1
|
Tuli TR, Mia M, Habib A. Integrated bioinformatics approach for the identification and validation of novel biomarkers in ACC progression and prognosis. Biomarkers 2025:1-15. [PMID: 40183287 DOI: 10.1080/1354750x.2025.2489453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
CONCLUSION In conclusion, the identified novel biomarkers and associated pathways, provides a comprehensive insight into the molecular mechanisms, prognosis, and potential clinical applications for the diagnosis and therapeutic interventions of ACC.
Collapse
Affiliation(s)
- Tonima Rahman Tuli
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Mijan Mia
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| | - Ahsan Habib
- Biotechnology and Genetic Engineering Discipline, Khulna University, Khulna, Bangladesh
| |
Collapse
|
2
|
Shen T, Wang W, Wang H, Zhu X, Zhu G. Mitochondrial miRNA miR-134-5p Play Oncogenic Role in Clear Cell Renal Cell Carcinoma. Biomolecules 2025; 15:445. [PMID: 40149981 PMCID: PMC11939903 DOI: 10.3390/biom15030445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/28/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial miRNAs (mitomiRs), which are miRNAs that located within mitochondria, have emerged as crucial regulators in a variety of human diseases, including multiple types of cancers. However, the specific role of mitomiRs in clear cell renal cell carcinoma (ccRCC) remains elusive. In this study, we employed a combination of experimental and bioinformatic approaches to uncover the diverse and abundant subcellular distribution of miRNAs within mitochondria in ccRCC. Notably, RNA sequencing after mitochondrial fractionation identified miR-134-5p as a miRNA predominantly detected in the mitochondria of 786O cells, and its expression is significantly upregulated compared to that in 293T cells. Differential expression and survival analyses from TCGA reveal that the upregulation of miR-134-5p is prevalent and closely associated with poor survival outcomes in ccRCC patients. Functionally, exogenous overexpression of miR-134-5p mimics promotes migration in both 786O and Caki-1 cells. Mechanistically, overexpressing the miR-134-5p mimic dramatically downregulates the mRNA levels of CHST6, SFXN2, and GRIK3, whereas the miR-134-5p inhibitor markedly upregulates their expression. Notably, these target mRNAs also predominantly detected in the mitochondria of 786O cells. The downregulated expression signatures of CHST6, SFXN2, and GRIK3 are also closely correlated with poor survival outcomes in ccRCC patients. Taken together, our work identifies a novel mitomiR, miR-134-5p, in ccRCC, provides potential targets that could serve as effective biomarkers for ccRCC diagnosis and prognosis, and opens new avenues for understanding the mitomiR-directed regulatory network in ccRCC progression.
Collapse
Affiliation(s)
- Tao Shen
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (H.W.); (X.Z.)
- Anhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Wei Wang
- Department of Geriatrics, Gerontology Institute of Anhui Province, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230026, China;
| | - Haiyang Wang
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (H.W.); (X.Z.)
- Anhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Xinyi Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (H.W.); (X.Z.)
- Anhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| | - Guoping Zhu
- Anhui Provincial Key Laboratory of Molecular Enzymology and Mechanism of Major Metabolic Diseases, College of Life Sciences, Anhui Normal University, Wuhu 241000, China; (H.W.); (X.Z.)
- Anhui Provincial Engineering Research Centre for Molecular Detection and Diagnostics, College of Life Sciences, Anhui Normal University, Wuhu 241000, China
| |
Collapse
|
3
|
Ranganathan Y, Kumar PR, Paramasivam SG, Krishnan RS. A Review of Connecting Bioinformatic Techniques to Rheumatoid Arthritis and its Associated Comorbidities. Curr Rheumatol Rev 2025; 21:25-36. [PMID: 38803169 DOI: 10.2174/0115733971302188240515075547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/29/2024]
Abstract
Rheumatoid Arthritis (RA) is a progressive autoimmune condition inflicting serious threats to people's life and health by causing severe pain and joint destruction. It affects not only bones and joints but also causes comorbid conditions and shortens the lifetime. The interactions and synergistic effects of comorbid disease with RA are not yet well studied. Hence, understanding how these conditions will collectively affect the progression and outcome of RA is the current area of research. Identification of RA and comorbidities associated with target genes may uncover diagnosis and treatment methodologies. This review is to provide an overview of the interlinking approach of Rheumatoid Arthritis with its comorbid conditions and its systemic complications using bioinformatic techniques which would be useful to identify the genes and pathways that are in common for both RA and comorbid diseases. It would also emphasize the significance of bioinformatics in comparing the pathological features of RA and comorbid diseases. With the help of bioinformatics, valuable insights into the mechanism underlying Rheumatoid arthritis and comorbid diseases would be better understood.
Collapse
Affiliation(s)
- Yeswanth Ranganathan
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| | - Pritam Ramesh Kumar
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| | - Sudhakar Gandhi Paramasivam
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| | - Ravi Shankar Krishnan
- Department of Biotechnology, University College of Engineering, BIT Campus, Anna University, Tiruchirappalli, 620024, India
| |
Collapse
|
4
|
Sun P, Wang Y, Zhou S, Liang J, Zhang B, Li P, Han R, Fei G, Cao C, Wang R. Exploring the shared pathogenic mechanisms of tuberculosis and COVID-19: emphasizing the role of VNN1 in severe COVID-19. Front Cell Infect Microbiol 2024; 14:1453466. [PMID: 39639868 PMCID: PMC11618882 DOI: 10.3389/fcimb.2024.1453466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Accepted: 10/28/2024] [Indexed: 12/07/2024] Open
Abstract
Background In recent years, COVID-19 and tuberculosis have emerged as major infectious diseases, significantly contributing to global mortality as respiratory illnesses. There is increasing evidence of a reciprocal influence between these diseases, exacerbating their incidence, severity, and mortality rates. Methods This study involved retrieving COVID-19 and tuberculosis data from the GEO database and identifying common differentially expressed genes. Machine learning techniques, specifically random forest analysis, were applied to pinpoint key genes for diagnosing COVID-19. The Cibersort algorithm was employed to estimate immune cell infiltration in individuals with COVID-19. Additionally, single-cell sequencing was used to study the distribution of VNN1 within immune cells, and molecular docking provided insights into potential drugs targeting these critical prognosis genes. Results GMNN, SCD, and FUT7 were identified as robust diagnostic markers for COVID-19 across training and validation datasets. Importantly, VNN1 was associated with the progression of severe COVID-19, showing a strong correlation with clinical indicators and immune cell infiltration. Single-cell sequencing demonstrated a predominant distribution of VNN1 in neutrophils, and molecular docking highlighted potential pharmacological targets for VNN1. Conclusions This study enhances our understanding of the shared pathogenic mechanisms underlying tuberculosis and COVID-19, providing essential insights that could improve the diagnosis and treatment of severe COVID-19 cases.
Collapse
Affiliation(s)
- Peng Sun
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Yue Wang
- Department of Infectious Diseases, Hefei Second People’s Hospital, Hefei, China
| | - Sijing Zhou
- Department of Occupational Disease, Hefei Third Clinical College of Anhui Medical University, Hefei, China
| | - Jiahui Liang
- Department of Breast Surgery, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Binbin Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Pulin Li
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Rui Han
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Guanghe Fei
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang, China
| | - Ran Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
5
|
Zhang X, Liu C, Cao L, Tang H, Jiang H, Hu C, Dong X, Zhou F, Qin K, Liu Q, Shen J, Zhou Y. Exploring the mechanisms of chronic obstructive pulmonary disease and Crohn's disease: a bioinformatics-based study. Sci Rep 2024; 14:27461. [PMID: 39523420 PMCID: PMC11551177 DOI: 10.1038/s41598-024-78697-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
This study explored the comorbid mechanisms between Crohn's disease (CD) and chronic obstructive pulmonary disease (COPD) using bioinformatics analysis. From the Gene Expression Omnibus (GEO) microarray dataset, 349 common differentially expressed genes (coDEGs) were identified, and 8 shared hub genes were found: CCL2, CXCL1, TLR2, ICAM1, PTPRC, ITGAX, PTGS2, and MMP9, which were vital for immune function and regulation of inflammatory responses. In addition, the study also analyzed the association between coDEGs and immune cell infiltration using the single-sample gene set enrichment algorithm (ssGSEA). Potential drugs related to these genes were identified using the connectivity map (CMap). These findings provided new perspectives for understanding the interaction between CD and COPD.
Collapse
Affiliation(s)
- Xinxin Zhang
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Caiping Liu
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Luqian Cao
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Hongguang Tang
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Haiyun Jiang
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Changjing Hu
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Xuehong Dong
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Feiyang Zhou
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Kunming Qin
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Qiang Liu
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China
| | - Jinyang Shen
- Department of Pharmacy, Jiangsu Ocean University, 59 Cangwu Road, Haizhou District, Lianyungang, 222000, Jiangsu, China.
| | - Yue Zhou
- Department of Pharmacy, Lianyungang Affiliated Hospital of Nanjing University of Chinese Medicine, No. 160, Chaoyang Middle Road, Haizhou District, Lianyungang, 222004, Jiangsu, China.
| |
Collapse
|
6
|
Yang Y, Sheng YH, Carreira P, Wang T, Zhao H, Wang R. Genome-wide assessment of shared genetic landscape of idiopathic pulmonary fibrosis and its comorbidities. Hum Genet 2024; 143:1223-1239. [PMID: 39103522 PMCID: PMC11485074 DOI: 10.1007/s00439-024-02696-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial lung disease accompanied by both local and systemic comorbidities. Genetic factors play a role in the development of IPF and certain associated comorbidities. Nevertheless, it is uncertain whether there are shared genetic factors underlying IPF and these comorbidities. To bridge this knowledge gap, we conducted a systematic investigation into the shared genetic architecture between IPF and ten prevalent heritable comorbidities (i.e., body mass index [BMI], coronary artery disease [CAD], chronic obstructive pulmonary disease [COPD], gastroesophageal reflux disease, lung cancer, major depressive disorder [MDD], obstructive sleep apnoea, pulmonary hypertension [PH], stroke, and type 2 diabetes), by utilizing large-scale summary data from their respective genome-wide association studies and multi-omics studies. We revealed significant (false discovery rate [FDR] < 0.05) and moderate genetic correlations between IPF and seven comorbidities, excluding lung cancer, MDD and PH. Evidence suggested a partially putative causal effect of IPF on CAD. Notably, we observed FDR-significant genetic enrichments in lung for the cross-trait between IPF and CAD and in liver for the cross-trait between IPF and COPD. Additionally, we identified 65 FDR-significant genes over-represented in 20 biological pathways related to the etiology of IPF, BMI, and COPD, including inflammation-related mucin gene clusters. Several of these genes were associated with clinically relevant drugs for the treatment of IPF, CAD, and/or COPD. Our results underscore the pervasive shared genetic basis between IPF and its common comorbidities and hold future implications for early diagnosis of IPF-related comorbidities, drug repurposing, and the development of novel therapies for IPF.
Collapse
Affiliation(s)
- Yuanhao Yang
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.
| | - Yong H Sheng
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia
- Cancer Program, QIMR Berghofer Medical Research Institute, Herston, QLD, Australia
| | - Patricia Carreira
- Immunology and Infectious Disease Division, John Curtin School of Medical Research, Australian National University, Acton, ACT, Australia
| | - Tong Wang
- Department of Health Statistics, School of Public Health, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Huiying Zhao
- Department of Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Ran Wang
- Mater Research Institute, The University of Queensland, Woolloongabba, QLD, Australia.
| |
Collapse
|
7
|
Alamin MH, Rahaman MM, Ferdousi F, Sarker A, Ali MA, Hossen MB, Sarker B, Kumar N, Mollah MNH. In-silico discovery of common molecular signatures for which SARS-CoV-2 infections and lung diseases stimulate each other, and drug repurposing. PLoS One 2024; 19:e0304425. [PMID: 39024368 PMCID: PMC11257407 DOI: 10.1371/journal.pone.0304425] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/12/2024] [Indexed: 07/20/2024] Open
Abstract
COVID-19 caused by SARS-CoV-2 is a global health issue. It is yet a severe risk factor to the patients, who are also suffering from one or more chronic diseases including different lung diseases. In this study, we explored common molecular signatures for which SARS-CoV-2 infections and different lung diseases stimulate each other, and associated candidate drug molecules. We identified both SARS-CoV-2 infections and different lung diseases (Asthma, Tuberculosis, Cystic Fibrosis, Pneumonia, Emphysema, Bronchitis, IPF, ILD, and COPD) causing top-ranked 11 shared genes (STAT1, TLR4, CXCL10, CCL2, JUN, DDX58, IRF7, ICAM1, MX2, IRF9 and ISG15) as the hub of the shared differentially expressed genes (hub-sDEGs). The gene ontology (GO) and pathway enrichment analyses of hub-sDEGs revealed some crucial common pathogenetic processes of SARS-CoV-2 infections and different lung diseases. The regulatory network analysis of hub-sDEGs detected top-ranked 6 TFs proteins and 6 micro RNAs as the key transcriptional and post-transcriptional regulatory factors of hub-sDEGs, respectively. Then we proposed hub-sDEGs guided top-ranked three repurposable drug molecules (Entrectinib, Imatinib, and Nilotinib), for the treatment against COVID-19 with different lung diseases. This recommendation is based on the results obtained from molecular docking analysis using the AutoDock Vina and GLIDE module of Schrödinger. The selected drug molecules were optimized through density functional theory (DFT) and observing their good chemical stability. Finally, we explored the binding stability of the highest-ranked receptor protein RELA with top-ordered three drugs (Entrectinib, Imatinib, and Nilotinib) through 100 ns molecular dynamic (MD) simulations with YASARA and Desmond module of Schrödinger and observed their consistent performance. Therefore, the findings of this study might be useful resources for the diagnosis and therapies of COVID-19 patients who are also suffering from one or more lung diseases.
Collapse
Affiliation(s)
- Muhammad Habibulla Alamin
- Faculty of Science, Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md. Matiur Rahaman
- Faculty of Science, Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
- Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Haining, P. R. China
| | - Farzana Ferdousi
- Faculty of Science, Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Arnob Sarker
- Faculty of Science, Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
- Faculty of Science, Department of Statistics, Bioinformatics Laboratory (Dry), University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Ahad Ali
- Faculty of Science, Department of Statistics, Bioinformatics Laboratory (Dry), University of Rajshahi, Rajshahi, Bangladesh
- Faculty of Science, Department of Chemistry, University of Rajshahi, Rajshahi, Bangladesh
| | - Md. Bayazid Hossen
- Faculty of Science, Department of Statistics, Bioinformatics Laboratory (Dry), University of Rajshahi, Rajshahi, Bangladesh
- Department of Agricultural and Applied Statistics, Bangladesh Agricultural University, Mymensingh, Bangladesh
| | - Bandhan Sarker
- Faculty of Science, Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Nishith Kumar
- Faculty of Science, Department of Statistics, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj, Bangladesh
| | - Md. Nurul Haque Mollah
- Faculty of Science, Department of Statistics, Bioinformatics Laboratory (Dry), University of Rajshahi, Rajshahi, Bangladesh
| |
Collapse
|
8
|
Wang L, Chen A, Zhang L, Zhang J, Wei S, Chen Y, Hu M, Mo Y, Li S, Zeng M, Li H, Liang C, Ren Y, Xu L, Liang W, Zhu X, Wang X, Sun D. Deciphering the molecular nexus between Omicron infection and acute kidney injury: a bioinformatics approach. Front Mol Biosci 2024; 11:1340611. [PMID: 39027131 PMCID: PMC11254815 DOI: 10.3389/fmolb.2024.1340611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/07/2024] [Indexed: 07/20/2024] Open
Abstract
Background The ongoing global health crisis of COVID-19, and particularly the challenges posed by recurrent infections of the Omicron variant, have significantly strained healthcare systems worldwide. There is a growing body of evidence indicating an increased susceptibility to Omicron infection in patients suffering from Acute Kidney Injury (AKI). However, the intricate molecular interplay between AKI and Omicron variant of COVID-19 remains largely enigmatic. Methods This study employed a comprehensive analysis of human RNA sequencing (RNA-seq) and microarray datasets to identify differentially expressed genes (DEGs) associated with Omicron infection in the context of AKI. We engaged in functional enrichment assessments, an examination of Protein-Protein Interaction (PPI) networks, and advanced network analysis to elucidate the cellular signaling pathways involved, identify critical hub genes, and determine the relevant controlling transcription factors and microRNAs. Additionally, we explored protein-drug interactions to highlight potential pharmacological interventions. Results Our investigation revealed significant DEGs and cellular signaling pathways implicated in both Omicron infection and AKI. We identified pivotal hub genes, including EIF2AK2, PLSCR1, GBP1, TNFSF10, C1QB, and BST2, and their associated regulatory transcription factors and microRNAs. Notably, in the murine AKI model, there was a marked reduction in EIF2AK2 expression, in contrast to significant elevations in PLSCR1, C1QB, and BST2. EIF2AK2 exhibited an inverse relationship with the primary AKI mediator, Kim-1, whereas PLSCR1 and C1QB demonstrated strong positive correlations with it. Moreover, we identified potential therapeutic agents such as Suloctidil, Apocarotenal, 3'-Azido-3'-deoxythymidine, among others. Our findings also highlighted a correlation between the identified hub genes and diseases like myocardial ischemia, schizophrenia, and liver cirrhosis. To further validate the credibility of our data, we employed an independent validation dataset to verify the hub genes. Notably, the expression patterns of PLSCR1, GBP1, BST2, and C1QB were consistent with our research findings, reaffirming the reliability of our results. Conclusion Our bioinformatics analysis has provided initial insights into the shared genetic landscape between Omicron COVID-19 infections and AKI, identifying potential therapeutic targets and drugs. This preliminary investigation lays the foundation for further research, with the hope of contributing to the development of innovative treatment strategies for these complex medical conditions.
Collapse
Affiliation(s)
- Li Wang
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Anning Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Lantian Zhang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Junwei Zhang
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Shuqi Wei
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yangxiao Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Mingliang Hu
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Yihao Mo
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Sha Li
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Min Zeng
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Huafeng Li
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Caixing Liang
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Yi Ren
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Liting Xu
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Wenhua Liang
- Nephrology Department, Southern Medical University Affiliated Longhua People’s Hospital, Shenzhen, China
| | - Xuejiao Zhu
- Department of Anesthesiology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xiaokai Wang
- Xuzhou First People’s Hospital, Xuzhou, Jiangsu, China
| | - Donglin Sun
- Department of Urology, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| |
Collapse
|
9
|
Rusu EC, Monfort-Lanzas P, Bertran L, Barrientos-Riosalido A, Solé E, Mahmoudian R, Aguilar C, Briansó S, Mohamed F, Garcia S, Camaron J, Auguet T. Towards understanding post-COVID-19 condition: A systematic meta-analysis of transcriptomic alterations with sex-specific insights. Comput Biol Med 2024; 175:108507. [PMID: 38657468 DOI: 10.1016/j.compbiomed.2024.108507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2023] [Revised: 03/26/2024] [Accepted: 04/21/2024] [Indexed: 04/26/2024]
Abstract
BACKGROUND Post COVID-19 Condition (PCC), characterized by lingering symptoms post-acute COVID-19, poses clinical challenges, highlighting the need to understand its underlying molecular mechanisms. This meta-analysis aims to shed light on the transcriptomic landscapes and sex-specific molecular dynamics intrinsic to PCC. METHODS A systematic review identified three studies suitable for comprehensive meta-analysis, encompassing 135 samples (57 PCC subjects and 78 recovered subjects). We performed meta-analysis on differential gene expression, a gene set enrichment analysis of Reactome pathways, and weighted gene co-expression network analysis (WGCNA). We performed a drug and disease enrichment analysis and also assessed sex-specific differences in expression patterns. KEY FINDINGS A clear difference was observed in the transcriptomic profiles of PCC subjects, with 530 differentially expressed genes (DEGs) identified. Enrichment analysis revealed that the altered pathways were predominantly implicated in cell cycle processes, immune dysregulation and histone modifications. Antioxidant compounds such as hesperitin were predominantly linked to the hub genes of the DEGs. Sex-specific analyses highlighted disparities in DEGs and altered pathways in male and female PCC patients, revealing a difference in the expression of ribosomal proteins. PCC in men was mostly linked to neuro-cardiovascular disorders, while women exhibited more diverse disorders, with a high index of respiratory conditions. CONCLUSION Our study reveals the intricate molecular processes underlying PCC, highlighting that the differences in molecular dynamics between males and females could be key to understanding and effectively managing the varied symptomatology of this condition.
Collapse
Affiliation(s)
- Elena Cristina Rusu
- GEMMAIR Research Unit (AGAUR) - Applied Medicine (URV), Department of Medicine and Surgery. University Rovira i Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain; Institute for Integrative Systems Biology (I2SysBio), University of Valencia and the Spanish National Research Council (CSIC), 46980, Valencia, Spain.
| | - Pablo Monfort-Lanzas
- Institute of Medical Biochemistry, Biocenter, Medical University of Innsbruck, 6020, Innsbruck, Austria; Institute of Bioinformatics, Biocenter, Medical University of Innsbruck, 6020, Innsbruck, Austria.
| | - Laia Bertran
- GEMMAIR Research Unit (AGAUR) - Applied Medicine (URV), Department of Medicine and Surgery. University Rovira i Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Andrea Barrientos-Riosalido
- GEMMAIR Research Unit (AGAUR) - Applied Medicine (URV), Department of Medicine and Surgery. University Rovira i Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Emilia Solé
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| | - Razieh Mahmoudian
- GEMMAIR Research Unit (AGAUR) - Applied Medicine (URV), Department of Medicine and Surgery. University Rovira i Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Carmen Aguilar
- GEMMAIR Research Unit (AGAUR) - Applied Medicine (URV), Department of Medicine and Surgery. University Rovira i Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain.
| | - Silvia Briansó
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| | - Fadel Mohamed
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| | - Susana Garcia
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| | - Javier Camaron
- Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| | - Teresa Auguet
- GEMMAIR Research Unit (AGAUR) - Applied Medicine (URV), Department of Medicine and Surgery. University Rovira i Virgili (URV), Health Research Institute Pere Virgili (IISPV), 43007, Tarragona, Spain; Internal Medicine Unit, Joan XXIII University Hospital of Tarragona, 43007, Tarragona, Spain.
| |
Collapse
|
10
|
Bi Y, Li T, Zhang S, Yang Y, Dong M. Bioinformatics-based analysis of the dialog between COVID-19 and RSA. Heliyon 2024; 10:e30371. [PMID: 38737245 PMCID: PMC11088317 DOI: 10.1016/j.heliyon.2024.e30371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 04/24/2024] [Accepted: 04/24/2024] [Indexed: 05/14/2024] Open
Abstract
Pregnant women infected with SARS-CoV-2 in early pregnancy may face an increased risk of miscarriage due to immune imbalance at the maternal-fetal interface. However, the molecular mechanisms underlying the crosstalk between COVID-19 infection and recurrent spontaneous abortion (RSA) remain poorly understood. This study aimed to elucidate the transcriptomic molecular dialog between COVID-19 and RSA. Based on bioinformatics analysis, 307 common differentially expressed genes were found between COVID-19 (GSE171110) and RSA (GSE165004). Common DEGs were mainly enriched in ribosome-related and cell cycle-related signaling pathways. Using degree algorithm, the top 10 hub genes (RPS27A, RPL5, RPS8, RPL4, RPS2, RPL30, RPL23A, RPL31, RPL26, RPL37A) were selected from the common DEGs based on their scores. The results of the qPCR were in general agreement with the results of the raw letter analysis. The top 10 candidate drugs were also selected based on P-values. In this study, we provide molecular markers, signaling pathways, and small molecule compounds that may associate COVID-19. These findings may increase the accurate diagnosis and treatment of COVID-19 patients.
Collapse
Affiliation(s)
- Yin Bi
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530000, China
| | - Ting Li
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530000, China
| | - Shun Zhang
- Department of Reproductive Medical Center, The Affiliated Hospital of Guilin Medical University, Guilin, Guangxi 541001, China
| | - Yihua Yang
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Guangxi Medical University, Ministry of Education, Nanning, 530000, China
| | - Mingyou Dong
- Guangxi Reproductive Medical Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530000, China
- The Key Laboratory of Molecular Pathology (For Hepatobiliary Diseases) of Guangxi, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise, 533000, China
| |
Collapse
|
11
|
Zhou X, Huang T, Pan H, Du A, Wu T, Lan J, Song Y, Lv Y, He F, Yuan K. Bioinformatics and system biology approaches to determine the connection of SARS-CoV-2 infection and intrahepatic cholangiocarcinoma. PLoS One 2024; 19:e0300441. [PMID: 38648205 PMCID: PMC11034673 DOI: 10.1371/journal.pone.0300441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/27/2024] [Indexed: 04/25/2024] Open
Abstract
INTRODUCTION Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causal agent of coronavirus disease 2019 (COVID-19), has infected millions of individuals worldwide, which poses a severe threat to human health. COVID-19 is a systemic ailment affecting various tissues and organs, including the lungs and liver. Intrahepatic cholangiocarcinoma (ICC) is one of the most common liver cancer, and cancer patients are particularly at high risk of SARS-CoV-2 infection. Nonetheless, few studies have investigated the impact of COVID-19 on ICC patients. METHODS With the methods of systems biology and bioinformatics, this study explored the link between COVID-19 and ICC, and searched for potential therapeutic drugs. RESULTS This study identified a total of 70 common differentially expressed genes (DEGs) shared by both diseases, shedding light on their shared functionalities. Enrichment analysis pinpointed metabolism and immunity as the primary areas influenced by these common genes. Subsequently, through protein-protein interaction (PPI) network analysis, we identified SCD, ACSL5, ACAT2, HSD17B4, ALDOA, ACSS1, ACADSB, CYP51A1, PSAT1, and HKDC1 as hub genes. Additionally, 44 transcription factors (TFs) and 112 microRNAs (miRNAs) were forecasted to regulate the hub genes. Most importantly, several drug candidates (Periodate-oxidized adenosine, Desipramine, Quercetin, Perfluoroheptanoic acid, Tetrandrine, Pentadecafluorooctanoic acid, Benzo[a]pyrene, SARIN, Dorzolamide, 8-Bromo-cAMP) may prove effective in treating ICC and COVID-19. CONCLUSION This study is expected to provide valuable references and potential drugs for future research and treatment of COVID-19 and ICC.
Collapse
Affiliation(s)
- Xinyi Zhou
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tengda Huang
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Hongyuan Pan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Ao Du
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Tian Wu
- NHC Key Laboratory of Transplant Engineering and Immunology, Regenerative Medicine Research Center, Frontiers Science Center for Disease-related Molecular Network, West China Hospital of Sichuan University, Chengdu, China
| | - Jiang Lan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yujia Song
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yue Lv
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Fang He
- Center of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Kefei Yuan
- Division of Liver Surgery, Department of General Surgery and Laboratory of Liver Surgery, and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
12
|
Nobel FA, Kamruzzaman M, Asaduzzaman M, Uddin MN, Ahammad H, Hasan MM, Kar TR, Juliana FM, Babu G, Islam MJ. Identification of Differentially Expressed Genes and Protein-Protein Interaction in Patients With COVID-19 and Diabetes Peripheral Neuropathy: A Bioinformatics and System Biology Approach. Cureus 2024; 16:e58548. [PMID: 38957825 PMCID: PMC11218505 DOI: 10.7759/cureus.58548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/18/2024] [Indexed: 07/04/2024] Open
Abstract
The coronavirus disease 2019 (COVID-19) pandemic has had a significant impact globally, resulting in a higher death toll and persistent health issues for survivors, particularly those with pre-existing medical conditions. Numerous studies have demonstrated a strong correlation between catastrophic COVID-19 results and diabetes. To gain deeper insights, we analysed the transcriptome dataset from COVID-19 and diabetic peripheral neuropathic patients. Using the R programming language, differentially expressed genes (DEGs) were identified and classified based on up and down regulations. The overlaps of DEGs were then explored between these groups. Functional annotation of those common DEGs was performed using Gene Ontology (GO), Kyoto Encyclopedia of Genes and Genomes (KEGG), Bio-Planet, Reactome, and Wiki pathways. A protein-protein interaction (PPI) network was created with bioinformatics tools to understand molecular interactions. Through topological analysis of the PPI network, we determined hub gene modules and explored gene regulatory networks (GRN). Furthermore, the study extended to suggesting potential drug molecules for the identified mutual DEG based on the comprehensive analysis. These approaches may contribute to understanding the molecular intricacies of COVID-19 in diabetic peripheral neuropathy patients through insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Fahim Alam Nobel
- Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, BGD
| | - Mohammad Kamruzzaman
- Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, BGD
| | - Mohammad Asaduzzaman
- Biochemistry and Molecular Biology, Noakhali Science and Technology University, Noakhali, BGD
| | - Mohammad Nasir Uddin
- Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, BGD
| | - Hasib Ahammad
- Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, BGD
| | - Mehedi Mahmudul Hasan
- Fisheries and Marine Science, Noakhali Science and Technology University, Noakhali, BGD
| | - Tanu Rani Kar
- Biochemistry and Molecular Biology, Primeasia University, Dhaka, BGD
| | - Farha Matin Juliana
- Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, BGD
| | - Golap Babu
- Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, BGD
| | - Mohammod Johirul Islam
- Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, BGD
| |
Collapse
|
13
|
Ettitaou A, Kabdy H, Oubella K, Raoui K, Oubahmane M, Aboufatima R, Elyazouli L, Garzoli S, Chait A. Molecular docking of quercetin: a promising approach for the development of new anti-inflammatory and analgesic drugs. Nat Prod Res 2024:1-10. [PMID: 38520257 DOI: 10.1080/14786419.2024.2333053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024]
Abstract
The aim of this study is to investigate the antinociceptive, anti-inflammatory and antipyretic effects of quercetin. Additionally, molecular docking studies were conducted to evaluate potential interactions between quercetin and various molecular targets. Animal models were used to conduct a comprehensive pharmacological investigation of quercetin. Evaluation of analgesic activity revealed a reduction in the number of abdominal cramps during the twisting test and inhibition of pain during the second phase of the formaldehyde test. Additionally, evaluation of its anti-inflammatory activity showed a reduction in ear oedema. However, it is important to note that quercetin administration has not been shown to significantly reduce yeast-induced hyperthermia. The docking study revealed the high inhibitory potential of quercetin against the COX-2 receptor.
Collapse
Affiliation(s)
- Amina Ettitaou
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Hamid Kabdy
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Khadija Oubella
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Karima Raoui
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Mehdi Oubahmane
- Laboratory of Molecular Chemistry, Department of Chemistry, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| | - Rachida Aboufatima
- Laboratory of Genie Biologic, Faculty of Sciences and Technics, Sultan Moulay Slimane University, Beni Mellal, Morocco
| | - Loubna Elyazouli
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| | - Stefania Garzoli
- Department of Chemistry and Technologies of Drug, Sapienza University, Rome, Italy
| | - Abderrahman Chait
- Laboratory of Pharmacology, Neurobiology, Anthropology and Environment, Department of Biology, Faculty of Sciences Semlalia, University Cadi Ayyad, Marrakech, Morocco
| |
Collapse
|
14
|
Li D, Chen R, Huang C, Zhang G, Li Z, Xu X, Wang B, Li B, Chu XM. Comprehensive bioinformatics analysis and systems biology approaches to identify the interplay between COVID-19 and pericarditis. Front Immunol 2024; 15:1264856. [PMID: 38455049 PMCID: PMC10918693 DOI: 10.3389/fimmu.2024.1264856] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 02/08/2024] [Indexed: 03/09/2024] Open
Abstract
Background Increasing evidence indicating that coronavirus disease 2019 (COVID-19) increased the incidence and related risks of pericarditis and whether COVID-19 vaccine is related to pericarditis has triggered research and discussion. However, mechanisms behind the link between COVID-19 and pericarditis are still unknown. The objective of this study was to further elucidate the molecular mechanisms of COVID-19 with pericarditis at the gene level using bioinformatics analysis. Methods Genes associated with COVID-19 and pericarditis were collected from databases using limited screening criteria and intersected to identify the common genes of COVID-19 and pericarditis. Subsequently, gene ontology, pathway enrichment, protein-protein interaction, and immune infiltration analyses were conducted. Finally, TF-gene, gene-miRNA, gene-disease, protein-chemical, and protein-drug interaction networks were constructed based on hub gene identification. Results A total of 313 common genes were selected, and enrichment analyses were performed to determine their biological functions and signaling pathways. Eight hub genes (IL-1β, CD8A, IL-10, CD4, IL-6, TLR4, CCL2, and PTPRC) were identified using the protein-protein interaction network, and immune infiltration analysis was then carried out to examine the functional relationship between the eight hub genes and immune cells as well as changes in immune cells in disease. Transcription factors, miRNAs, diseases, chemicals, and drugs with high correlation with hub genes were predicted using bioinformatics analysis. Conclusions This study revealed a common gene interaction network between COVID-19 and pericarditis. The screened functional pathways, hub genes, potential compounds, and drugs provided new insights for further research on COVID-19 associated with pericarditis.
Collapse
Affiliation(s)
- Daisong Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Ruolan Chen
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Chao Huang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Guoliang Zhang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhaoqing Li
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xiaojian Xu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Banghui Wang
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Bing Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Department of Dermatology, The Affiliated Haici Hospital of Qingdao University, Qingdao, China
| | - Xian-Ming Chu
- Department of Cardiology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Department of Cardiology, The Affiliated Cardiovascular Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
15
|
Xu J, Abdulsalam Khaleel R, Zaidan HK, Faisal Mutee A, Fahmi Fawy K, Gehlot A, Abbas AH, Arias Gonzáles JL, Amin AH, Ruiz-Balvin MC, Imannezhad S, Bahrami A, Akhavan-Sigari R. Discovery of common molecular signatures and drug repurposing for COVID-19/Asthma comorbidity: ACE2 and multi-partite networks. Cell Cycle 2024; 23:405-434. [PMID: 38640424 PMCID: PMC11529202 DOI: 10.1080/15384101.2024.2340859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 01/15/2024] [Accepted: 04/04/2024] [Indexed: 04/21/2024] Open
Abstract
Angiotensin-converting enzyme 2 (ACE2) is identified as the functional receptor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causative agent of the ongoing global coronavirus disease-2019 (COVID-19) pandemic. This study aimed to elucidate potential therapeutic avenues by scrutinizing approved drugs through the identification of the genetic signature associated with SARS-CoV-2 infection in individuals with asthma. This exploration was conducted through an integrated analysis, encompassing interaction networks between the ACE2 receptor and common host (co-host) factors implicated in COVID-19/asthma comorbidity. The comprehensive analysis involved the identification of common differentially expressed genes (cDEGs) and hub-cDEGs, functional annotations, interaction networks, gene set variation analysis (GSVA), gene set enrichment analysis (GSEA), and module construction. Interaction networks were used to identify overlapping disease modules and potential drug targets. Computational biology and molecular docking analyzes were utilized to discern functional drug modules. Subsequently, the impact of the identified drugs on the expression of hub-cDEGs was experimentally validated using a mouse model. A total of 153 cDEGs or co-host factors associated with ACE2 were identified in the COVID-19 and asthma comorbidity. Among these, seven significant cDEGs and proteins - namely, HRAS, IFNG, JUN, CDH1, TLR4, ICAM1, and SCD-were recognized as pivotal host factors linked to ACE2. Regulatory network analysis of hub-cDEGs revealed eight top-ranked transcription factors (TFs) proteins and nine microRNAs as key regulatory factors operating at the transcriptional and post-transcriptional levels, respectively. Molecular docking simulations led to the proposal of 10 top-ranked repurposable drug molecules (Rapamycin, Ivermectin, Everolimus, Quercetin, Estradiol, Entrectinib, Nilotinib, Conivaptan, Radotinib, and Venetoclax) as potential treatment options for COVID-19 in individuals with comorbid asthma. Validation analysis demonstrated that Rapamycin effectively inhibited ICAM1 expression in the HDM-stimulated mice group (p < 0.01). This study unveils the common pathogenesis and genetic signature underlying asthma and SARS-CoV-2 infection, delineated by the interaction networks of ACE2-related host factors. These findings provide valuable insights for the design and discovery of drugs aimed at more effective therapeutics within the context of lung disease comorbidities.
Collapse
Affiliation(s)
- Jiajun Xu
- College of Veterinary & Life Sciences, the University of Glasgow, Glasgow, UK
| | | | | | | | - Khaled Fahmi Fawy
- Department of Chemistry, Faculty of Science, King Khalid University, Abha, Saudi Arabia
| | - Anita Gehlot
- Uttaranchal Institute of Technology, Uttaranchal University, Dehradun, India
| | | | - José Luis Arias Gonzáles
- Department of Social Sciences, Faculty of Social Studies, University of British Columbia, Vancouver, Canada
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | | | - Shima Imannezhad
- Department of Pediatrics, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abolfazl Bahrami
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Reza Akhavan-Sigari
- Department of Neurosurgery, University Medical Center Tuebingen, Tuebingen, Germany
- Department of Health Care Management and Clinical Research, Collegium Humanum, Warsaw, Poland
| |
Collapse
|
16
|
Chu Y, Li M, Sun M, Wang J, Xin W, Xu L. Gene crosstalk between COVID-19 and preeclampsia revealed by blood transcriptome analysis. Front Immunol 2024; 14:1243450. [PMID: 38259479 PMCID: PMC10800816 DOI: 10.3389/fimmu.2023.1243450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 12/14/2023] [Indexed: 01/24/2024] Open
Abstract
Background The extensive spread of coronavirus disease 2019 (COVID-19) has led to a rapid increase in global mortality. Preeclampsia is a commonly observed pregnancy ailment characterized by high maternal morbidity and mortality rates, in addition to the restriction of fetal growth within the uterine environment. Pregnant individuals afflicted with vascular disorders, including preeclampsia, exhibit an increased susceptibility to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection via mechanisms that have not been fully delineated. Additionally, the intricate molecular mechanisms underlying preeclampsia and COVID-19 have not been fully elucidated. This study aimed to discern commonalities in gene expression, regulators, and pathways shared between COVID-19 and preeclampsia. The objective was to uncover potential insights that could contribute to novel treatment strategies for both COVID-19 and preeclampsia. Method Transcriptomic datasets for COVID-19 peripheral blood (GSE152418) and preeclampsia blood (GSE48424) were initially sourced from the Gene Expression Omnibus (GEO) database. Subsequent to that, we conducted a subanalysis by selecting females from the GSE152418 dataset and employed the "Deseq2" package to identify genes that exhibited differential expression. Simultaneously, the "limma" package was applied to identify differentially expressed genes (DEGs) in the preeclampsia dataset (GSE48424). Following that, an intersection analysis was conducted to identify the common DEGs obtained from both the COVID-19 and preeclampsia datasets. The identified shared DEGs were subsequently utilized for functional enrichment analysis, transcription factor (TF) and microRNAs (miRNA) prediction, pathway analysis, and identification of potential candidate drugs. Finally, to validate the bioinformatics findings, we collected peripheral blood mononuclear cell (PBMC) samples from healthy individuals, COVID-19 patients, and Preeclampsia patients. The abundance of the top 10 Hub genes in both diseases was assessed using real-time quantitative polymerase chain reaction (RT-qPCR). Result A total of 355 overlapping DEGs were identified in both preeclampsia and COVID-19 datasets. Subsequent ontological analysis, encompassing Gene Ontology (GO) functional assessment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis, revealed a significant association between the two conditions. Protein-protein interactions (PPIs) were constructed using the STRING database. Additionally, the top 10 hub genes (MRPL11, MRPS12, UQCRH, ATP5I, UQCRQ, ATP5D, COX6B1, ATP5O, ATP5H, NDUFA6) were selected based on their ranking scores using the degree algorithm, which considered the shared DEGs. Moreover, transcription factor-gene interactions, protein-drug interactions, co-regulatory networks of DEGs and miRNAs, and protein-drug interactions involving the shared DEGs were also identified in the datasets. Finally, RT-PCR results confirmed that 10 hub genes do exhibit distinct expression profiles in the two diseases. Conclusion This study successfully identified overlapping DEGs, functional pathways, and regulatory elements between COVID-19 and preeclampsia. The findings provide valuable insights into the shared molecular mechanisms and potential therapeutic targets for both diseases. The validation through RT-qPCR further supports the distinct expression profiles of the identified hub genes in COVID-19 and preeclampsia, emphasizing their potential roles as biomarkers or therapeutic targets in these conditions.
Collapse
Affiliation(s)
| | | | | | | | | | - Lin Xu
- Department of Obstetrics, the Affiliated Hospital of Qingdao University, Qingdao, China
| |
Collapse
|
17
|
Zhang P, He J, Gan Y, Shang Q, Chen H, Zhao W, Cui J, Shen G, Li Y, Jiang X, Zhu G, Ren H. Unravelling diagnostic clusters and immune landscapes of cuproptosis patterns in intervertebral disc degeneration through dry and wet experiments. Aging (Albany NY) 2023; 15:15599-15623. [PMID: 38159257 PMCID: PMC10781477 DOI: 10.18632/aging.205449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 12/07/2023] [Indexed: 01/03/2024]
Abstract
Cuproptosis is a manner of mitochondrial cell death induced by copper. However, cuproptosis modulators' molecular processes in intervertebral disc degeneration (IDD) are still unclear. To better understand the processes of cuproptosis regulators in IDD, a thorough analysis of cuproptosis regulators in the diagnostic biomarkers and subtype determination of IDD was conducted. Then we collected clinical IDD samples and successfully established IDD model in vivo and in vitro, and carried out real-time quantitative polymerase chain reaction (RT-qPCR) validation of significant cuproptosis modulators. Totally we identified 8 crucial cuproptosis regulators in the present research. Using a random forest model, we isolated 8 diagnostic cuproptosis modulators for the prediction of IDD risk. Then, based on our following decision curve analysis, we selected the five diagnostic cuproptosis regulators with importance scores greater than two and built a nomogram model. Using a consensus clustering method, we divided IDD patients into two cuproptosis clusters (clusterA and clusterB) based on the important cuproptosis regulators. Additionally, each sample's cuproptosis value was evaluated using principal component analysis in order to quantify the cuproptosis clusters. Patients in clusterB had higher cuproptosis scores than patients in clusterA. Moreover, we found that clusterB was involved in the immunity of natural killer cell, while clusterA was related to activated CD4 T cell, activated B cell, etc. Notably, cuproptosis modulators detected by RT-qPCR showed generally consistent expression levels with the bioinformatics results. To sum up, cuproptosis modulators play a crucial role in the pathogenic process of IDD, providing biomarkers and immunotherapeutic approaches for IDD.
Collapse
Affiliation(s)
- Peng Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Jiahui He
- The Affiliated TCM Hospital of Guangzhou Medical University, Guangzhou 510130, China
| | - Yanchi Gan
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Qi Shang
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Honglin Chen
- Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Wenhua Zhao
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Jianchao Cui
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, China
| | - Gengyang Shen
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Yuwei Li
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215007, China
| | - Xiaobing Jiang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| | - Guangye Zhu
- Suzhou TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Suzhou 215007, China
| | - Hui Ren
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou 510260, China
| |
Collapse
|
18
|
Guo J, Zhang Y, Gao Y, Li S, Xu G, Tian Z, Xu Q, Li X, Li Y, Zhang Y. Systematical analyses of large-scale transcriptome reveal viral infection-related genes and disease comorbidities. ARTIFICIAL CELLS, NANOMEDICINE, AND BIOTECHNOLOGY 2023; 51:453-465. [PMID: 37651591 DOI: 10.1080/21691401.2023.2252477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 08/13/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
Perturbation of transcriptome in viral infection patients is a recurrent theme impacting symptoms and mortality, yet a detailed understanding of pertinent transcriptome and identification of robust biomarkers is not complete. In this study, we manually collected 23 datasets related to 6,197 blood transcriptomes across 16 types of respiratory virus infections. We applied a comprehensive systems biology approach starting with whole-blood transcriptomes combined with multilevel bioinformatics analyses to characterize the expression, functional pathways, and protein-protein interaction (PPI) networks to identify robust biomarkers and disease comorbidities. Robust gene markers of infection with different viruses were identified, which can accurately classify the normal and infected patients in train and validation cohorts. The biological processes (BP) of different viruses showed great similarity and enriched in infection and immune response pathways. Network-based analyses revealed that a variety of viral infections were associated with nervous system diseases, neoplasms and metabolic diseases, and significantly correlated with brain tissues. In summary, our manually collected transcriptomes and comprehensive analyses reveal key molecular markers and disease comorbidities in the process of viral infection, which could provide a valuable theoretical basis for the prevention of subsequent public health events for respiratory virus infections.
Collapse
Affiliation(s)
- Jing Guo
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Ya Zhang
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yueying Gao
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Si Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Gang Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Zhanyu Tian
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Qi Xu
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Xia Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| | - Yongsheng Li
- Key Laboratory of Tropical Translational Medicine of Ministry of Education, College of Biomedical Information and Engineering, Hainan Women and Children's Medical Center, Hainan Medical University, Haikou, Hainan, China
| | - Yunpeng Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, China
| |
Collapse
|
19
|
Muneeb Hassan M, Ameeq M, Jamal F, Tahir MH, Mendy JT. Prevalence of covid-19 among patients with chronic obstructive pulmonary disease and tuberculosis. Ann Med 2023; 55:285-291. [PMID: 36594409 PMCID: PMC9815254 DOI: 10.1080/07853890.2022.2160491] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND The exhaustive information about non-communicable diseases associated with COVID-19 and severe acute respiratory syndrome corona virus-2 (SARS-CoV-2) are getting easier to find in the literature. However, there is a lack of knowledge regarding tuberculosis (TB) and chronic obstructed pulmonary disease (COPD), with numerous infections in COVID-19 patients. OBJECTIVES Priority is placed on determining the patient's prognosis based on the presence or absence of TB and COPD. Additionally, a comparison is made between the risk of death and the likelihood of recovery in terms of time in COVID-19 patients who have either COPD or TB. METHODOLOGY At the DHQ Hospital in Muzaffargarh, Punjab, Pakistan, 498 COVID-19 patients with TB and COPD were studied retrospectively. The duration of study started in February 2022 and concluded in August 2022. The Kaplan-Meier curves described time-to-death and time-to-recovery stratified by TB and COPD status. The Wilcoxon test compared the survival rates of people with TB and COPD in two matched paired groups and their status differences with their standard of living. RESULTS The risk of death in COVID-19 patients with TB was 1.476 times higher than in those without (95% CI: 0.949-2.295). The recovery risk in COVID-19 patients with TB was 0.677 times lower than in those without (95% CI: 0.436-1.054). Similarly, patients with TB had a significantly shorter time to death (p=.001) and longer time to recovery (p=.001). CONCLUSIONS According to the findings, the most significant contributor to an increased risk of morbidity and mortality in TB and COPD patients was the COVID-19.KEY MESSAGESSARS-Cov-19 is a new challenge for the universe in terms of prevention and treatment for people with tuberculosis and chronic obstructive pulmonary disease, among other diseases.Propensity score matching to control for potential biases.Compared to hospitalized patients with and without (TB and COPD) had an equivalently higher mortality rate.
Collapse
Affiliation(s)
| | - Muhammad Ameeq
- Department of Statistics, The Islamia University, Bahawalpur, Pakistan
| | - Farrukh Jamal
- Department of Statistics, The Islamia University, Bahawalpur, Pakistan
| | - Muhammad H Tahir
- Department of Statistics, The Islamia University, Bahawalpur, Pakistan
| | - John T Mendy
- Department of Mathematics, School of Arts and Science, University of The Gambia, Serekunda, The Gambia
| |
Collapse
|
20
|
Zhao X, Liang Q, Li H, Jing Z, Pei D. Single-cell RNA sequencing and multiple bioinformatics methods to identify the immunity and ferroptosis-related biomarkers of SARS-CoV-2 infections to ischemic stroke. Aging (Albany NY) 2023; 15:8237-8257. [PMID: 37606960 PMCID: PMC10497002 DOI: 10.18632/aging.204966] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 07/20/2023] [Indexed: 08/23/2023]
Abstract
BACKGROUND Since December 2019, Coronavirus disease 2019 (COVID-19) induced by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has resulted in significant morbidity and mortality worldwide. There is an increased risk of ischemic stroke (IS) associated with COVID-19. However, few studies have been reported to explain the potential correlation between COVID-19 and IS. METHODS We investigated the relationship and relevant mechanisms between COVID-19 and IS using single-cell RNA sequencing and multiple bioinformatics approaches. RESULTS By intersecting differentially expressed genes and WGCNA critical module genes, we obtained 73 COVID-19-related IS genes. According to the KEGG pathway analysis, the COVID-19-related IS disease genes were significantly enriched in the hematopoietic cell lineage pathway, ribosome pathway, COVID-19 pathway and primary immunodeficiency pathway. Finally, three genes associated with immunity (B4GALT5, CRISPLD2, F5) and two genes associated with ferroptosis (ACSL1, CREB5) were identified up-regulated in COVID-19-related IS. Significantly, it was found that all five genes were highly expressed in monocytes by single cell RNA sequencing. CONCLUSION We believe these genes (B4GALT5, CRISPLD2, F5, ACSL1, CREB5) may regulate the immune response and ferroptosis of multiple immune cells, mainly including monocytes, which may contribute to the development of COVID-19-related IS. In addition, these genes may be potential targets for the treatment of COVID-19-related IS.
Collapse
Affiliation(s)
- Xiang Zhao
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Qingyu Liang
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Hao Li
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Zhitao Jing
- Department of Neurosurgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Dongmei Pei
- Department of Family Medicine, Shengjing Hospital, China Medical University, Shenyang, Liaoning 110001, China
| |
Collapse
|
21
|
Xi YJ, Guo Q, Zhang R, Duan GS, Zhang SX. Identifying cellular senescence associated genes involved in the progression of end-stage renal disease as new biomarkers. BMC Nephrol 2023; 24:231. [PMID: 37553608 PMCID: PMC10408218 DOI: 10.1186/s12882-023-03285-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Accepted: 07/28/2023] [Indexed: 08/10/2023] Open
Abstract
BACKGROUND Cellular senescence plays an essential role in the development and progression of end-stage renal disease (ESRD). However, the detailed mechanisms phenomenon remains unclear. METHODS The mRNA expression profiling dataset GSE37171 was taken from the Gene Expression Omnibus (GEO) database. The cell senescence-associated hub genes were selected by applying protein-protein interaction (PPI), followed by correlation analysis, gene interaction analysis, Gene Ontology (GO), and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analysis. We next explored the relationships of hub genes with miRNAs, TFs, and diseases. The absolute abundance of eight immune cells and two stromal cells were calculated by MCPcount and the correlation of hub genes with these ten cells was analyzed. Lasso was used to selecting for trait genes. ROC curves and DCA decision curves were used to assess the accuracy and predictive power of the trait genes. RESULTS A total of 65 cellular senescence signature genes were identified among patients and controls. The PPI network screened out ten hub genes. GO and KEGG indicated that ten hub genes were associated with ESRD progression. Transcription factor gene interactions and common regulatory networks of miRNAs were also identified in the datasets. The hub genes were significantly correlated with immune cells and stromal cells. Then the lasso model was constructed to screen out the five most relevant signature genes (FOS, FOXO3, SIRT1, TP53, SMARCA4). The area under the ROC curve (AUC) showed that these five characteristic genes have good resolving power for the diagnostic model. CONCLUSIONS Our findings suggested that cellular senescence-associated genes played an important role in the development of ESRD and immune regulation.
Collapse
Affiliation(s)
- Yu-Jia Xi
- Department of Urology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China
| | - Qiang Guo
- Department of Urology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Ran Zhang
- School of Public Health, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Guo-Sheng Duan
- Fifth School of Clinical Medicine, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, Shanxi Province, China
| | - Sheng-Xiao Zhang
- Key Laboratory of Cellular Physiology at Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi Province, China.
- Department of Rheumatology, Second Hospital of Shanxi Medical University, 382 Wuyi Road, Taiyuan, 030001, Shanxi Province, China.
| |
Collapse
|
22
|
Zhou H, Xu M, Hu P, Li Y, Ren C, Li M, Pan Y, Wang S, Liu X. Identifying hub genes and common biological pathways between COVID-19 and benign prostatic hyperplasia by machine learning algorithms. Front Immunol 2023; 14:1172724. [PMID: 37426635 PMCID: PMC10328422 DOI: 10.3389/fimmu.2023.1172724] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Accepted: 06/07/2023] [Indexed: 07/11/2023] Open
Abstract
Background COVID-19, a serious respiratory disease that has the potential to affect numerous organs, is a serious threat to the health of people around the world. The objective of this article is to investigate the potential biological targets and mechanisms by which SARS-CoV-2 affects benign prostatic hyperplasia (BPH) and related symptoms. Methods We downloaded the COVID-19 datasets (GSE157103 and GSE166253) and the BPH datasets (GSE7307 and GSE132714) from the Gene Expression Omnibus (GEO) database. In GSE157103 and GSE7307, differentially expressed genes (DEGs) were found using the "Limma" package, and the intersection was utilized to obtain common DEGs. Further analyses followed, including those using Protein-Protein Interaction (PPI), Gene Ontology (GO) function enrichment analysis, and the Kyoto Encyclopedia of Genes and Genomes (KEGG). Potential hub genes were screened using three machine learning methods, and they were later verified using GSE132714 and GSE166253. The CIBERSORT analysis and the identification of transcription factors, miRNAs, and drugs as candidates were among the subsequent analyses. Results We identified 97 common DEGs from GSE157103 and GSE7307. According to the GO and KEGG analyses, the primary gene enrichment pathways were immune-related pathways. Machine learning methods were used to identify five hub genes (BIRC5, DNAJC4, DTL, LILRB2, and NDC80). They had good diagnostic properties in the training sets and were validated in the validation sets. According to CIBERSORT analysis, hub genes were closely related to CD4 memory activated of T cells, T cells regulatory and NK cells activated. The top 10 drug candidates (lucanthone, phytoestrogens, etoposide, dasatinib, piroxicam, pyrvinium, rapamycin, niclosamide, genistein, and testosterone) will also be evaluated by the P value, which is expected to be helpful for the treatment of COVID-19-infected patients with BPH. Conclusion Our findings reveal common signaling pathways, possible biological targets, and promising small molecule drugs for BPH and COVID-19. This is crucial to understand the potential common pathogenic and susceptibility pathways between them.
Collapse
Affiliation(s)
- Hang Zhou
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Mingming Xu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Ping Hu
- Department of Orthopedics, Tianjin Medical University General Hospital, Tianjin, China
| | - Yuezheng Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Congzhe Ren
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Muwei Li
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Yang Pan
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Shangren Wang
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaoqiang Liu
- Department of Urology, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
23
|
Chen A, Sun Z, Sun D, Huang M, Fang H, Zhang J, Qian G. Integrative bioinformatics and validation studies reveal KDM6B and its associated molecules as crucial modulators in Idiopathic Pulmonary Fibrosis. Front Immunol 2023; 14:1183871. [PMID: 37275887 PMCID: PMC10235501 DOI: 10.3389/fimmu.2023.1183871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 05/08/2023] [Indexed: 06/07/2023] Open
Abstract
Background Idiopathic Pulmonary Fibrosis (IPF) can be described as a debilitating lung disease that is characterized by the complex interactions between various immune cell types and signaling pathways. Chromatin-modifying enzymes are significantly involved in regulating gene expression during immune cell development, yet their role in IPF is not well understood. Methods In this study, differential gene expression analysis and chromatin-modifying enzyme-related gene data were conducted to identify hub genes, common pathways, immune cell infiltration, and potential drug targets for IPF. Additionally, a murine model was employed for investigating the expression levels of candidate hub genes and determining the infiltration of different immune cells in IPF. Results We identified 33 differentially expressed genes associated with chromatin-modifying enzymes. Enrichment analyses of these genes demonstrated a strong association with histone lysine demethylation, Sin3-type complexes, and protein demethylase activity. Protein-protein interaction network analysis further highlighted six hub genes, specifically KDM6B, KDM5A, SETD7, SUZ12, HDAC2, and CHD4. Notably, KDM6B expression was significantly increased in the lungs of bleomycin-induced pulmonary fibrosis mice, showing a positive correlation with fibronectin and α-SMA, two essential indicators of pulmonary fibrosis. Moreover, we established a diagnostic model for IPF focusing on KDM6B and we also identified 10 potential therapeutic drugs targeting KDM6B for IPF treatment. Conclusion Our findings suggest that molecules related to chromatin-modifying enzymes, primarily KDM6B, play a critical role in the pathogenesis and progression of IPF.
Collapse
Affiliation(s)
- Anning Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhun Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Donglin Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Meiying Huang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Hongwei Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jinyuan Zhang
- Department of Pain, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Guojun Qian
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
24
|
Li P, Li T, Zhang Z, Dai X, Zeng B, Li Z, Li Z. Bioinformatics and system biology approach to identify the influences among COVID-19, ARDS and sepsis. Front Immunol 2023; 14:1152186. [PMID: 37261353 PMCID: PMC10227520 DOI: 10.3389/fimmu.2023.1152186] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/27/2023] [Indexed: 06/02/2023] Open
Abstract
Background Severe coronavirus disease 2019 (COVID -19) has led to severe pneumonia or acute respiratory distress syndrome (ARDS) worldwide. we have noted that many critically ill patients with COVID-19 present with typical sepsis-related clinical manifestations, including multiple organ dysfunction syndrome, coagulopathy, and septic shock. The molecular mechanisms that underlie COVID-19, ARDS and sepsis are not well understood. The objectives of this study were to analyze potential molecular mechanisms and identify potential drugs for the treatment of COVID-19, ARDS and sepsis using bioinformatics and a systems biology approach. Methods Three RNA-seq datasets (GSE171110, GSE76293 and GSE137342) from Gene Expression Omnibus (GEO) were employed to detect mutual differentially expressed genes (DEGs) for the patients with the COVID-19, ARDS and sepsis for functional enrichment, pathway analysis, and candidate drugs analysis. Results We obtained 110 common DEGs among COVID-19, ARDS and sepsis. ARG1, FCGR1A, MPO, and TLR5 are the most influential hub genes. The infection and immune-related pathways and functions are the main pathways and molecular functions of these three diseases. FOXC1, YY1, GATA2, FOXL, STAT1 and STAT3 are important TFs for COVID-19. mir-335-5p, miR-335-5p and hsa-mir-26a-5p were associated with COVID-19. Finally, the hub genes retrieved from the DSigDB database indicate multiple drug molecules and drug-targets interaction. Conclusion We performed a functional analysis under ontology terms and pathway analysis and found some common associations among COVID-19, ARDS and sepsis. Transcription factors-genes interaction, protein-drug interactions, and DEGs-miRNAs coregulatory network with common DEGs were also identified on the datasets. We believe that the candidate drugs obtained in this study may contribute to the effective treatment of COVID-19.
Collapse
Affiliation(s)
- Peiyu Li
- Department of Gastroenterology, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, China
| | - Tao Li
- Department of Critical Care Medicine, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
- Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Zhiming Zhang
- Department of Anesthesiology, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Xingui Dai
- Department of Critical Care Medicine, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Bin Zeng
- Department of Anesthesiology, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Zhen Li
- Department of Anesthesiology, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
| | - Zhiwang Li
- Department of Anesthesiology, The First People’s Hospital of Chenzhou, Chenzhou, Hunan, China
| |
Collapse
|
25
|
Deng X, Luo Y, Guan T, Guo X. Identification of the Genetic Influence of SARS-CoV-2 Infections on IgA Nephropathy Based on Bioinformatics Method. Kidney Blood Press Res 2023; 48:367-384. [PMID: 37040729 PMCID: PMC10308545 DOI: 10.1159/000529687] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 02/09/2023] [Indexed: 04/13/2023] Open
Abstract
INTRODUCTION Coronavirus disease-2019 (COVID-19) is caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection. It was initially detected in Wuhan, China, in December 2019. In March 2020, the World Health Organization (WHO) declared COVID-19 a global pandemic. Compared to healthy individuals, patients with IgA nephropathy (IgAN) are at a higher risk of SARS-CoV-2 infection. However, the potential mechanisms remain unclear. This study explores the underlying molecular mechanisms and therapeutic agents for the management of IgAN and COVID-19 using the bioinformatics and system biology way. METHODS We first downloaded GSE73953 and GSE164805 from the Gene Expression Omnibus (GEO) database to obtain common differentially expressed genes (DEGs). Then, we performed the functional enrichment analysis, pathway analysis, protein-protein interaction (PPI) analysis, gene regulatory networks analysis, and potential drug analysis on these common DEGs. RESULTS We acquired 312 common DEGs from the IgAN and COVID-19 datasets and used various bioinformatics tools and statistical analyses to construct the PPI network to extract hub genes. Besides, we performed gene ontology (GO) and pathway analyses to reveal the common correlation between IgAN and COVID-19. Finally, on the basis of common DEGs, we determined the interactions between DEGs-miRNAs, the transcription factor-genes (TFs-genes), protein-drug, and gene-disease networks. CONCLUSION We successfully identified hub genes that may act as biomarkers of COVID-19 and IgAN and also screened out some potential drugs to provide new ideas for COVID-19 and IgAN treatment.
Collapse
Affiliation(s)
- Xiaoqi Deng
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Yu Luo
- School of Medicine, Xiamen University, Xiamen, China
| | - Tianjun Guan
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Xiaodan Guo
- Department of Nephrology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
26
|
Zhang C, Ma Z, Nan X, Wang W, Zeng X, Chen J, Cai Z, Wang J. Comprehensive analysis to identify the influences of SARS-CoV-2 infections to inflammatory bowel disease. Front Immunol 2023; 14:1024041. [PMID: 36817436 PMCID: PMC9936160 DOI: 10.3389/fimmu.2023.1024041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 01/23/2023] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Coronavirus disease 2019 (COVID-19) and inflammatory bowel disease (IBD) are both caused by a disordered immune response and have direct and profound impacts on health care services. In this study, we implemented transcriptomic and single-cell analysis to detect common molecular and cellular intersections between COVID-19 and IBD that help understand the linkage of COVID-19 to the IBD patients. METHODS Four RNA-sequencing datasets (GSE147507, GSE126124, GSE9686 and GSE36807) from Gene Expression Omnibus (GEO) database are extracted to detect mutual differentially expressed genes (DEGs) for IBD patients with the infection of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to find shared pathways, candidate drugs, hub genes and regulatory networks. Two single-cell RNA sequencing (scRNA-eq) datasets (GSE150728, PRJCA003980) are used to analyze the immune characteristics of hub genes and the proportion of immune cell types, so as to find common immune responses between COVID-19 and IBD. RESULTS A total of 121 common DEGs were identified among four RNA-seq datasets, and were all involved in the functional enrichment analysis related to inflammation and immune response. Transcription factors-DEGs interactions, miRNAs-DEGs coregulatory networks, and protein-drug interactions were identified based on these datasets. Protein-protein interactions (PPIs) was built and 59 hub genes were identified. Moreover, scRNA-seq of peripheral blood monocyte cells (PBMCs) from COVID-19 patients revealed a significant increase in the proportion of CD14+ monocytes, in which 38 of 59 hub genes were highly enriched. These genes, encoding inflammatory cytokines, were also highly expressed in inflammatory macrophages (IMacrophage) of intestinal tissues of IBD patients. CONCLUSIONS We conclude that COVID-19 may promote the progression of IBD through cytokine storms. The candidate drugs and DEGs-regulated networks may suggest effective therapeutic methods for both COVID-19 and IBD.
Collapse
Affiliation(s)
- Chengyan Zhang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zeyu Ma
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xi Nan
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Wenhui Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xianchang Zeng
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinming Chen
- Department of Anorectal, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
- Department of Orthopaedics of the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jianli Wang
- Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, China
| |
Collapse
|
27
|
Identification of Potential Key Genes and Prognostic Biomarkers of Lung Cancer Based on Bioinformatics. BIOMED RESEARCH INTERNATIONAL 2023; 2023:2152432. [PMID: 36714024 PMCID: PMC9876670 DOI: 10.1155/2023/2152432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 10/31/2022] [Accepted: 11/17/2022] [Indexed: 01/19/2023]
Abstract
Objective To analyze and identify the core genes related to the expression and prognosis of lung cancer including lung adenocarcinoma (LUAD) and lung squamous cell carcinoma (LUSC) by bioinformatics technology, with the aim of providing a reference for clinical treatment. Methods Five sets of gene chips, GSE7670, GSE151102, GSE33532, GSE43458, and GSE19804, were obtained from the Gene Expression Omnibus (GEO) database. After using GEO2R to analyze the differentially expressed genes (DEGs) between lung cancer and normal tissues online, the common DEGs of the five sets of chips were obtained using a Venn online tool and imported into the Database for Annotation, Visualization, and Integrated Discovery (DAVID) database for Gene Ontology (GO) enrichment and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analyses. The protein-protein interaction (PPI) network was constructed by STRING online software for further study, and the core genes were determined by Cytoscape software and KEGG pathway enrichment analysis. The clustering heat map was drawn by Excel software to verify its accuracy. In addition, we used the University of Alabama at Birmingham Cancer (UALCAN) website to analyze the expression of core genes in P53 mutation status, confirmed the expression of crucial core genes in lung cancer tissues with Gene Expression Profiling Interactive Analysis (GEPIA) and GEPIA2 online software, and evaluated their prognostic value in lung cancer patients with the Kaplan-Meier online plotter tool. Results CHEK1, CCNB1, CCNB2, and CDK1 were selected. The expression levels of these four genes in lung cancer tissues were significantly higher than those in normal tissues. Their increased expression was negatively correlated with lung cancer patients (including LUAD and LUSC) prognosis and survival rate. Conclusion CHEK1, CCNB1, CCNB2, and CDK1 are the critical core genes of lung cancer and are highly expressed in lung cancer. They are negatively correlated with the prognosis of lung cancer patients (including LUAD and LUSC) and closely related to the formation and prediction of lung cancer. They are valuable predictors and may be predictive biomarkers of lung cancer.
Collapse
|
28
|
Bioinformatics Prediction and Experimental Validation Identify a Novel Cuproptosis-Related Gene Signature in Human Synovial Inflammation during Osteoarthritis Progression. Biomolecules 2023; 13:biom13010127. [PMID: 36671512 PMCID: PMC9855951 DOI: 10.3390/biom13010127] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 11/27/2022] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
Osteoarthritis (OA) is the one of most common joint diseases worldwide. Cuproptosis, which had been discovered lately, is a novel form of cell death induced by copper. Our purpose is to study the relationship between cuproptosis-related genes (CRGs) and inflammatory microenvironments in patients with OA and identify characteristic cuproptosis-related biomarkers. First, the combinatory analysis of OA transcriptome data from five datasets identified differentially expressed CRGs associated with OA. Then, we applied single-sample gene set enrichment analysis (ssGSEA) to evaluate immune-cell infiltration and immune-function levels in OA patients and normal controls, respectively. Hub CRGs for OA were mined based on the random forest (RF) model, and a nomogram prediction model was constructed based on them. In total, four differentially expressed CRGs were identified through bioinformatics analysis and confirmed by RT-qPCR. FDX1 and LIPT1 were expressed at a high level in OA, while DBT and DLST were expressed higher in the normal group. In total, 10 CRGs were found to be significantly correlated with immune landscape. Four hub CRGs were subsequently obtained by the RF analysis as potential biomarkers for OA. We constructed an OA predictive model based on these four CRGs (DBT, DLST, FDX1, and LIPT1).
Collapse
|
29
|
Patel SK, Surve J, Parmar J, Ahmed K, Bui FM, Al-Zahrani FA. Recent Advances in Biosensors for Detection of COVID-19 and Other Viruses. IEEE Rev Biomed Eng 2023; 16:22-37. [PMID: 36197867 PMCID: PMC10009816 DOI: 10.1109/rbme.2022.3212038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 06/28/2022] [Accepted: 09/23/2022] [Indexed: 11/06/2022]
Abstract
This century has introduced very deadly, dangerous, and infectious diseases to humankind such as the influenza virus, Ebola virus, Zika virus, and the most infectious SARS-CoV-2 commonly known as COVID-19 and have caused epidemics and pandemics across the globe. For some of these diseases, proper medications, and vaccinations are missing and the early detection of these viruses will be critical to saving the patients. And even the vaccines are available for COVID-19, the new variants of COVID-19 such as Delta, and Omicron are spreading at large. The available virus detection techniques take a long time, are costly, and complex and some of them generates false negative or false positive that might cost patients their lives. The biosensor technique is one of the best qualified to address this difficult challenge. In this systematic review, we have summarized recent advancements in biosensor-based detection of these pandemic viruses including COVID-19. Biosensors are emerging as efficient and economical analytical diagnostic instruments for early-stage illness detection. They are highly suitable for applications related to healthcare, wearable electronics, safety, environment, military, and agriculture. We strongly believe that these insights will aid in the study and development of a new generation of adaptable virus biosensors for fellow researchers.
Collapse
Affiliation(s)
- Shobhit K. Patel
- Department of Computer EngineeringMarwadi UniversityRajkot360003India
| | - Jaymit Surve
- Department of Electrical EngineeringMarwadi UniversityRajkot360003India
| | - Juveriya Parmar
- Department of Mechanical and Materials EngineeringUniversity of Nebraska - LincolnNebraska68588USA
- Department of Electronics and Communication EngineeringMarwadi UniversityRajkot360003India
| | - Kawsar Ahmed
- Department of Electrical and Computer EngineeringUniversity of SaskatchewanSaskatoonSKS79 5A9Canada
- Group of Bio-PhotomatiX, Department of Information and Communication TechnologyMawlana Bhashani Science and Technology UniversitySantoshTangail1902Bangladesh
| | - Francis M. Bui
- Department of Electrical and Computer EngineeringUniversity of SaskatchewanSaskatoonSKS79 5A9Canada
| | | |
Collapse
|
30
|
Snigdha M, Akter A, Amin MA, Islam MZ. Bioinformatics approach to analyse COVID-19 biomarkers accountable for generation of intracranial aneurysm in COVID-19 patients. INFORMATICS IN MEDICINE UNLOCKED 2023; 39:101247. [PMID: 37159621 PMCID: PMC10141791 DOI: 10.1016/j.imu.2023.101247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 04/17/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
COVID-19 became a health emergency on January 30, 2020. SARS-CoV-2 is the causative agent of the coronavirus disease known as COVID-19 and can develop cardiometabolic and neurological disorders. Intracranial aneurysm (IA) is considered the most significant reason for hemorrhagic stroke,and it accounts for approximately 85% of all subarachnoid hemorrhages (SAH). Retinoid signaling abnormalities may explain COVID-19's pathogenesis with inhibition of AEH2, from which COVID-19 infection may enhance aneurysm formation and rupture due to abrupt blood pressure changes, endothelial cell injury, and systemic inflammation. The objective of this study was to investigate the potential biomarkers, differentially expressed genes (DEGs), and metabolic pathways associated with both COVID-19 and intracranial aneurysm (IA) using simulation databases like DIsGeNET. The purpose was to confirm prior findings and gain a comprehensive understanding of the underlying mechanisms that contribute to the development of these conditions. We combined the regulated genes to describe intracranial aneurysm formation in COVID-19. To determine DEGs in COVID-19 and IA patient tissues, we compared gene expression transcriptomic datasets from healthy and diseased individuals. There were 41 differentially expressed genes (DEGs) shared by both the COVID-19 and IA datasets (27 up-regulated genes and 14 down-regulated genes). Using protein-protein interaction analysis, we were able to identify hub proteins (C3, NCR1, IL10RA, OXTR, RSAD2, CD38, IL10RB, MX1, IL10, GFAP, IFIT3, XAF1, USP18, OASL, IFI6, EPSTI1, CMPK2, and ISG15), which were not described as key proteins for both COVID-19 and IA before. We also used Gene Ontology analysis (6 significant ontologies were validated), Pathway analysis (the top 20 were validated), TF-Gene interaction analysis, Gene miRNA analysis, and Drug-Protein interaction analysis methods to comprehend the extensive connection between COVID-19 and IA. In Drug-Protein interaction analysis, we have gotten the following three drugs: LLL-3348, CRx139, and AV41 against IL10 which was both common for COVID-19 and IA disease. Our study with different cabalistic methods has showed the interaction between the proteins and pathways with drug analysis which may direct further treatment development for certain diseases.
Collapse
Affiliation(s)
- Mahajabin Snigdha
- Department of Pharmacy, Islamic University, Kushtia, 7003, Bangladesh
| | - Azifa Akter
- Department of Pharmacy, Islamic University, Kushtia, 7003, Bangladesh
| | - Md Al Amin
- Department of Computer Science & Engineering, Prime University, Dhaka, 1216, Bangladesh
| | - Md Zahidul Islam
- Department of Information & Communication Technology, Islamic University, Kushtia, 7003, Bangladesh
| |
Collapse
|
31
|
Bu F, Guan R, Wang W, Liu Z, Yin S, Zhao Y, Chai J. Bioinformatics and systems biology approaches to identify the effects of COVID-19 on neurodegenerative diseases: A review. Medicine (Baltimore) 2022; 101:e32100. [PMID: 36626425 PMCID: PMC9750669 DOI: 10.1097/md.0000000000032100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), causing coronavirus disease (COVID-19), has been devastated by COVID-19 in an increasing number of countries and health care systems around the world since its announcement of a global pandemic on 11 March 2020. During the pandemic, emerging novel viral mutant variants have caused multiple outbreaks of COVID-19 around the world and are prone to genetic evolution, causing serious damage to human health. As confirmed cases of COVID-19 spread rapidly, there is evidence that SARS-CoV-2 infection involves the central nervous system (CNS) and peripheral nervous system (PNS), directly or indirectly damaging neurons and further leading to neurodegenerative diseases (ND), but the molecular mechanisms of ND and CVOID-19 are unknown. We employed transcriptomic profiling to detect several major diseases of ND: Alzheimer 's disease (AD), Parkinson' s disease (PD), and multiple sclerosis (MS) common pathways and molecular biomarkers in association with COVID-19, helping to understand the link between ND and COVID-19. There were 14, 30 and 19 differentially expressed genes (DEGs) between COVID-19 and Alzheimer 's disease (AD), Parkinson' s disease (PD) and multiple sclerosis (MS), respectively; enrichment analysis showed that MAPK, IL-17, PI3K-Akt and other signaling pathways were significantly expressed; the hub genes (HGs) of DEGs between ND and COVID-19 were CRH, SST, TAC1, SLC32A1, GAD2, GAD1, VIP and SYP. Analysis of transcriptome data suggests multiple co-morbid mechanisms between COVID-19 and AD, PD, and MS, providing new ideas and therapeutic strategies for clinical prevention and treatment of COVID-19 and ND.
Collapse
Affiliation(s)
- Fan Bu
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
- * Correspondence: Fan Bu, Heilongjiang University of Chinese Medicine, Haerbin 150040, Heilongjiang Province, China (e-mail: )
| | - Ruiqian Guan
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
- Heilongjiang University of Chinese Medicine Affiliated Second Hospital, Haerbin, Heilongjiang Province, China
| | - Wanyu Wang
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| | - Zhao Liu
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| | - Shijie Yin
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| | - Yonghou Zhao
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
- Heilongjiang University of Chinese Medicine Affiliated Second Hospital, Haerbin, Heilongjiang Province, China
| | - Jianbo Chai
- Heilongjiang University of Chinese Medicine, Haerbin, Heilongjiang Province, China
| |
Collapse
|
32
|
The identification and validation of hub genes associated with advanced IPF by weighted gene co-expression network analysis. Funct Integr Genomics 2022; 22:1127-1138. [PMID: 36107393 DOI: 10.1007/s10142-022-00901-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 09/08/2022] [Accepted: 09/08/2022] [Indexed: 01/18/2023]
|
33
|
Jiang ST, Liu YG, Zhang L, Sang XT, Xu YY, Lu X. Systems biology approach reveals a common molecular basis for COVID-19 and non-alcoholic fatty liver disease (NAFLD). Eur J Med Res 2022; 27:251. [PMCID: PMC9664052 DOI: 10.1186/s40001-022-00865-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 10/24/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Patients with non-alcoholic fatty liver disease (NAFLD) may be more susceptible to coronavirus disease 2019 (COVID-19) and even more likely to suffer from severe COVID-19. Whether there is a common molecular pathological basis for COVID-19 and NAFLD remains to be identified. The present study aimed to elucidate the transcriptional alterations shared by COVID-19 and NAFLD and to identify potential compounds targeting both diseases.
Methods
Differentially expressed genes (DEGs) for COVID-19 and NAFLD were extracted from the GSE147507 and GSE89632 datasets, and common DEGs were identified using the Venn diagram. Subsequently, we constructed a protein–protein interaction (PPI) network based on the common DEGs and extracted hub genes. Then, we performed gene ontology (GO) and pathway analysis of common DEGs. In addition, transcription factors (TFs) and miRNAs regulatory networks were constructed, and drug candidates were identified.
Results
We identified a total of 62 common DEGs for COVID-19 and NAFLD. The 10 hub genes extracted based on the PPI network were IL6, IL1B, PTGS2, JUN, FOS, ATF3, SOCS3, CSF3, NFKB2, and HBEGF. In addition, we also constructed TFs–DEGs, miRNAs–DEGs, and protein–drug interaction networks, demonstrating the complex regulatory relationships of common DEGs.
Conclusion
We successfully extracted 10 hub genes that could be used as novel therapeutic targets for COVID-19 and NAFLD. In addition, based on common DEGs, we propose some potential drugs that may benefit patients with COVID-19 and NAFLD.
Collapse
|
34
|
Yan C, Niu Y, Wang X. Blood transcriptome analysis revealed the crosstalk between COVID-19 and HIV. Front Immunol 2022; 13:1008653. [PMID: 36389792 PMCID: PMC9650272 DOI: 10.3389/fimmu.2022.1008653] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Accepted: 09/29/2022] [Indexed: 08/08/2023] Open
Abstract
BACKGROUND The severe coronavirus disease 2019 (COVID-19) is an infectious disease caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which has resulted in the most devastating pandemic in modern history. Human immunodeficiency virus (HIV) destroys immune system cells and weakens the body's ability to resist daily infections and diseases. Furthermore, HIV-infected individuals had double COVID-19 mortality risk and experienced worse COVID-related outcomes. However, the existing research still lacks the understanding of the molecular mechanism underlying crosstalk between COVID-19 and HIV. The aim of our work was to illustrate blood transcriptome crosstalk between COVID-19 and HIV and to provide potential drugs that might be useful for the treatment of HIV-infected COVID-19 patients. METHODS COVID-19 datasets (GSE171110 and GSE152418) were downloaded from Gene Expression Omnibus (GEO) database, including 54 whole-blood samples and 33 peripheral blood mononuclear cells samples, respectively. HIV dataset (GSE37250) was also obtained from GEO database, containing 537 whole-blood samples. Next, the "Deseq2" package was used to identify differentially expressed genes (DEGs) between COVID-19 datasets (GSE171110 and GSE152418) and the "limma" package was utilized to identify DEGs between HIV dataset (GSE37250). By intersecting these two DEG sets, we generated common DEGs for further analysis, containing Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and Gene Ontology (GO) functional enrichment analysis, protein-protein interaction (PPI) analysis, transcription factor (TF) candidate identification, microRNAs (miRNAs) candidate identification and drug candidate identification. RESULTS In this study, a total of 3213 DEGs were identified from the merged COVID-19 dataset (GSE171110 and GSE152418), and 1718 DEGs were obtained from GSE37250 dataset. Then, we identified 394 common DEGs from the intersection of the DEGs in COVID-19 and HIV datasets. GO and KEGG enrichment analysis indicated that common DEGs were mainly gathered in chromosome-related and cell cycle-related signal pathways. Top ten hub genes (CCNA2, CCNB1, CDC20, TOP2A, AURKB, PLK1, BUB1B, KIF11, DLGAP5, RRM2) were ranked according to their scores, which were screened out using degree algorithm on the basis of common DEGs. Moreover, top ten drug candidates (LUCANTHONE, Dasatinib, etoposide, Enterolactone, troglitazone, testosterone, estradiol, calcitriol, resveratrol, tetradioxin) ranked by their P values were screened out, which maybe be beneficial for the treatment of HIV-infected COVID-19 patients. CONCLUSION In this study, we provide potential molecular targets, signaling pathways, small molecular compounds, and promising biomarkers that contribute to worse COVID-19 prognosis in patients with HIV, which might contribute to precise diagnosis and treatment for HIV-infected COVID-19 patients.
Collapse
Affiliation(s)
- Cheng Yan
- *Correspondence: Cheng Yan, ; Xuannian Wang,
| | | | | |
Collapse
|
35
|
Lu L, Qin J, Chen J, Yu N, Miyano S, Deng Z, Li C. Recent computational drug repositioning strategies against SARS-CoV-2. Comput Struct Biotechnol J 2022; 20:5713-5728. [PMID: 36277237 PMCID: PMC9575573 DOI: 10.1016/j.csbj.2022.10.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 10/12/2022] [Accepted: 10/12/2022] [Indexed: 11/08/2022] Open
Abstract
We performed a comprehensive review of computational drug repositioning methods applied to COVID-19 based on differing data types including sequence data, expression data, structure data and interaction data. We found that graph theory and neural network were the most used strategies for drug repositioning in the case of COVID-19. Integrating different levels of data may improve the success rate for drug repositioning.
Since COVID-19 emerged in 2019, significant levels of suffering and disruption have been caused on a global scale. Although vaccines have become widely used, the virus has shown its potential for evading immunities or acquiring other novel characteristics. Whether current drug treatments are still effective for people infected with Omicron remains unclear. Due to the long development cycles and high expense requirements of de novo drug development, many researchers have turned to consider drug repositioning in the search to find effective treatments for COVID-19. Here, we review such drug repositioning and combination efforts towards providing better handling. For potential drugs under consideration, aspects of both structure and function require attention, with specific categories of sequence, expression, structure, and interaction, the key parameters for investigation. For different data types, we show the corresponding differing drug repositioning methods that have been exploited. As incorporating drug combinations can increase therapeutic efficacy and reduce toxicity, we also review computational strategies to reveal drug combination potential. Taken together, we found that graph theory and neural network were the most used strategy with high potential towards drug repositioning for COVID-19. Integrating different levels of data may further improve the success rate of drug repositioning.
Collapse
Affiliation(s)
- Lu Lu
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiale Qin
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Hangzhou, China
| | - Jiandong Chen
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,School of Public Health, Undergraduate School of Zhejiang University, Hangzhou, China
| | - Na Yu
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Satoru Miyano
- M&D Data Science Center, Tokyo Medical and Dental University, Tokyo, Japan
| | - Zhenzhong Deng
- Xinhua Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, China,Corresponding authors at: Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (C. Li).
| | - Chen Li
- Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China,Zhejiang Provincial Key Laboratory of Genetic & Developmental Disorders, Zhejiang University School of Medicine, Hangzhou, China,Alibaba-Zhejiang University Joint Research Center of Future Digital Healthcare, Hangzhou, China,Corresponding authors at: Department of Human Genetics, Department of Ultrasound, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China (C. Li).
| |
Collapse
|
36
|
Fang H, Sun Z, Chen Z, Chen A, Sun D, Kong Y, Fang H, Qian G. Bioinformatics and systems-biology analysis to determine the effects of Coronavirus disease 2019 on patients with allergic asthma. Front Immunol 2022; 13:988479. [PMID: 36211429 PMCID: PMC9537444 DOI: 10.3389/fimmu.2022.988479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/30/2022] [Indexed: 12/05/2022] Open
Abstract
Background The coronavirus disease (COVID-19) pandemic has posed a significant challenge for global health systems. Increasing evidence shows that asthma phenotypes and comorbidities are major risk factors for COVID-19 symptom severity. However, the molecular mechanisms underlying the association between COVID-19 and asthma are poorly understood. Therefore, we conducted bioinformatics and systems biology analysis to identify common pathways and molecular biomarkers in patients with COVID-19 and asthma, as well as potential molecular mechanisms and candidate drugs for treating patients with both COVID-19 and asthma. Methods Two sets of differentially expressed genes (DEGs) from the GSE171110 and GSE143192 datasets were intersected to identify common hub genes, shared pathways, and candidate drugs. In addition, murine models were utilized to explore the expression levels and associations of the hub genes in asthma and lung inflammation/injury. Results We discovered 157 common DEGs between the asthma and COVID-19 datasets. A protein–protein-interaction network was built using various combinatorial statistical approaches and bioinformatics tools, which revealed several hub genes and critical modules. Six of the hub genes were markedly elevated in murine asthmatic lungs and were positively associated with IL-5, IL-13 and MUC5AC, which are the key mediators of allergic asthma. Gene Ontology and pathway analysis revealed common associations between asthma and COVID-19 progression. Finally, we identified transcription factor–gene interactions, DEG–microRNA coregulatory networks, and potential drug and chemical-compound interactions using the hub genes. Conclusion We identified the top 15 hub genes that can be used as novel biomarkers of COVID-19 and asthma and discovered several promising candidate drugs that might be helpful for treating patients with COVID-19 and asthma.
Collapse
Affiliation(s)
- Hongwei Fang
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhun Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Zhouyi Chen
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Anning Chen
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Donglin Sun
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yan Kong
- Department of Anesthesiology (High-Tech Branch), The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Hao Fang
- Department of Anesthesiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Anesthesiology, Minhang Hospital, Fudan University, Shanghai, China
- *Correspondence: Guojun Qian, ; Hao Fang,
| | - Guojun Qian
- Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
- *Correspondence: Guojun Qian, ; Hao Fang,
| |
Collapse
|
37
|
Lv Y, Zhang T, Cai J, Huang C, Zhan S, Liu J. Bioinformatics and systems biology approach to identify the pathogenetic link of Long COVID and Myalgic Encephalomyelitis/Chronic Fatigue Syndrome. Front Immunol 2022; 13:952987. [PMID: 36189286 PMCID: PMC9524193 DOI: 10.3389/fimmu.2022.952987] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Accepted: 08/18/2022] [Indexed: 11/13/2022] Open
Abstract
Background The COVID-19 pandemic, caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), is a global crisis. Although many people recover from COVID-19 infection, they are likely to develop persistent symptoms similar to those of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) after discharge. Those constellations of symptoms persist for months after infection, called Long COVID, which may lead to considerable financial burden and healthcare challenges. However, the mechanisms underlying Long COVID and ME/CFS remain unclear. Methods We collected the genes associated with Long COVID and ME/CFS in databases by restricted screening conditions and clinical sample datasets with limited filters. The common genes for Long COVID and ME/CFS were finally obtained by taking the intersection. We performed several advanced bioinformatics analyses based on common genes, including gene ontology and pathway enrichment analyses, protein-protein interaction (PPI) analysis, transcription factor (TF)-gene interaction network analysis, transcription factor-miRNA co-regulatory network analysis, and candidate drug analysis prediction. Results We found nine common genes between Long COVID and ME/CFS and gained a piece of detailed information on their biological functions and signaling pathways through enrichment analysis. Five hub proteins (IL-6, IL-1B, CD8A, TP53, and CXCL8) were collected by the PPI network. The TF-gene and TF-miRNA coregulatory networks were demonstrated by NetworkAnalyst. In the end, 10 potential chemical compounds were predicted. Conclusion This study revealed common gene interaction networks of Long COVID and ME/CFS and predicted potential therapeutic drugs for clinical practice. Our findings help to identify the potential biological mechanism between Long COVID and ME/CFS. However, more laboratory and multicenter evidence is required to explore greater mechanistic insight before clinical application in the future.
Collapse
Affiliation(s)
- Yongbiao Lv
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Tian Zhang
- The First Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Junxiang Cai
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Chushuan Huang
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Shaofeng Zhan
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianbo Liu
- The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, China
| |
Collapse
|
38
|
Network-Based Data Analysis Reveals Ion Channel-Related Gene Features in COVID-19: A Bioinformatic Approach. Biochem Genet 2022; 61:471-505. [PMID: 36104591 PMCID: PMC9473477 DOI: 10.1007/s10528-022-10280-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 09/01/2022] [Indexed: 11/02/2022]
Abstract
Coronavirus disease 2019 (COVID-19) seriously threatens human health and has been disseminated worldwide. Although there are several treatments for COVID-19, its control is currently suboptimal. Therefore, the development of novel strategies to treat COVID-19 is necessary. Ion channels are located on the membranes of all excitable cells and many intracellular organelles and are key components involved in various biological processes. They are a target of interest when searching for drug targets. This study aimed to reveal the relevant molecular features of ion channel genes in COVID-19 based on bioinformatic analyses. The RNA-sequencing data of patients with COVID-19 and healthy subjects (GSE152418 and GSE171110 datasets) were obtained from the Gene Expression Omnibus (GEO) database. Ion channel genes were selected from the Hugo Gene Nomenclature Committee (HGNC) database. The RStudio software was used to process the data based on the corresponding R language package to identify ion channel-associated differentially expressed genes (DEGs). Based on the DEGs, Gene Ontology (GO) functional and pathway enrichment analyses were performed using the Enrichr web tool. The STRING database was used to generate a protein-protein interaction (PPI) network, and the Cytoscape software was used to screen for hub genes in the PPI network based on the cytoHubba plug-in. Transcription factors (TF)-DEG, DEG-microRNA (miRNA) and DEG-disease association networks were constructed using the NetworkAnalyst web tool. Finally, the screened hub genes as drug targets were subjected to enrichment analysis based on the DSigDB using the Enrichr web tool to identify potential therapeutic agents for COVID-19. A total of 29 ion channel-associated DEGs were identified. GO functional analysis showed that the DEGs were integral components of the plasma membrane and were mainly involved in inorganic cation transmembrane transport and ion channel activity functions. Pathway analysis showed that the DEGs were mainly involved in nicotine addiction, calcium regulation in the cardiac cell and neuronal system pathways. The top 10 hub genes screened based on the PPI network included KCNA2, KCNJ4, CACNA1A, CACNA1E, NALCN, KCNA5, CACNA2D1, TRPC1, TRPM3 and KCNN3. The TF-DEG and DEG-miRNA networks revealed significant TFs (FOXC1, GATA2, HINFP, USF2, JUN and NFKB1) and miRNAs (hsa-mir-146a-5p, hsa-mir-27a-3p, hsa-mir-335-5p, hsa-let-7b-5p and hsa-mir-129-2-3p). Gene-disease association network analysis revealed that the DEGs were closely associated with intellectual disability and cerebellar ataxia. Drug-target enrichment analysis showed that the relevant drugs targeting the hub genes CACNA2D1, CACNA1A, CACNA1E, KCNA2 and KCNA5 were gabapentin, gabapentin enacarbil, pregabalin, guanidine hydrochloride and 4-aminopyridine. The results of this study provide a valuable basis for exploring the mechanisms of ion channel genes in COVID-19 and clues for developing therapeutic strategies for COVID-19.
Collapse
|
39
|
Lu L, Liu LP, Gui R, Dong H, Su YR, Zhou XH, Liu FX. Discovering common pathogenetic processes between COVID-19 and sepsis by bioinformatics and system biology approach. Front Immunol 2022; 13:975848. [PMID: 36119022 PMCID: PMC9471316 DOI: 10.3389/fimmu.2022.975848] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Accepted: 08/04/2022] [Indexed: 11/17/2022] Open
Abstract
Corona Virus Disease 2019 (COVID-19), an acute respiratory infectious disease caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), has spread rapidly worldwide, resulting in a pandemic with a high mortality rate. In clinical practice, we have noted that many critically ill or critically ill patients with COVID-19 present with typical sepsis-related clinical manifestations, including multiple organ dysfunction syndrome, coagulopathy, and septic shock. In addition, it has been demonstrated that severe COVID-19 has some pathological similarities with sepsis, such as cytokine storm, hypercoagulable state after blood balance is disrupted and neutrophil dysfunction. Considering the parallels between COVID-19 and non-SARS-CoV-2 induced sepsis (hereafter referred to as sepsis), the aim of this study was to analyze the underlying molecular mechanisms between these two diseases by bioinformatics and a systems biology approach, providing new insights into the pathogenesis of COVID-19 and the development of new treatments. Specifically, the gene expression profiles of COVID-19 and sepsis patients were obtained from the Gene Expression Omnibus (GEO) database and compared to extract common differentially expressed genes (DEGs). Subsequently, common DEGs were used to investigate the genetic links between COVID-19 and sepsis. Based on enrichment analysis of common DEGs, many pathways closely related to inflammatory response were observed, such as Cytokine-cytokine receptor interaction pathway and NF-kappa B signaling pathway. In addition, protein-protein interaction networks and gene regulatory networks of common DEGs were constructed, and the analysis results showed that ITGAM may be a potential key biomarker base on regulatory analysis. Furthermore, a disease diagnostic model and risk prediction nomogram for COVID-19 were constructed using machine learning methods. Finally, potential therapeutic agents, including progesterone and emetine, were screened through drug-protein interaction networks and molecular docking simulations. We hope to provide new strategies for future research and treatment related to COVID-19 by elucidating the pathogenesis and genetic mechanisms between COVID-19 and sepsis.
Collapse
Affiliation(s)
- Lu Lu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Le-Ping Liu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
- Department of Pediatrics, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Hang Dong
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Yan-Rong Su
- Department of Laboratory Medicine, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Xiong-Hui Zhou
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
| | - Feng-Xia Liu
- Department of Blood Transfusion, The Third Xiangya Hospital of Central South University, Changsha, China
- *Correspondence: Feng-Xia Liu,
| |
Collapse
|
40
|
Li XY, Wang JB, An HB, Wen MZ, You JX, Yang XT. Effect of SARS-CoV-2 infection on asthma patients. Front Med (Lausanne) 2022; 9:928637. [PMID: 35983093 PMCID: PMC9378965 DOI: 10.3389/fmed.2022.928637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
BackgroundSARS-CoV-2 causes coronavirus disease 2019 (COVID-19), a new coronavirus pneumonia, and containing such an international pandemic catastrophe remains exceedingly difficult. Asthma is a severe chronic inflammatory airway disease that is becoming more common around the world. However, the link between asthma and COVID-19 remains unknown. Through bioinformatics analysis, this study attempted to understand the molecular pathways and discover potential medicines for treating COVID-19 and asthma.MethodsTo investigate the relationship between SARS-CoV-2 and asthma patients, a transcriptome analysis was used to discover shared pathways and molecular signatures in asthma and COVID-19. Here, two RNA-seq data (GSE147507 and GSE74986) from the Gene Expression Omnibus were used to detect differentially expressed genes (DEGs) in asthma and COVID-19 patients to find the shared pathways and the potential drug candidates.ResultsThere were 66 DEGs in all that were classified as common DEGs. Using a protein-protein interaction (PPI) network created using various bioinformatics techniques, five hub genes were found. We found that asthma has some shared links with the progression of COVID-19. Additionally, protein-drug interactions with common DEGs were also identified in the datasets.ConclusionWe investigated possible links between COVID-19 and asthma using bioinformatics databases, which might be useful in treating COVID-19 patients. More studies on populations affected by these diseases are needed to elucidate the molecular mechanism behind their association.
Collapse
Affiliation(s)
- Xin-yu Li
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- Department of Neurosurgery, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jing-bing Wang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Hong-bang An
- Affiliated Hospital of Weifang Medical University, School of Clinical Medicine, Weifang Medical University, Weifang, China
| | - Ming-zhe Wen
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-xiong You
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
| | - Xi-tao Yang
- Department of Interventional Therapy, Multidisciplinary Team of Vascular Anomalies, Shanghai Ninth People’s Hospital, Shanghai Jiao Tong University, Shanghai, China
- *Correspondence: Xi-tao Yang,
| |
Collapse
|
41
|
Huang J, Wang Y, Zha Y, Zeng X, Li W, Zhou M. Transcriptome Analysis Reveals Hub Genes Regulating Autophagy in Patients With Severe COVID-19. Front Genet 2022; 13:908826. [PMID: 35923698 PMCID: PMC9340158 DOI: 10.3389/fgene.2022.908826] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/06/2022] [Indexed: 11/21/2022] Open
Abstract
Background: The COVID-19 pandemic has currently developed into a worldwide threat to humankind. Importantly, patients with severe COVID-19 are believed to have a higher mortality risk than those with mild conditions. However, despite the urgent need to develop novel therapeutic strategies, the biological features and pathogenic mechanisms of severe COVID-19 are poorly understood. Methods: Here, peripheral blood mononuclear cells (PBMCs) from four patients with severe COVID-19, four patients with mild COVID-19, and four healthy controls were examined by RNA sequencing (RNA-Seq). We conducted gene expression analysis and Venn diagrams to detect specific differentially expressed genes (DEGs) in patients with severe disease compared with those with mild conditions. Gene Ontology (GO) enrichment analysis was performed to identify the significant biological processes, and protein–protein interaction networks were constructed to extract hub genes. These hub genes were then subjected to regulatory signatures and protein–chemical interaction analysis for certain regulatory checkpoints and identification of potent chemical agents. Finally, to demonstrate the cell type-specific expression of these genes, we performed single-cell RNA-Seq analyses using an online platform. Results: A total of 144 DEGs were specifically expressed in severe COVID-19, and GO enrichment analysis revealed a significant association of these specific DEGs with autophagy. Hub genes such as MVB12A, CHMP6, STAM, and VPS37B were then found to be most significantly involved in the biological processes of autophagy at the transcriptome level. In addition, six transcription factors, including SRF, YY1, CREB1, PPARG, NFIC, and GATA2, as well as miRNAs, namely, hsa-mir-1-3p, and potent chemical agents such as copper sulfate and cobalt chloride, may cooperate in regulating the autophagy hub genes. Furthermore, classical monocytes may play a central role in severe COVID-19. Conclusion: We suggest that autophagy plays a crucial role in severe COVID-19. This study might facilitate a more profound knowledge of the biological characteristics and progression of COVID-19 and the development of novel therapeutic approaches to achieve a breakthrough in the current COVID-19 pandemic.
Collapse
Affiliation(s)
- Jinfeng Huang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China
| | - Yimeng Wang
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Yawen Zha
- Department of Radiation Oncology Ⅱ, Zhongshan People’s Hospital, Zhongshan, China
| | - Xin Zeng
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenxing Li
- Department of Biochemistry and Molecular Biology, School of Basic Medicine, Southern Medical University, Guangzhou, China
| | - Meijuan Zhou
- Department of Radiation Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, China
- Jiangmen Central Hospital, Affiliated Jiangmen Hospital of Sun Yat-sen University, Jiangmen, China
- *Correspondence: Meijuan Zhou,
| |
Collapse
|
42
|
Al-Mustanjid M, Mahmud SMH, Akter F, Rahman MS, Hossen MS, Rahman MH, Moni MA. Systems biology models to identify the influence of SARS-CoV-2 infections to the progression of human autoimmune diseases. INFORMATICS IN MEDICINE UNLOCKED 2022; 32:101003. [PMID: 35818398 PMCID: PMC9259025 DOI: 10.1016/j.imu.2022.101003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 06/25/2022] [Accepted: 06/25/2022] [Indexed: 11/20/2022] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has been circulating since 2019, and its global dominance is rising. Evidences suggest the respiratory illness SARS-CoV-2 has a sensitive affect on causing organ damage and other complications to the patients with autoimmune diseases (AD), posing a significant risk factor. The genetic interrelationships and molecular appearances between SARS-CoV-2 and AD are yet unknown. We carried out the transcriptomic analytical framework to delve into the SARS-CoV-2 impacts on AD progression. We analyzed both gene expression microarray and RNA-Seq datasets from SARS-CoV-2 and AD affected tissues. With neighborhood-based benchmarks and multilevel network topology, we obtained dysfunctional signaling and ontological pathways, gene disease (diseasesome) association network and protein-protein interaction network (PPIN), uncovered essential shared infection recurrence connectivities with biological insights underlying between SARS-CoV-2 and AD. We found a total of 77, 21, 9, 54 common DEGs for SARS-CoV-2 and inflammatory bowel disorder (IBD), SARS-CoV-2 and rheumatoid arthritis (RA), SARS-CoV-2 and systemic lupus erythematosus (SLE) and SARS-CoV-2 and type 1 diabetes (T1D). The enclosure of these common DEGs with bimolecular networks revealed 10 hub proteins (FYN, VEGFA, CTNNB1, KDR, STAT1, B2M, CD3G, ITGAV, TGFB3). Drugs such as amlodipine besylate, vorinostat, methylprednisolone, and disulfiram have been identified as a common ground between SARS-CoV-2 and AD from drug repurposing investigation which will stimulate the optimal selection of medications in the battle against this ongoing pandemic triggered by COVID-19.
Collapse
Affiliation(s)
- Md Al-Mustanjid
- Department of Software Engineering, Faculty of Science and Information Technology, Daffodil International University, Dhaka-1207, Bangladesh
| | - S M Hasan Mahmud
- Department of Computer Science, American International University-Bangladesh, Dhaka, 1229, Bangladesh
| | - Farzana Akter
- Department of Software Engineering, Faculty of Science and Information Technology, Daffodil International University, Dhaka-1207, Bangladesh
| | - Md Shazzadur Rahman
- Department of Computer Science & Engineering, Faculty of Science and Information Technology, Daffodil International University, Dhaka-1207, Bangladesh
| | - Md Sajid Hossen
- Department of Software Engineering, Faculty of Science and Information Technology, Daffodil International University, Dhaka-1207, Bangladesh
| | - Md Habibur Rahman
- Department of Computer Science and Engineering, Islamic University, Kushtia-7003, Bangladesh
| | - Mohammad Ali Moni
- Department of Computer Science and Engineering, Pabna Science & Technology University, Pabna, 6600, Bangladesh
| |
Collapse
|
43
|
Kang X, Wen X, Liang J, Liu L, Zhang Y, Wang Q, Zhao H. The Biological Interaction of SARS-CoV-2 Infection and Osteoporosis: A Preliminary Study. Front Cell Dev Biol 2022; 10:917907. [PMID: 35646907 PMCID: PMC9130749 DOI: 10.3389/fcell.2022.917907] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 04/27/2022] [Indexed: 11/13/2022] Open
Abstract
The COVID-19 pandemic caused by the severe acute coronavirus disease 2 (SARS-CoV-2) virus represents an ongoing threat to human health and well-being. Notably, many COVID-19 patients suffer from complications consistent with osteoporosis (OP) following disease resolution yet the mechanistic links between SARS-CoV-2 infection and OP remain to be clarified. The present study was thus developed to explore the potential basis for this link by employing transcriptomic analyses to identify signaling pathways and biomarkers associated with OP and SARS-CoV-2. Specifically, a previously published RNA-sequencing dataset (GSE152418) from Gene Expression Omnibus (GEO) was used to identify the differentially expressed genes (DEGs) in OP patients and individuals infected with SARS-CoV-2 as a means of exploring the underlying molecular mechanisms linking these two conditions. In total, 2,885 DEGs were identified by analyzing the COVID-19 patient dataset, with shared DEGs then being identified by comparison of these DEGs with those derived from an OP patient dataset. Hub genes were identified through a series of bioinformatics approaches and protein-protein interaction analyses. Predictive analyses of transcription factor/gene interactions, protein/drug interactions, and DEG/miRNA networks associated with these DEGs were also conducted. Together, these data highlight promising candidate drugs with the potential to treat both COVID-19 and OP.
Collapse
Affiliation(s)
- Xin Kang
- Department of Sports Medicine, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Xiaodong Wen
- Department of Foot and Ankle Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Jingqi Liang
- Department of Foot and Ankle Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Liang Liu
- Department of Foot and Ankle Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Yan Zhang
- Department of Foot and Ankle Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Qiong Wang
- Department of Foot and Ankle Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| | - Hongmou Zhao
- Department of Foot and Ankle Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
44
|
Hu H, Tang N, Zhang F, Li L, Li L. Bioinformatics and System Biology Approach to Identify the Influences of COVID-19 on Rheumatoid Arthritis. Front Immunol 2022; 13:860676. [PMID: 35464423 PMCID: PMC9021444 DOI: 10.3389/fimmu.2022.860676] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/16/2022] [Indexed: 02/05/2023] Open
Abstract
Background Severe coronavirus disease 2019 (COVID -19) has led to a rapid increase in mortality worldwide. Rheumatoid arthritis (RA) was a high-risk factor for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection, whereas the molecular mechanisms underlying RA and CVOID-19 are not well understood. The objectives of this study were to analyze potential molecular mechanisms and identify potential drugs for the treatment of COVID-19 and RA using bioinformatics and a systems biology approach. Methods Two Differentially expressed genes (DEGs) sets extracted from GSE171110 and GSE1775544 datasets were intersected to generate common DEGs, which were used for functional enrichment, pathway analysis, and candidate drugs analysis. Results A total of 103 common DEGs were identified in the two datasets between RA and COVID-19. A protein-protein interaction (PPI) was constructed using various combinatorial statistical methods and bioinformatics tools. Subsequently, hub genes and essential modules were identified from the PPI network. In addition, we performed functional analysis and pathway analysis under ontological conditions and found that there was common association between RA and progression of COVID-19 infection. Finally, transcription factor-gene interactions, protein-drug interactions, and DEGs-miRNAs coregulatory networks with common DEGs were also identified in the datasets. Conclusion We successfully identified the top 10 hub genes that could serve as novel targeted therapy for COVID-19 and screened out some potential drugs useful for COVID-19 patients with RA.
Collapse
Affiliation(s)
- Huan Hu
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China.,Clinical Medical College, Guizhou Medical University, Guiyang, China
| | - Nana Tang
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Facai Zhang
- Department of Urology/Institute of Urology, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Medical Intensive Care Unit, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Long Li
- Department of Rheumatology and Immunology, The Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
45
|
Bioinformatics and Network-based Approaches for Determining Pathways, Signature Molecules, and Drug Substances connected to Genetic Basis of Schizophrenia etiology. Brain Res 2022; 1785:147889. [PMID: 35339428 DOI: 10.1016/j.brainres.2022.147889] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 02/28/2022] [Accepted: 03/21/2022] [Indexed: 12/12/2022]
Abstract
Knowledge of heterogeneous etiology and pathophysiology of schizophrenia (SZP) is reasonably inadequate and non-deterministic due to its inherent complexity and underlying vast dynamics related to genetic mechanisms. The evolution of large-scale transcriptome-wide datasets and subsequent development of relevant, robust technologies for their analyses show promises toward elucidating the genetic basis of disease pathogenesis, its early risk prediction, and predicting drug molecule targets for therapeutic intervention. In this research, we have scrutinized the genetic basis of SZP through functional annotation and network-based system biology approaches. We have determined 96 overlapping differentially expressed genes (DEGs) from 2 microarray datasets and subsequently identified their interconnecting networks to reveal transcriptome signatures like hub proteins (FYN, RAD51, SOCS3, XIAP, AKAP13, PIK3C2A, CBX5, GATA3, EIF3K, and CDKN2B), transcription factors and miRNAs. In addition, we have employed gene set enrichment to highlight significant gene ontology (e.g., positive regulation of microglial cell activation) and relevant pathways (such as axon guidance and focal adhesion) interconnected to the genes associated with SZP. Finally, we have suggested candidate drug substances like Luteolin HL60 UP as a possible therapeutic target based on these key molecular signatures.
Collapse
|
46
|
Hasan MM, Khan Z, Chowdhury MS, Khan MA, Moni MA, Rahman MH. In silico molecular docking and ADME/T analysis of Quercetin compound with its evaluation of broad-spectrum therapeutic potential against particular diseases. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100894] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
|
47
|
Mahbub NI, Hasan MI, Rahman MH, Naznin F, Islam MZ, Moni MA. Identifying molecular signatures and pathways shared between Alzheimer's and Huntington's disorders: A bioinformatics and systems biology approach. INFORMATICS IN MEDICINE UNLOCKED 2022. [DOI: 10.1016/j.imu.2022.100888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022] Open
|
48
|
Kircher M, Chludzinski E, Krepel J, Saremi B, Beineke A, Jung K. Augmentation of Transcriptomic Data for Improved Classification of Patients with Respiratory Diseases of Viral Origin. Int J Mol Sci 2022; 23:ijms23052481. [PMID: 35269624 PMCID: PMC8910329 DOI: 10.3390/ijms23052481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 02/17/2022] [Accepted: 02/21/2022] [Indexed: 02/01/2023] Open
Abstract
To better understand the molecular basis of respiratory diseases of viral origin, high-throughput gene-expression data are frequently taken by means of DNA microarray or RNA-seq technology. Such data can also be useful to classify infected individuals by molecular signatures in the form of machine-learning models with genes as predictor variables. Early diagnosis of patients by molecular signatures could also contribute to better treatments. An approach that has rarely been considered for machine-learning models in the context of transcriptomics is data augmentation. For other data types it has been shown that augmentation can improve classification accuracy and prevent overfitting. Here, we compare three strategies for data augmentation of DNA microarray and RNA-seq data from two selected studies on respiratory diseases of viral origin. The first study involves samples of patients with either viral or bacterial origin of the respiratory disease, the second study involves patients with either SARS-CoV-2 or another respiratory virus as disease origin. Specifically, we reanalyze these public datasets to study whether patient classification by transcriptomic signatures can be improved when adding artificial data for training of the machine-learning models. Our comparison reveals that augmentation of transcriptomic data can improve the classification accuracy and that fewer genes are necessary as explanatory variables in the final models. We also report genes from our signatures that overlap with signatures presented in the original publications of our example data. Due to strict selection criteria, the molecular role of these genes in the context of respiratory infectious diseases is underlined.
Collapse
Affiliation(s)
- Magdalena Kircher
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Buenteweg 17p, 30559 Hannover, Germany; (M.K.); (J.K.); (B.S.)
| | - Elisa Chludzinski
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hannover, Germany; (E.C.); (A.B.)
| | - Jessica Krepel
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Buenteweg 17p, 30559 Hannover, Germany; (M.K.); (J.K.); (B.S.)
| | - Babak Saremi
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Buenteweg 17p, 30559 Hannover, Germany; (M.K.); (J.K.); (B.S.)
| | - Andreas Beineke
- Department of Pathology, University of Veterinary Medicine Hannover, Buenteweg 17, 30559 Hannover, Germany; (E.C.); (A.B.)
| | - Klaus Jung
- Institute for Animal Breeding and Genetics, University of Veterinary Medicine Hannover, Buenteweg 17p, 30559 Hannover, Germany; (M.K.); (J.K.); (B.S.)
- Correspondence: ; Tel.: +49-511-953-8878
| |
Collapse
|
49
|
Forni D, Cagliani R, Pontremoli C, Clerici M, Sironi M. The substitution spectra of coronavirus genomes. Brief Bioinform 2022; 23:bbab382. [PMID: 34518866 PMCID: PMC8499949 DOI: 10.1093/bib/bbab382] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/24/2021] [Accepted: 08/25/2021] [Indexed: 12/23/2022] Open
Abstract
The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has triggered an unprecedented international effort to sequence complete viral genomes. We leveraged this wealth of information to characterize the substitution spectrum of SARS-CoV-2 and to compare it with those of other human and animal coronaviruses. We show that, once nucleotide composition is taken into account, human and most animal coronaviruses display a mutation spectrum dominated by C to U and G to U substitutions, a feature that is not shared by other positive-sense RNA viruses. However, the proportions of C to U and G to U substitutions tend to decrease as divergence increases, suggesting that, whatever their origin, a proportion of these changes is subsequently eliminated by purifying selection. Analysis of the sequence context of C to U substitutions showed little evidence of apolipoprotein B mRNA editing catalytic polypeptide-like (APOBEC)-mediated editing and such contexts were similar for SARS-CoV-2 and Middle East respiratory syndrome coronavirus sampled from different hosts, despite different repertoires of APOBEC3 proteins in distinct species. Conversely, we found evidence that C to U and G to U changes affect CpG dinucleotides at a frequency higher than expected. Whereas this suggests ongoing selective reduction of CpGs, this effect alone cannot account for the substitution spectra. Finally, we show that, during the first months of SARS-CoV-2 pandemic spread, the frequency of both G to U and C to U substitutions increased. Our data suggest that the substitution spectrum of SARS-CoV-2 is determined by an interplay of factors, including intrinsic biases of the replication process, avoidance of CpG dinucleotides and other constraints exerted by the new host.
Collapse
Affiliation(s)
- Diego Forni
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| | - Rachele Cagliani
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| | - Chiara Pontremoli
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| | - Mario Clerici
- Department of Physiopathology and Transplantation, University of Milan, Milan, Italy
- Don C. Gnocchi Foundation ONLUS, IRCCS, Milan, Italy
| | - Manuela Sironi
- Scientific Institute IRCCS E. MEDEA, Bioinformatics, Bosisio Parini, Italy
| |
Collapse
|
50
|
Systems Biology and Bioinformatics approach to Identify blood based signatures molecules and drug targets of patient with COVID-19. INFORMATICS IN MEDICINE UNLOCKED 2022; 28:100840. [PMID: 34981034 PMCID: PMC8716147 DOI: 10.1016/j.imu.2021.100840] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) infection results in the development of a highly contagious respiratory ailment known as new coronavirus disease (COVID-19). Despite the fact that the prevalence of COVID-19 continues to rise, it is still unclear how people become infected with SARS-CoV-2 and how patients with COVID-19 become so unwell. Detecting biomarkers for COVID-19 using peripheral blood mononuclear cells (PBMCs) may aid in drug development and treatment. This research aimed to find blood cell transcripts that represent levels of gene expression associated with COVID-19 progression. Through the development of a bioinformatics pipeline, two RNA-Seq transcriptomic datasets and one microarray dataset were studied and discovered 102 significant differentially expressed genes (DEGs) that were shared by three datasets derived from PBMCs. To identify the roles of these DEGs, we discovered disease-gene association networks and signaling pathways, as well as we performed gene ontology (GO) studies and identified hub protein. Identified significant gene ontology and molecular pathways improved our understanding of the pathophysiology of COVID-19, and our identified blood-based hub proteins TPX2, DLGAP5, NCAPG, CCNB1, KIF11, HJURP, AURKB, BUB1B, TTK, and TOP2A could be used for the development of therapeutic intervention. In COVID-19 subjects, we discovered effective putative connections between pathological processes in the transcripts blood cells, suggesting that blood cells could be used to diagnose and monitor the disease’s initiation and progression as well as developing drug therapeutics.
Collapse
|