate these biomarkers and unravel the mechanisms of Omicron-induced pathology. Our work lays the groundwork for future studies aimed at developing targeted interventions to mitigate the effects of this challenging variant.

Acknowledgements

We thank the Immunology Department of the First Affiliated Hospital of Guangzhou Medical University for providing the experimental environment.

Declarations

Informed consent was obtained from all participants, and approval was granted by the Ethics Committee of Guangzhou Medical University Affiliated First Hospital (Ethics Numbers: 202001134 and 202115202).
Not applicable.

Competing interests

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

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Anhänge
Literatur
1.
Zurück zum Zitat Kandeel M, et al. Omicron variant genome evolution and phylogenetics. J Med Virol. 2022;94(4):1627–32.PubMedCrossRef Kandeel M, et al. Omicron variant genome evolution and phylogenetics. J Med Virol. 2022;94(4):1627–32.PubMedCrossRef
3.
Zurück zum Zitat Dhama K, et al. Global emerging omicron variant of SARS-CoV-2: impacts, challenges and strategies. J Infect Public Health. 2023;16(1):4–14.PubMedCrossRef Dhama K, et al. Global emerging omicron variant of SARS-CoV-2: impacts, challenges and strategies. J Infect Public Health. 2023;16(1):4–14.PubMedCrossRef
5.
10.
11.
Zurück zum Zitat Semple JW, Italiano JE Jr, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11(4):264–74.PubMedCrossRef Semple JW, Italiano JE Jr, Freedman J. Platelets and the immune continuum. Nat Rev Immunol. 2011;11(4):264–74.PubMedCrossRef
12.
Zurück zum Zitat Sevilya Z, et al. Differential platelet activation through an interaction with spike proteins of different SARS-CoV-2 variants. J Thromb Thrombolysis. 2023;56(4):538–47.PubMedCrossRef Sevilya Z, et al. Differential platelet activation through an interaction with spike proteins of different SARS-CoV-2 variants. J Thromb Thrombolysis. 2023;56(4):538–47.PubMedCrossRef
14.
Zurück zum Zitat Jackson CB, et al. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1):3–20.PubMedCrossRef Jackson CB, et al. Mechanisms of SARS-CoV-2 entry into cells. Nat Rev Mol Cell Biol. 2022;23(1):3–20.PubMedCrossRef
15.
Zurück zum Zitat Wang H, et al. Multi-omics blood atlas reveals unique features of immune and platelet responses to SARS-CoV-2 omicron breakthrough infection. Immunity. 2023;56(6):1410-1428.e8.PubMedPubMedCentralCrossRef Wang H, et al. Multi-omics blood atlas reveals unique features of immune and platelet responses to SARS-CoV-2 omicron breakthrough infection. Immunity. 2023;56(6):1410-1428.e8.PubMedPubMedCentralCrossRef
16.
Zurück zum Zitat SB MJ, et al. Biomarkers of coagulation, endothelial, platelet function, and fibrinolysis in patients with COVID-19: a prospective study. Sci Rep. 2024;14(1):2011.ADSCrossRef SB MJ, et al. Biomarkers of coagulation, endothelial, platelet function, and fibrinolysis in patients with COVID-19: a prospective study. Sci Rep. 2024;14(1):2011.ADSCrossRef
17.
Zurück zum Zitat Bakiera J, et al. Novel inflammatory markers in patients with severe COVID-19 and a pulmonary thrombotic event. Cent Eur J Immunol. 2023;48(1):167.PubMedPubMedCentralCrossRef Bakiera J, et al. Novel inflammatory markers in patients with severe COVID-19 and a pulmonary thrombotic event. Cent Eur J Immunol. 2023;48(1):167.PubMedPubMedCentralCrossRef
18.
19.
Zurück zum Zitat Ho AM, et al. Label-free proteomics differences in the dorsolateral prefrontal cortex between bipolar disorder patients with and without psychosis. J Affect Disord. 2020;270:165–73.PubMedPubMedCentralCrossRef Ho AM, et al. Label-free proteomics differences in the dorsolateral prefrontal cortex between bipolar disorder patients with and without psychosis. J Affect Disord. 2020;270:165–73.PubMedPubMedCentralCrossRef
20.
Zurück zum Zitat Che J, et al. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with the progression of gastroesophageal cancer. BMC Cancer. 2023;23(1):1238.PubMedPubMedCentralCrossRef Che J, et al. Untargeted serum metabolomics reveals potential biomarkers and metabolic pathways associated with the progression of gastroesophageal cancer. BMC Cancer. 2023;23(1):1238.PubMedPubMedCentralCrossRef
21.
22.
23.
Zurück zum Zitat Jin S, et al. A case of multiple thrombosis and septic shock in a critically ill patient with omicron infection. J Infect Public Health. 2022;15(11):1321–5.PubMedPubMedCentralCrossRef Jin S, et al. A case of multiple thrombosis and septic shock in a critically ill patient with omicron infection. J Infect Public Health. 2022;15(11):1321–5.PubMedPubMedCentralCrossRef
24.
Zurück zum Zitat Wettstein L, et al. Native and activated antithrombin inhibits TMPRSS2 activity and SARS-CoV-2 infection. J Med Virol. 2023;95(1):e28124.PubMedCrossRef Wettstein L, et al. Native and activated antithrombin inhibits TMPRSS2 activity and SARS-CoV-2 infection. J Med Virol. 2023;95(1):e28124.PubMedCrossRef
25.
Zurück zum Zitat Zhang H, et al. Epidemiological and clinical features of SARS-CoV-2 omicron variant infection in Quanzhou, Fujian province: a retrospective study. Sci Rep. 2023;13(1):22152.ADSPubMedPubMedCentralCrossRef Zhang H, et al. Epidemiological and clinical features of SARS-CoV-2 omicron variant infection in Quanzhou, Fujian province: a retrospective study. Sci Rep. 2023;13(1):22152.ADSPubMedPubMedCentralCrossRef
26.
Zurück zum Zitat Levi M, van der Poll T. Inflammation and coagulation. Crit Care Med. 2010;38(2 Suppl):S26-34.PubMedCrossRef Levi M, van der Poll T. Inflammation and coagulation. Crit Care Med. 2010;38(2 Suppl):S26-34.PubMedCrossRef
27.
Zurück zum Zitat Levi M, et al. Infection and inflammation and the coagulation system. Cardiovasc Res. 2003;60(1):26–39.ADSPubMedCrossRef Levi M, et al. Infection and inflammation and the coagulation system. Cardiovasc Res. 2003;60(1):26–39.ADSPubMedCrossRef
28.
Zurück zum Zitat Garcia C, et al. SARS-CoV-2 Omicron variant infection affects blood platelets, a comparative analysis with Delta variant. Front Immunol. 2023;14:1231576.PubMedPubMedCentralCrossRef Garcia C, et al. SARS-CoV-2 Omicron variant infection affects blood platelets, a comparative analysis with Delta variant. Front Immunol. 2023;14:1231576.PubMedPubMedCentralCrossRef
29.
Zurück zum Zitat Krygier A, et al. Molecular pathogenesis of fibrosis, thrombosis and surfactant dysfunction in the lungs of severe COVID-19 patients. Biomolecules. 2022;12(12):1845.PubMedPubMedCentralCrossRef Krygier A, et al. Molecular pathogenesis of fibrosis, thrombosis and surfactant dysfunction in the lungs of severe COVID-19 patients. Biomolecules. 2022;12(12):1845.PubMedPubMedCentralCrossRef
30.
Zurück zum Zitat Shih YT, et al. Vinculin phosphorylation impairs vascular endothelial junctions promoting atherosclerosis. Eur Heart J. 2023;44(4):304–18.PubMedCrossRef Shih YT, et al. Vinculin phosphorylation impairs vascular endothelial junctions promoting atherosclerosis. Eur Heart J. 2023;44(4):304–18.PubMedCrossRef
32.
Zurück zum Zitat Demali KA. Vinculin–a dynamic regulator of cell adhesion. Trends Biochem Sci. 2004;29(11):565–7.PubMedCrossRef Demali KA. Vinculin–a dynamic regulator of cell adhesion. Trends Biochem Sci. 2004;29(11):565–7.PubMedCrossRef
33.
Zurück zum Zitat Artman L, et al. Planning your every move: the role of β-actin and its post-transcriptional regulation in cell motility. Semin Cell Dev Biol. 2014;34:33–43.PubMedCrossRef Artman L, et al. Planning your every move: the role of β-actin and its post-transcriptional regulation in cell motility. Semin Cell Dev Biol. 2014;34:33–43.PubMedCrossRef
37.
Zurück zum Zitat Atanasova VS, et al. Thrombospondin-1 is a major activator of TGF-β signaling in recessive dystrophic epidermolysis bullosa fibroblasts. J Invest Dermatol. 2019;139(7):1497-1505.e5.PubMedCrossRef Atanasova VS, et al. Thrombospondin-1 is a major activator of TGF-β signaling in recessive dystrophic epidermolysis bullosa fibroblasts. J Invest Dermatol. 2019;139(7):1497-1505.e5.PubMedCrossRef
38.
Zurück zum Zitat Sun J, et al. USF2 knockdown downregulates THBS1 to inhibit the TGF-β signaling pathway and reduce pyroptosis in sepsis-induced acute kidney injury. Pharmacol Res. 2022;176:105962.PubMedCrossRef Sun J, et al. USF2 knockdown downregulates THBS1 to inhibit the TGF-β signaling pathway and reduce pyroptosis in sepsis-induced acute kidney injury. Pharmacol Res. 2022;176:105962.PubMedCrossRef
39.
Zurück zum Zitat Vekariya U, et al. Identification of MΦ specific POTEE expression: Its role in mTORC2 activation via protein-protein interaction in TAMs. Cell Immunol. 2019;335:30–40.PubMedCrossRef Vekariya U, et al. Identification of MΦ specific POTEE expression: Its role in mTORC2 activation via protein-protein interaction in TAMs. Cell Immunol. 2019;335:30–40.PubMedCrossRef
41.
Zurück zum Zitat Sjöblom B, Salmazo A, Djinović-Carugo K. Alpha-actinin structure and regulation. Cell Mol Life Sci. 2008;65(17):2688–701.PubMedCrossRef Sjöblom B, Salmazo A, Djinović-Carugo K. Alpha-actinin structure and regulation. Cell Mol Life Sci. 2008;65(17):2688–701.PubMedCrossRef
42.
Zurück zum Zitat Xie GF, et al. High ACTN1 is associated with poor prognosis, and ACTN1 silencing suppresses cell proliferation and metastasis in oral squamous cell carcinoma. Drug Des Devel Ther. 2020;14:1717–27.PubMedPubMedCentralCrossRef Xie GF, et al. High ACTN1 is associated with poor prognosis, and ACTN1 silencing suppresses cell proliferation and metastasis in oral squamous cell carcinoma. Drug Des Devel Ther. 2020;14:1717–27.PubMedPubMedCentralCrossRef
43.
Zurück zum Zitat Boutilier JK, et al. Variable cardiac α-actin (Actc1) expression in early adult skeletal muscle correlates with promoter methylation. Biochim Biophys Acta Gene Regul Mech. 2017;1860(10):1025–36.PubMedCrossRef Boutilier JK, et al. Variable cardiac α-actin (Actc1) expression in early adult skeletal muscle correlates with promoter methylation. Biochim Biophys Acta Gene Regul Mech. 2017;1860(10):1025–36.PubMedCrossRef
44.
Zurück zum Zitat Liu B, et al. Cytoskeleton remodeling mediated by circRNA-YBX1 phase separation suppresses the metastasis of liver cancer. Proc Natl Acad Sci U S A. 2023;120(30):e2220296120.PubMedPubMedCentralCrossRef Liu B, et al. Cytoskeleton remodeling mediated by circRNA-YBX1 phase separation suppresses the metastasis of liver cancer. Proc Natl Acad Sci U S A. 2023;120(30):e2220296120.PubMedPubMedCentralCrossRef
45.
47.
Zurück zum Zitat Paluch EK, Aspalter IM, Sixt M. Focal adhesion-independent cell migration. Annu Rev Cell Dev Biol. 2016;32:469–90.PubMedCrossRef Paluch EK, Aspalter IM, Sixt M. Focal adhesion-independent cell migration. Annu Rev Cell Dev Biol. 2016;32:469–90.PubMedCrossRef
48.
Zurück zum Zitat Mishra YG, Manavathi B. Focal adhesion dynamics in cellular function and disease. Cell Signal. 2021;85:110046.PubMedCrossRef Mishra YG, Manavathi B. Focal adhesion dynamics in cellular function and disease. Cell Signal. 2021;85:110046.PubMedCrossRef
49.
Zurück zum Zitat Bui TM, Wiesolek HL, Sumagin R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J Leukoc Biol. 2020;108(3):787–99.PubMedCrossRef Bui TM, Wiesolek HL, Sumagin R. ICAM-1: A master regulator of cellular responses in inflammation, injury resolution, and tumorigenesis. J Leukoc Biol. 2020;108(3):787–99.PubMedCrossRef
50.
Zurück zum Zitat Sumagin R, Lomakina E, Sarelius IH. Leukocyte-endothelial cell interactions are linked to vascular permeability via ICAM-1-mediated signaling. Am J Physiol Heart Circ Physiol. 2008;295(3):H969-h977.PubMedPubMedCentralCrossRef Sumagin R, Lomakina E, Sarelius IH. Leukocyte-endothelial cell interactions are linked to vascular permeability via ICAM-1-mediated signaling. Am J Physiol Heart Circ Physiol. 2008;295(3):H969-h977.PubMedPubMedCentralCrossRef
51.
Zurück zum Zitat Cervenka I, Agudelo LZ, Ruas JL. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science. 2017;357(6349):eaaf9794.PubMedCrossRef Cervenka I, Agudelo LZ, Ruas JL. Kynurenines: tryptophan’s metabolites in exercise, inflammation, and mental health. Science. 2017;357(6349):eaaf9794.PubMedCrossRef
52.
Zurück zum Zitat Fiore A, Murray PJ. Tryptophan and indole metabolism in immune regulation. Curr Opin Immunol. 2021;70:7–14.PubMedCrossRef Fiore A, Murray PJ. Tryptophan and indole metabolism in immune regulation. Curr Opin Immunol. 2021;70:7–14.PubMedCrossRef
53.
Zurück zum Zitat Prudêncio APA, et al. Red meat intake, indole-3-Acetate, and Dorea longicatena together affect insulin resistance after gastric bypass. Nutrients. 2023;15(5):1185.PubMedPubMedCentralCrossRef Prudêncio APA, et al. Red meat intake, indole-3-Acetate, and Dorea longicatena together affect insulin resistance after gastric bypass. Nutrients. 2023;15(5):1185.PubMedPubMedCentralCrossRef
54.
Zurück zum Zitat Krishnan S, et al. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Rep. 2018;23(4):1099–111.PubMedPubMedCentralCrossRef Krishnan S, et al. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Rep. 2018;23(4):1099–111.PubMedPubMedCentralCrossRef
55.
Zurück zum Zitat Rabanal Basalo A, et al. A randomized, double-blind study on the efficacy of oral domperidone versus placebo for reducing SARS-CoV-2 viral load in mild-to-moderate COVID-19 patients in primary health care. Ann Med. 2023;55(2):2268535.PubMedPubMedCentralCrossRef Rabanal Basalo A, et al. A randomized, double-blind study on the efficacy of oral domperidone versus placebo for reducing SARS-CoV-2 viral load in mild-to-moderate COVID-19 patients in primary health care. Ann Med. 2023;55(2):2268535.PubMedPubMedCentralCrossRef
56.
Zurück zum Zitat Lan Y, et al. Upregulation of girdin delays endothelial cell apoptosis via promoting engulfment of platelets. Mol Biol Rep. 2023;50(10):8111–20.PubMedCrossRef Lan Y, et al. Upregulation of girdin delays endothelial cell apoptosis via promoting engulfment of platelets. Mol Biol Rep. 2023;50(10):8111–20.PubMedCrossRef
Metadaten
Titel
Deciphering the omicron variant: integrated omics analysis reveals critical biomarkers and pathophysiological pathways
verfasst von
Qianyue Yang
Zhiwei Lin
Mingshan Xue
Yueting Jiang
Libing Chen
Jiahong Chen
Yuhong Liao
Jiali Lv
Baojun Guo
Peiyan Zheng
Huimin Huang
Baoqing Sun
Publikationsdatum
01.12.2024
Verlag
BioMed Central
Erschienen in
Journal of Translational Medicine / Ausgabe 1/2024
Elektronische ISSN: 1479-5876
DOI
https://doi.org/10.1186/s12967-024-05022-z

Weitere Artikel der Ausgabe 1/2024

Journal of Translational Medicine 1/2024 Zur Ausgabe

Leitlinien kompakt für die Innere Medizin

Mit medbee Pocketcards sicher entscheiden.

Seit 2022 gehört die medbee GmbH zum Springer Medizin Verlag

Ist Fasten vor Koronarinterventionen wirklich nötig?

Wenn Eingriffe wie eine Koronarangiografie oder eine Koronarangioplastie anstehen, wird häufig empfohlen, in den Stunden zuvor nüchtern zu bleiben. Ein französisches Forscherteam hat diese Maßnahme hinterfragt.

Typ-2-Diabetes: Ernährungsunsicherheit vervierfacht Risiko für schwere Hypoglykämien

04.06.2024 Typ-2-Diabetes Nachrichten

Wenn ältere Menschen mit Typ-2-Diabetes Schwierigkeiten beim Beschaffen und Zubereiten von Mahlzeiten haben, geht dies mit einem deutlich gesteigerten Risiko für schwere Hypoglykämien einher.

Mehr Brustkrebs, aber weniger andere gynäkologische Tumoren mit Levonorgestrel-IUS

04.06.2024 Levonorgestrel Nachrichten

Unter Frauen, die ein Levonorgestrel-freisetzendes intrauterines System (IUS) verwenden, ist die Brustkrebsrate um 13% erhöht. Dafür kommt es deutlich seltener zu Endometrium-, Zervix- und Ovarialkarzinomen.

GLP-1-Agonist Semaglutid wirkt kardio- und nephroprotektiv

03.06.2024 Semaglutid Nachrichten

Der GLP-1-Agonist Semaglutid hat in der FLOW-Studie bewiesen, dass sich damit die Progression chronischer Nierenerkrankungen bei Patienten mit Typ-2-Diabetes bremsen lässt. Auch in kardiovaskulärer Hinsicht war die Therapie erfolgreich.

Update Innere Medizin

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