1
|
Zhou W, Li H, Zhang J, Liu C, Liu D, Chen X, Ouyang J, Zeng T, Peng S, Ouyang F, Long Y, Li Y. Identification and mechanism analysis of biomarkers related to butyrate metabolism in COVID-19 patients. Ann Med 2025; 57:2477301. [PMID: 40074706 PMCID: PMC11905318 DOI: 10.1080/07853890.2025.2477301] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Butyrate may inhibit SARS-CoV-2 replication and affect the development of COVID-19. However, there have been no systematic comprehensive analyses of the role of butyrate metabolism-related genes (BMRGs) in COVID-19. METHODS We performed differential expression analysis of BMRGs in the brain, liver and pancreas of COVID-19 patients and controls in GSE157852 and GSE151803. The differentially expressed genes (DEGs) and module genes between COVID-19 patients and healthy controls in GSE171110 were screened through 'limma' and 'WGANA' R package, respectively, followed by an intersection with BMRGs via 'ggvenn' R package. Six machine learning algorithms were employed to determine the best model for identifying biomarkers, and receiver operating characteristic (ROC) curves were plotted to evaluate the diagnostic value of the biomarkers in COVID-19. Moreover, the differences in immune-infiltrating cells between the COVID-19 and control groups were compared using CIBERSORT. The differences in immune cells and expression levels of biomarkers in immune cells among different tissues were analysed using GSE171668. RESULTS The BMRGs were the most different in the brain between the COVID-19 and control groups, including 21 upregulated and 16 downregulated genes. Five important common BMRGs were screened as biomarkers for COVID-19 using XGBoost, namely CCNB1, CCNA2, BRCA1, HBB and HSPA5, with increased diagnostic performance. Enrichment analysis revealed that these five genes were related to the cell cycle, cell proliferation and cell senescence. The infiltrating abundance of 12 immune cells was different between the COVID-19 and control groups. Finally, the expression levels of HSPA5, BRCA1 and HBB were higher in annotated cells than in CCNB1 and CCNA2, and there were four different types of immune cells in the liver, heart, lungs and kidneys. CONCLUSIONS These five genes may be potential biomarkers of butyrate metabolism in COVID-19 patients. These findings provide a direction for further studies on the molecular mechanisms underlying COVID-19.
Collapse
Affiliation(s)
- Wenchao Zhou
- Department of Assisted Reproductive Centre, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
- Department of Gynecology, The Second Affiliated Hospital, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Hui Li
- Department of Assisted Reproductive Centre, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Juan Zhang
- Department of Assisted Reproductive Centre, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Changsheng Liu
- Department of Scientific Research, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Dan Liu
- Department of Assisted Reproductive Centre, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Xupeng Chen
- Department of Gynecology, The Second Affiliated Hospital, Hunan Province Key Laboratory of Tumor Cellular & Molecular Pathology, Cancer Research Institute, Hengyang Medical School, University of South China, Hengyang, China
| | - Jing Ouyang
- Department of Infectious Disease, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Tian Zeng
- Department of Scientific Research, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Shuang Peng
- Department of Infectious Disease, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Fan Ouyang
- Department of Cardiology, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yunzhu Long
- Department of Infectious Disease, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| | - Yukun Li
- Department of Assisted Reproductive Centre, Xiangya Hospital Zhuzhou Central South University, Central South University, Zhuzhou, China
| |
Collapse
|
2
|
Nadendla EK, Alluri P, Sundaram B, Kumar SP, Chadchan SB, Sarkar R, Kanneganti TD. HCK regulates NLRP12-mediated PANoptosis. Proc Natl Acad Sci U S A 2025; 122:e2422079122. [PMID: 40408404 DOI: 10.1073/pnas.2422079122] [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: 10/24/2024] [Accepted: 04/16/2025] [Indexed: 05/25/2025] Open
Abstract
NOD-like receptors (NLRs) are a highly conserved family of cytosolic pattern recognition receptors that drive innate immune responses against pathogens, pathogen-associated molecular patterns, damage-associated molecular patterns, and homeostatic disruptions. Within the NLR family, NLRP12 was recently identified as a key regulator of PANoptosis, which is an innate immune, lytic cell death pathway initiated by innate immune sensors and driven by caspases and RIPKs through PANoptosome complexes. While NLRP12 activation is critical for maintaining homeostasis, aberrant activation has been implicated in a broad range of disorders, including cancers and metabolic, infectious, autoinflammatory, and hemolytic diseases. However, the molecular mechanisms of NLRP12 activation remain poorly understood. Here, we identified hematopoietic cell kinase (HCK) as a regulator of NLRP12-mediated PANoptosis. HCK expression was significantly upregulated in response to NLRP12-PANoptosome triggers. Moreover, Hck knockdown inhibited NLRP12-mediated PANoptosis. Computational analyses identified residues in the putative interaction interface between NLRP12 and HCK, suggesting that HCK likely binds NLRP12 in the region between its NACHT domain and pyrin domain (PYD); removal of the NLRP12 PYD abrogated this interaction in vitro. Overall, our work identifies HCK as a regulator of NLRP12-mediated PANoptosis, suggesting that it may serve as a potential therapeutic target for mitigating inflammation and pathology.
Collapse
Affiliation(s)
- Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Priyanshu Alluri
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Balamurugan Sundaram
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | | - Sangappa B Chadchan
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | - Roman Sarkar
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN 38105
| | | |
Collapse
|
3
|
Ma X, Huang T, Song Y, Pan H, Du A, Zhou X, Zeng Y, Yuan K. Bioinformatics and system biology approach to discover the common pathogenetic processes between COVID-19 and chronic hepatitis B. PLoS One 2025; 20:e0323708. [PMID: 40408617 DOI: 10.1371/journal.pone.0323708] [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: 02/22/2024] [Accepted: 04/12/2025] [Indexed: 05/25/2025] Open
Abstract
INTRODUCTION The ongoing coronavirus disease 2019 (COVID-19) pandemic, caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), presents a significant global public health threat. Concurrently, hepatitis B virus (HBV) remains a significant public health challenge. While previous studies have indicated an association between COVID-19 and chronic hepatitis B, the common underlying pathogenesis of these diseases remains incompletely understood. METHODS To investigate the shared molecular mechanisms between chronic HBV infection and COVID-19, a comprehensive investigation was conducted using bioinformatics and systems biology. Specifically, we utilized RNA-seq datasets (GSE196822 and GSE83148) to identify differentially expressed genes (DEGs) associated with both SARS-CoV-2 and HBV infection. Subsequently, these common DEGs were utilized to identify shared pathways, hub genes, transcriptional regulatory networks, and potential drugs. The differential expression of hub genes in both COVID-19 and HBV was verified using the GSE171110 and GSE94660 datasets, respectively. RESULTS From the 106 shared DEGs identified, immune-related pathways were found to play a role in the development and progression of chronic hepatitis B and COVID-19. Protein-protein interaction (PPI) network analysis revealed 8 hub genes: CDK1, E2F7, E2F8, TYMS, KIF20A, CENPE, TPX2, HMMR, CD8A, GZMA. In the validation set, the expression of hub genes was statistically significant in both the COVID-19 group and the HBV group compared with the healthy control group. Transcriptional regulatory network analysis identified 155 microRNAs (miRNAs) and 43 transcription factors (TFs) as potential regulatory signals. Notably, we identified potential therapeutic drugs for HBV chronic infection and COVID-19, including progesterone, estradiol, dasatinib, aspirin, etoposide, irinotecan hydrochloride, phorbol 12-myristate 13-acetate, lucanthone, calcitriol. CONCLUSION This research elucidates potential molecular targets, signaling pathways, and promising small molecule compounds that could aid in the treatment of chronic HBV infection and COVID-19.
Collapse
Affiliation(s)
- Xiao Ma
- 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
| | - 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
| | - 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
| | - 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
| | - Yong Zeng
- 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
| | - 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
|
4
|
Wu J, Gao P, Yang C, Zhuang F, Luo Y, Wen F, Zhang P, Wang L, Xie H, Dai C, Zhao D, Li C, Deng H, Deng Z, Chen C. Targeting mitochondrial complex I of CD177 + neutrophils alleviates lung ischemia-reperfusion injury. Cell Rep Med 2025; 6:102140. [PMID: 40398393 DOI: 10.1016/j.xcrm.2025.102140] [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: 06/20/2024] [Revised: 01/01/2025] [Accepted: 04/24/2025] [Indexed: 05/23/2025]
Abstract
Primary graft dysfunction (PGD) is the leading cause of early morbidity and mortality following lung transplantation, with neutrophils playing a central role in its inflammatory pathology. Here, we employ single-cell RNA sequencing and spatial transcriptomics to investigate neutrophil subtypes in the lung ischemia-reperfusion injury (IRI) model. We identify CD177+ neutrophils as an activated subpopulation that significantly contributes to lung injury and serves as an early biomarker for predicting severe PGD in human lung transplant recipients (area under the curve [AUC] = 0.871). CD177+ neutrophils exhibit elevated oxidative phosphorylation and increased mitochondrial complex I activity, driving inflammation and the formation of neutrophil extracellular traps. Targeting mitochondrial function with the complex I inhibitor IACS-010759 reduces CD177+ neutrophil activation and alleviates lung injury in both mouse IRI and rat left lung transplant models. These findings provide a comprehensive landscape of CD177+ neutrophil-driven inflammation in lung IRI and highlight its potential value for future early diagnosis and therapeutic interventions.
Collapse
Affiliation(s)
- Junqi Wu
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Peigen Gao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chenlu Yang
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Fenghui Zhuang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Yunzhe Luo
- BGI Research, Beijing, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | | | | | - Long Wang
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Chenyang Dai
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Deping Zhao
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China
| | - Chongwu Li
- Department of Thoracic and Cardiovascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| | | | | | - Chang Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China; Shanghai Engineering Research Center of Lung Transplantation, Shanghai, China.
| |
Collapse
|
5
|
Zhong X, Xie T, Wang SY, Xu ZS, Chi XX, Lan QS, Xie BW, Sun QL, Yuan L, Lan QY, Zhao ZX, Pan BR, Feng H, Lu L, Wang YY, Wang X, Dong C. Alveolar macrophages critically control infection by seasonal human coronavirus OC43 to avoid severe pneumonia. Cell Rep 2025; 44:115531. [PMID: 40222012 DOI: 10.1016/j.celrep.2025.115531] [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: 12/25/2023] [Revised: 11/06/2024] [Accepted: 03/17/2025] [Indexed: 04/15/2025] Open
Abstract
Seasonal coronaviruses, similar to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), only cause severe respiratory symptoms in a small fraction of infected individuals. However, the host factors that determine the variable responses to coronavirus infection remain unclear. Here, we use seasonal human coronavirus OC43 (HCoV-OC43) infection as an asymptomatic model that triggers both innate and adaptive immune responses in mice. Interestingly, innate sensing pathways as well as adaptive immune cells are not essential in protection against HCoV-OC43. Instead, alveolar macrophage (AMΦ) deficiency in mice results in COVID-19-like severe pneumonia post HCoV-OC43 infection, with abundant neutrophil infiltration, neutrophil extracellular trap (NET) release, and exaggerated pro-inflammatory cytokine production. Mechanistically, AMΦ efficiently phagocytose HCoV-OC43, effectively blocking virus spread, whereas, in their absence, HCoV-OC43 triggers Toll-like receptor (TLR)-dependent chemokine production to cause pneumonia. These findings reveal the central role of AMΦ in defending against seasonal HCoV-OC43 with clinical implications for human immunopathology associated with coronavirus infection.
Collapse
Affiliation(s)
- Xuan Zhong
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Tian Xie
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Su-Yun Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Zhi-Sheng Xu
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Xin-Xin Chi
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Qiao-Shuai Lan
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Bo-Wen Xie
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China; Tsinghua-Peking Center for Life Sciences, Beijing 100084, China
| | - Qin-Li Sun
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Lei Yuan
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Qiu-Yan Lan
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Zi-Xuan Zhao
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Bi-Rui Pan
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Han Feng
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Lu Lu
- MOE/NHC/CAMS Key Laboratory of Medical Molecular Virology, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Yan-Yi Wang
- Key Laboratory of Virology and Biosafety, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, Hubei 430207, China
| | - Xiaohu Wang
- Institute for Immunology and School of Basic Medical Sciences, Tsinghua University, Beijing 100084, China
| | - Chen Dong
- Research Unit of Immune Regulation and Immune Diseases of Chinese Academy of Medical Sciences, Shanghai Jiao Tong University School of Medicine-Affiliated Renji Hospital, Shanghai 200127, China; Westlake University School of Medicine, Hangzhou, Zhejiang 310030, China.
| |
Collapse
|
6
|
Ding X, Chen X, Liu Y, He J, Zhou Y, Li J. Elucidating the inhibitory role of miR-140-5p in SARS-CoV-2 infection. Int Immunopharmacol 2025; 152:114395. [PMID: 40090081 DOI: 10.1016/j.intimp.2025.114395] [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: 03/08/2024] [Revised: 12/10/2024] [Accepted: 02/27/2025] [Indexed: 03/18/2025]
Abstract
The ongoing COVID-19 pandemic, caused by the novel coronavirus SARS-CoV-2, has necessitated an urgent need for understanding the molecular mechanisms underlying viral infection and host response. MicroRNAs (miRNAs) have emerged as key regulators in viral pathogenesis, mediating complex interactions between the virus and the host's cellular machinery. In this study, we identify miR-140-5p as a significant factor in the regulation of SARS-CoV-2 entry into host cells. Through comprehensive RNA sequencing analysis of peripheral blood mononuclear cells from COVID-19 patients, we observed significant alterations in the expression of miR-140-5p and its target genes during infection. Further bioinformatics analysis revealed that miR-140-5p targets are predominantly associated with endocytosis-related signaling pathways, suggesting a mechanism by which miR-140-5p may influence SARS-CoV-2 entry. Experimental validation using miR-140-5p mimics demonstrated a significant reduction in viral entry across multiple SARS-CoV-2 variants, confirming the inhibitory role of miR-140-5p on viral replication. These findings suggest that miR-140-5p could potentially be explored as a target for inhibiting viral entry, providing new insights into the role of host miRNAs in SARS-CoV-2 infection and the development of antiviral strategies.
Collapse
Affiliation(s)
- Xiaoyan Ding
- College of Basic Medicine, Army Medical University, Chongqing 400038, China; Department of Pediatrics, Ludwig-Maximilians University of Munich, Munich 80337, Germany
| | - Xiaozhong Chen
- College of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Yuheng Liu
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing 400016, China; Chongqing Key Laboratory of Drug Metabolism, Chongqing Medical University, Chongqing 400016, China
| | - Jiuxiang He
- College of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Yuxin Zhou
- College of Basic Medicine, Army Medical University, Chongqing 400038, China
| | - Jintao Li
- College of Basic Medicine, Army Medical University, Chongqing 400038, China.
| |
Collapse
|
7
|
Hu M, Oliveira APBN, Fang Z, Feng Y, Miranda M, Kowli S, Arunachalam PS, Vasudevan G, Hui HSY, Grifoni A, Sette A, Litvack M, Rouphael N, Suthar MS, Ji X, Maecker HT, Hagan T, Dhillon G, Nicolls MR, Pulendran B. Altered baseline immunological state and impaired immune response to SARS-CoV-2 mRNA vaccination in lung transplant recipients. Cell Rep Med 2025; 6:102050. [PMID: 40187358 PMCID: PMC12047491 DOI: 10.1016/j.xcrm.2025.102050] [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: 08/28/2024] [Revised: 12/08/2024] [Accepted: 03/07/2025] [Indexed: 04/07/2025]
Abstract
The effectiveness of COVID-19 mRNA vaccines is diminished in organ transplant patients. Using a multi-omics approach, we investigate the immunological state of lung transplant (LTX) recipients at baseline and after SARS-CoV-2 mRNA vaccination compared to healthy controls (HCs). LTX patients exhibit a baseline immune profile resembling severe COVID-19 and sepsis, characterized by elevated pro-inflammatory cytokines (e.g., EN-RAGE [also known as S100A12], interleukin [IL]-6), reduced human leukocyte antigen (HLA)-DR expression on monocytes and dendritic cells, impaired cytokine production, and increased plasma microbial products. Single-cell RNA sequencing identifies an enriched monocyte cluster in LTX patients marked by high S100A family expression and reduced cytokine and antigen presentation genes. Post vaccination, LTX patients show diminished antibody, B cell, and T cell responses, along with blunted innate immune signatures. Integrative analysis links these altered baseline immunological features to impaired vaccine responses. These findings provide critical insights into the immunosuppressed condition of LTX recipients and their reduced vaccine-induced adaptive and innate immune responses.
Collapse
Affiliation(s)
- Mengyun Hu
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Ana Paula B N Oliveira
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Zhuoqing Fang
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Yupeng Feng
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Institute of Infectious Diseases, Guangzhou Eighth People's Hospital, Guangzhou Medical University, Guangzhou, China
| | - Molly Miranda
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Sangeeta Kowli
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Prabhu S Arunachalam
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Department of Immunobiology, University of Arizona, Tucson, AZ, USA
| | - Gowri Vasudevan
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Harold Sai-Yin Hui
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Alba Grifoni
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA
| | - Alessandro Sette
- Center for Infectious Disease and Vaccine Research, La Jolla Institute for Immunology, La Jolla, CA, USA; Department of Pathology, University of California, San Diego, La Jolla, CA 92093, USA
| | - Matthew Litvack
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA, USA
| | - Nadine Rouphael
- Hope Clinic of the Emory Vaccine Center, Department of Medicine, Division of Infectious Diseases, Emory University School of Medicine, Decatur, GA, USA
| | - Mehul S Suthar
- Department of Pediatrics, Emory Vaccine Center, Emory National Primate Research Center, Atlanta, GA, USA
| | - Xuhuai Ji
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA
| | - Thomas Hagan
- Department of Infectious Diseases, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Gundeep Dhillon
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Mark R Nicolls
- Division of Pulmonary, Allergy and Critical Care Medicine, Stanford University, Stanford, CA, USA
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA; Department of Pathology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
8
|
Ejtehadifar M, Zahedi S, Gameiro P, Cabeçadas J, Rodriguez MS, da Silva MG, Beck HC, Matthiesen R, Carvalho AS. Proteome alterations in peripheral immune cells of DLBCL patients and evidence of cancer extracellular vesicles involvement. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167842. [PMID: 40222457 DOI: 10.1016/j.bbadis.2025.167842] [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: 01/13/2025] [Revised: 03/14/2025] [Accepted: 04/08/2025] [Indexed: 04/15/2025]
Abstract
Diffuse large B-cell lymphoma (DLBCL) is an aggressive disease and a frequent form of non-Hodgkin lymphoma. Given the primary localization of DLBCL and the effect of tumors on the systemic immune response, we investigated the proteome of DLBCL patients' and healthy donors (HDs') peripheral immune cells (PICs). Since the ubiquitin-proteasome system has a vital role in proteome regulation and immune cells' functions, this study also explores the potential impact of DLBCL secretome on the polyubiquitination level in PICs. PICs from DLBCL patients and HDs were isolated and analyzed by mass spectrometry-based proteomics. The analysis resulted in 135 down and 51 upregulated proteins (adjusted p-value <0.05). Unsupervised principal component analysis revealed distinct proteomic profiles between DLBCL and HDs. Functional enrichment analysis for comparison between DLBCL and HDs-PICs proteome identified immune-related pathways such as innate immune system, specifically neutrophil degranulation, Fcγ receptor-dependent phagocytosis, and JAK-STAT signaling after IL-12 stimulation as downregulated. Proteomics analysis of DLBCL-PICs also showed dysregulation of proteostasis factors. This prompted the investigation of the effect of tumor secretome on viability and polyubiquitination level in mononuclear immune cells. Therefore, human HD peripheral blood mononuclear cells (PBMCs) were cultured in the presence of DLBCL cell line-derived soluble factors, small-EVs, and large-EVs in vitro. Our results revealed that exposure of mainly small-EVs, and large-EVs to HD PBMCs increased the polyubiquitination in PBMCs and decreased PIC viability. These findings suggest impaired immune responses in DLBCL-PICs, with tumor secretome-inducing polyubiquitination and reduced PIC viability.
Collapse
Affiliation(s)
- Mostafa Ejtehadifar
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Sara Zahedi
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal
| | - Paula Gameiro
- Instituto Português de Oncologia, Departament of Hematology, Lisbon, Portugal
| | - José Cabeçadas
- Instituto Português de Oncologia, Departament of Hematology, Lisbon, Portugal
| | - Manuel S Rodriguez
- Laboratoire de Chimie de Coordination (LCC) - UPR 8241 CNRS, UMR 152 Pharma-Dev, Université de Toulouse, IRD, UPS, 31400 and BMolecular, Centre Pierre Potiers, 31100 Toulouse, France
| | | | - Hans Christian Beck
- Centre for Clinical Proteomics, Department of Clinical Biochemistry, Odense University Hospital, DK-5000 Odense C, Denmark
| | - Rune Matthiesen
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal.
| | - Ana Sofia Carvalho
- iNOVA4Health, NOVA Medical School, Faculdade de Ciências Médicas, Universidade NOVA de Lisboa, Lisboa, Portugal.
| |
Collapse
|
9
|
Xu H, Zhan M, Wu Z, Chen J, Zhao Y, Feng F, Wang F, Li Y, Zhang S, Liu Y. Aberrant expansion of CD177 + neutrophils promotes endothelial dysfunction in systemic lupus erythematosus via neutrophil extracellular traps. J Autoimmun 2025; 152:103399. [PMID: 40088615 DOI: 10.1016/j.jaut.2025.103399] [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: 07/23/2024] [Revised: 02/22/2025] [Accepted: 03/10/2025] [Indexed: 03/17/2025]
Abstract
OBJECTIVE Aberrant neutrophil activation is implicated in the pathogenesis of systemic lupus erythematosus (SLE) and its related comorbidities. We found that CD177 was one of the most highly up-regulated genes at the transcriptional level in purified neutrophils from SLE patients. In this study, we aimed to explore the role of CD177+ neutrophils in the pathogenesis of SLE. METHODS Expression of CD177 was analyzed by neutrophil transcriptome and flow cytometry. CD177+ neutrophils and CD177-neutrophils were isolated to determine the role of neutrophils-derived NETs in endothelium dysfunction. Wild type and CD177-/- murine model of lupus were analyzed for organ involvement, endothelium-dependent vasorelaxation, serum autoantibodies, and innate and adaptive immune responses in an imiquimod (IMQ)-induced lupus model. RESULTS CD177MFI-hi neutrophils and CD177MFI-hi low-density granulocytes (LDGs) were expanded in active SLE, which were weakly but significantly associated with disease activity. CD177+neutrophils displayed enhanced production of reactive oxygen species (ROS) and NETs, which impaired the murine aortic endothelium-dependent vasorelaxation and induced endothelial cell apoptosis. Moreover, CD177-/- mice exposed to IMQ showed alleviated splenomegaly, endothelium-dependent vasorelaxation, and renal immune complex deposition. CONCLUSIONS Our findings indicated that CD177 MFI-hi may serve as a novel biomarker for monitoring disease activity in SLE. Further, CD177+ neutrophils may play a vasculopathic role in cardiovascular disease (CVD) via NETs formation, suggesting that specific targeting CD177+ neutrophil subset may have therapeutic effect in SLE but reducing the levels of NETs-prone neutrophils.
Collapse
MESH Headings
- Lupus Erythematosus, Systemic/immunology
- Lupus Erythematosus, Systemic/pathology
- Lupus Erythematosus, Systemic/metabolism
- Lupus Erythematosus, Systemic/etiology
- Animals
- Extracellular Traps/immunology
- Extracellular Traps/metabolism
- Neutrophils/immunology
- Neutrophils/metabolism
- Mice
- Humans
- Mice, Knockout
- Disease Models, Animal
- Endothelium, Vascular/immunology
- Endothelium, Vascular/pathology
- Endothelium, Vascular/metabolism
- Isoantigens/metabolism
- Isoantigens/genetics
- GPI-Linked Proteins/genetics
- GPI-Linked Proteins/metabolism
- Reactive Oxygen Species/metabolism
- Female
- Male
- Receptors, Cell Surface/metabolism
- Receptors, Cell Surface/genetics
- Neutrophil Activation/immunology
- Endothelial Cells
- Vasodilation
Collapse
Affiliation(s)
- Honglin Xu
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Minghua Zhan
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, 100730, China
| | - Ziyan Wu
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Jianing Chen
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yanling Zhao
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Futai Feng
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Fang Wang
- Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongzhe Li
- Department of Clinical Laboratory, State Key Laboratory of Complex, Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Shulan Zhang
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, State Key Laboratory of Complex Severe and Rare Diseases, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| | - Yudong Liu
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, 100730, China; Department of Rheumatology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Clinical Immunology Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
10
|
Arunachalam V, Tran KN, Hoy W, Lea RA, Nagaraj SH. Regional autozygosity association with albumin-to-creatinine ratio reveals a novel FTO region in an Indigenous Australian population. Eur J Hum Genet 2025:10.1038/s41431-025-01799-9. [PMID: 39994404 DOI: 10.1038/s41431-025-01799-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 01/10/2025] [Accepted: 01/23/2025] [Indexed: 02/26/2025] Open
Abstract
The genetic distinctiveness of Indigenous Australian populations is well established, yet the Tiwi population remains underrepresented in genetic research. Due to their prolonged geographic isolation, these populations are prone to increased runs of homozygosity (ROH). We investigated the genetic diversity of the Tiwi population, isolated from mainland Australia for decades, based on ROH and their associations with clinical traits. We analyzed 455 whole genome sequences to identify population structure via PCA and performed a comparison with UK Biobank, Melanesian, and Polynesian cohorts. ROH assessment and genome-wide and regional measures of homozygosity were used to explore associations between clinical traits and autozygosity. Our analysis revealed distinct genetic characteristics of the Tiwi population that aligned closely with those of the Melanesian cohort. Tiwi individuals exhibited an increased burden of ROH, particularly in LINC0109, FMLN1, and RPL17P45 genes on chromosomes 2, 17, and 18, respectively, indicating prolonged isolation and genetic drift. A positive correlation was observed between genomic FROH and albumin-to-creatinine ratio (ACR) levels, suggesting a potential link between autozygosity and renal health markers. Furthermore, regional autozygosity association analysis revealed an association between elevated ACR and a region in FTO, implicating its role in obesity, kidney disease, and cardiovascular conditions. Importantly, we found that this association is strong under the recessive model. This research lays a robust foundation for further exploration of ROH mapping and its implications for disease susceptibility within Indigenous communities worldwide.
Collapse
Affiliation(s)
- Vignesh Arunachalam
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Kim N Tran
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Wendy Hoy
- Faculty of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Rodney A Lea
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia
| | - Shivashankar H Nagaraj
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD, Australia.
| |
Collapse
|
11
|
Viz-Lasheras S, Gómez-Carballa A, Pardo-Seco J, Bello X, Rivero-Calle I, Dacosta AI, Kaforou M, Habgood-Coote D, Cunnington AJ, Emonts M, Herberg JA, Wright VJ, Carrol ED, Paulus SC, Zenz W, Kohlfürst DS, Van der Flier M, de Groot R, Schlapbach LJ, Agyeman P, Pollard AJ, Fink C, Kuijpers TT, Anderson S, Calvo C, Martínez-Padilla MDC, Pérez-Aragón A, Gómez-Sánchez E, Valencia-Ramos J, Giménez-Sánchez F, Alonso-Quintela P, Moreno-Galarraga L, von Both U, Pokorn M, Zavadska D, Tsolia M, Vermont CL, Moll HA, Levin M, Martinón-Torres F, Salas A, on behalf of EUCLIDS, DIAMONDS, GENDRES and, PERFORM consortia. A 5-transcript signature for discriminating viral and bacterial etiology in pediatric pneumonia. iScience 2025; 28:111747. [PMID: 39906557 PMCID: PMC11791257 DOI: 10.1016/j.isci.2025.111747] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Revised: 10/24/2024] [Accepted: 01/02/2025] [Indexed: 02/06/2025] Open
Abstract
Pneumonia stands as the primary cause of death among children under five, yet current diagnosis methods often result in inadequate or unnecessary treatments. Our research seeks to address this gap by identifying host transcriptomic biomarkers in the blood of children with definitive viral and bacterial pneumonia. We performed RNA sequencing on 192 prospectively collected whole blood samples, including 38 controls and 154 pneumonia cases, uncovering a 5-transcript signature (genes FAM20A, BAG3, TDRD9, MXRA7, and KLF14) that effectively distinguishes bacterial from viral pneumonia (area under the curve (AUC): 0.95 [0.88-1.00]). Initial validation using combined definitive and probable cases yielded an AUC of 0.87 [0.77-0.97], while full validation in a new prospective cohort of 32 patients achieved an AUC of 0.92 [0.83-1.00]. This robust signature holds significant potential to enhance diagnostics accuracy for pediatric pneumonia, reducing diagnostic delays and unnecessary treatments and potentially transforming clinical practice.
Collapse
Affiliation(s)
- Sandra Viz-Lasheras
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Alberto Gómez-Carballa
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Jacobo Pardo-Seco
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Xabier Bello
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - Irene Rivero-Calle
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Ana Isabel Dacosta
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | | | | | - Marieke Emonts
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
| | - Jethro A. Herberg
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Victoria J. Wright
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Enitan D. Carrol
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
| | - Stephane C. Paulus
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Werner Zenz
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
| | - Daniela S. Kohlfürst
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
| | - Michiel Van der Flier
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Ronald de Groot
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
| | - Luregn J. Schlapbach
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
| | - Philipp Agyeman
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Andrew J. Pollard
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
| | - Colin Fink
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
| | - Taco T. Kuijpers
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
| | - Suzanne Anderson
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
| | - Cristina Calvo
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
| | | | - Ana Pérez-Aragón
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
| | - Esteban Gómez-Sánchez
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
| | - Juan Valencia-Ramos
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
| | | | - Paula Alonso-Quintela
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
| | - Laura Moreno-Galarraga
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
| | - Ulrich von Both
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Marko Pokorn
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
| | - Dace Zavadska
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
| | - María Tsolia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
| | | | | | - Michael Levin
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
| | - Federico Martinón-Torres
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
| | - Antonio Salas
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
| | - on behalf of EUCLIDS
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - DIAMONDS
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - GENDRES and
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| | - PERFORM consortia
- Unidade de Xenética, Instituto de Ciencias Forenses, Facultade de Medicina, Universidade de Santiago de Compostela, and Genética de Poblaciones en Biomedicina (GenPoB) Research Group, Instituto de Investigación Sanitaria (IDIS), 15706 Hospital Clínico Universitario de Santiago (SERGAS), Galicia, Spain
- Genetics, Vaccines and Infections Research Group (GenViP), Instituto de Investigación Sanitaria de Santiago, 15706 Universidade de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Respiratorias (CIBER-ES), Madrid, Spain
- Translational Pediatrics and Infectious Diseases, Department of Pediatrics, 15706 Hospital Clínico Universitario de Santiago de Compostela, Santiago de Compostela, Galicia, Spain
- Department of Infectious Disease, Imperial College London, London W2 1PG, UK
- Great North Children’s Hospital, Paediatric Immunology, Infectious Diseases & Allergy, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne NE1 4LP, UK
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne NE2 4HH, UK
- NIHR Newcastle Biomedical Research Centre based at Newcastle upon Tyne Hospitals NHS Trust and Newcastle University, Newcastle upon Tyne NE4 5PL, UK
- Department of Infectious Diseases, Alder Hey Children’s NHS Foundation Trust, Liverpool L12 2AP, UK
- Department of Clinical Infection, Microbiology and Immunology, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK
- Department of Paediatrics, University of Oxford and the NIHR Oxford Biomedical Research Centre, Oxford OX3 9DU, UK
- Department of General Paediatrics, Medical University of Graz, Graz, Auenbruggerplatz 34/2 8036, Graz, Austria
- Pediatric Infectious Diseases and Immunology, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Utrecht 3508 AB, the Netherlands
- Pediatric Infectious Diseases and Immunology, Amalia Children’s Hospital, and Section Pediatric Infectious Diseases, Laboratory of Medical Immunology, Department of Laboratory Medicine, Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen 6500 HB, the Netherlands
- Department of Intensive Care and Neonatology, and Children’s Research Center, University Children’s Hospital Zürich, University of Zürich, Zürich, Switzerland
- Department of Pediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Micropathology Ltd, University of Warwick, Warwick CV4 7EZ, UK
- Division of Pediatric Immunology, Rheumatology and Infectious diseases, Emma Children’s Hospital, Amsterdam Univiersyt Medical Center (Amsterdam UMC), Amsterdam 1105 AZ, the Netherlands
- Medical Research Council Unit at the London School of Hygiene & Tropical Medicine, Banjul, The Gambia
- General Pediatrics, Infectious and Tropical Diseases Department, Hospital La Paz, 28046 Madrid, Spain
- La Paz Research Institute (IdiPAZ), 28029 Madrid, Spain
- Faculty of Medicine, Universidad Autónoma de Madrid (UAM), 28049 Madrid, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Infecciosas (CIBERINFEC), Madrid, Spain
- Red de Investigación Traslacional en Infectología Pediátrica (RITIP), Madrid, Spain
- Unidad de Cuidados Intensivos Pediátricos, Complejo Hospitalario de Jaen, Jaen, Spain
- Hospital Universitario Virgen de las Nieves, Servicio de Pediatría, Granada, Spain
- Department of Pediatric Intensive Care Unit, Hospital Universitario de Burgos, Burgos, Spain
- Instituto Hispalense de Pediatría, Instituto Balmis de Vacunas, Almería, Spain
- Neonatal Intensive Care Unit, Complejo Asistencial Universitario de León, León, Spain
- Department of Pediatrics, Complejo Hospitalario de Navarra, Servicio Navarro de Salud, Pamplona, Spain
- IdiSNA (Instituto de Investigación Sanitaria de Navarra), Navarra Institute for Health Research, Pamplona, Spain
- Infectious Diseases, Department of Pediatrics, Dr von Hauner Children’s Hospital, University Hospital, LMU Munich, Munich, Germany
- Division of Paediatrics, University Medical Centre Ljubljana and Medical Faculty, University of Ljubljana, Ljubljana, Slovenia
- Children’s Clinical University Hospital, Rīga Stradins University, Rïga, Latvia
- Second Department of Paediatrics, National and Kapodistrian University of Athens (NKUA), School of Medicine, Panagiotis & Aglaia, Kyriakou Children’s Hospital, Athens, Greece
- Department of Pediatrics, Erasmus MC, Rotterdam, the Netherlands
| |
Collapse
|
12
|
Williams DJ, Gautam S, Creech CB, Jimenez N, Anderson EJ, Bosinger SE, Grimes T, Arnold SR, McCullers JA, Goll J, Edwards KM, Ramilo O. Transcriptomic Biomarkers Associated With Microbiological Etiology and Disease Severity in Childhood Pneumonia. J Infect Dis 2025; 231:e277-e289. [PMID: 39397536 PMCID: PMC11841634 DOI: 10.1093/infdis/jiae491] [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: 03/21/2024] [Revised: 08/28/2024] [Accepted: 10/10/2024] [Indexed: 10/15/2024] Open
Abstract
BACKGROUND Challenges remain in discerning microbiologic etiology and disease severity in childhood pneumonia. Defining host transcriptomic profiles during illness may facilitate improved diagnostic and prognostic approaches. METHODS Using whole blood RNA sequencing from 222 hospitalized children with radiographic pneumonia and 45 age-matched controls, we identified differentially expressed (DE) genes that best identified children according to detected microbial pathogens (viral only vs bacterial only and typical vs atypical bacterial [with or without [±] viral co-detection]) and an ordinal measure of phenotypic severity (moderate, severe, very severe). RESULTS Overall, 135 (61%) children had viral-only detections, 15 (7%) had typical bacterial detections (± viral co-detections), and 26 (12%) had atypical bacterial detections (± viral co-detections). Eleven DE genes distinguished between viral-only and bacterial-only detections. Sixteen DE genes distinguished between atypical and typical bacterial detections (± viral co-detections). Nineteen DE genes distinguished between levels of pneumonia severity, including 4 genes also identified in the viral-only versus bacterial-only model (IGHGP, PI3, CD177, RAP1GAP1) and 4 genes from the typical versus atypical bacterial model (PRSS23, IFI27, OLFM4, ABO). CONCLUSIONS We identified transcriptomic biomarkers associated with microbial detections and phenotypic severity in children hospitalized with pneumonia. These DE genes are promising candidates for validation and translation into diagnostic and prognostic tools.
Collapse
Affiliation(s)
- Derek J Williams
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr Children's Hospital at Vanderbilt University Medical Center, Nashville, Tennessee
| | - Shruti Gautam
- Department of Biomedical Data Science and Bioinformatics, The Emmes Company, LLC, Rockville, Maryland
| | - C Buddy Creech
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr Children's Hospital at Vanderbilt University Medical Center, Nashville, Tennessee
| | - Natalia Jimenez
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr Children's Hospital at Vanderbilt University Medical Center, Nashville, Tennessee
| | - Evan J Anderson
- Departments of Pediatrics and Medicine, Emory University School of Medicine
| | - Steven E Bosinger
- Departments of Pathology and Laboratory Medicine, Emory University, and Emory National Primate Research Center, Atlanta, Georgia
| | - Tyler Grimes
- Department of Biomedical Data Science and Bioinformatics, The Emmes Company, LLC, Rockville, Maryland
| | - Sandra R Arnold
- Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital
| | - Jonathan A McCullers
- Department of Pediatrics, University of Tennessee Health Sciences Center and Le Bonheur Children's Hospital
| | - Johannes Goll
- Department of Biomedical Data Science and Bioinformatics, The Emmes Company, LLC, Rockville, Maryland
| | - Kathryn M Edwards
- Department of Pediatrics, Vanderbilt University School of Medicine and the Monroe Carell Jr Children's Hospital at Vanderbilt University Medical Center, Nashville, Tennessee
| | - Octavio Ramilo
- Department of Infectious Diseases, St Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
13
|
Agidigbi TS, Fram B, Molloy I, Riedel M, Wiznia D, Oh I. CD177, MYBL2, and RRM2 Are Potential Biomarkers for Musculoskeletal Infections. Clin Orthop Relat Res 2025:00003086-990000000-01897. [PMID: 39915095 DOI: 10.1097/corr.0000000000003402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 01/13/2025] [Indexed: 05/16/2025]
Abstract
BACKGROUND Biomarkers of infection are measurable indicators that reflect the presence of an infection in the body. They are particularly valuable for detecting infections and tracking treatment responses. Previous transcriptome analysis of peripheral blood mononuclear cells (PBMCs) collected from patients during the active phase of diabetic foot infection identified the upregulation of several genes, including a neutrophil-specific cell surface glycoprotein, CD177, an Myb-related transcription factor 2 (MYBL2), and ribonucleotide reductase regulatory subunit M2 (RRM2). We aimed to investigate whether these observations in diabetic foot infections could be extrapolated to other musculoskeletal infections. QUESTIONS/PURPOSES (1) Are the protein concentrations of CD177, MYBL2, and RRM2 elevated in serum or PBMCs of patients with musculoskeletal infections? (2) Do serum and PBMC concentrations of CD177, MYBL2, and RRM2 decrease in response to antibiotic therapy? (3) Can these biomarkers give diagnostic accuracy and differentiate patients with musculoskeletal infections from controls? METHODS From April 2023 to June 2024, we treated 26 patients presenting with clinical symptoms and signs of acute musculoskeletal infections, including elevated inflammatory markers (white blood cell [WBC] and C-reactive protein [CRP]) and local changes such as swelling, erythema, tenderness or pain, warmth, purulent drainage, sinus tract, or wound leading to bone or hardware. Diagnosis included periprosthetic joint infection (PJI), foot and ankle infection (FAI), fracture-related infection (FRI), and septic arthritis of the native joints. Patients with chronic recurrent osteomyelitis, PJI, or FRI were excluded from the study. Among the 26 patients deemed potentially eligible, 19% (5) were excluded for the following reasons: prison inmate (1), unable to provide consent because of severe sepsis (1), mental illness (1), and declined to participate (2). Of the 81% (21) of patients who provided consent, cultures from 9.5% (2) were negative. These two patients were ultimately diagnosed with inflammatory arthritis: gout (1) and rheumatoid arthritis (1); thus, the musculoskeletal infection group for analysis consisted of 73.1% (19 of 26) of patients. A control group of 21 patients undergoing elective foot or ankle deformity correction surgery without infections or systemic inflammation was included. Because foot or ankle deformity is highly unlikely to influence the immunologic profile of the subjects, we believed that these patients would serve as an appropriate control group. Other than the absence of infection and the lower prevalence of diabetes mellitus, the control group was comparable to the study group in terms of demographics and clinical factors, including age and sex distribution. We collected blood samples from both patients and controls and quantified CD177, MYBL2, and RRM2 RNA transcription levels in the PBMC using qRT-PCR. We also assessed protein concentrations in the serum and PBMC using an enzyme-linked immunosorbent assay. A comparative analysis of the three biomarkers was performed on 19 patients with musculoskeletal infections with positive cultures and 21 controls to assess their diagnostic potential using the unpaired nonparametric t-test with the Mann-Whitney test. We obtained 8-week follow-up blood samples from seven patients with musculoskeletal infections who clinically healed. Healing was defined by normalization of inflammatory markers (WBC and CRP) and absence of swelling, erythema, local tenderness or pain, warmth, purulent drainage, sinus tract, or open wound. We performed a comparative analysis of the seven patients during active infection and after treatment to determine a change in the level of CD177, MYBL2, and RRM2 in their serum and PBMCs. These findings were also compared with those of the control group. We evaluated the diagnostic accuracy of CD177, MYBL2, and RRM2 for musculoskeletal infections using receiver operating characteristic (ROC) curve analysis. RESULTS The musculoskeletal infections group showed a larger increased serum and PBMC concentrations of CD177, MYBL2, and RRM2 proteins compared with the control group. The mean protein concentrations of CD177, MYBL2, and RRM2 were increased in the serum and PBMC of the musculoskeletal infections group compared with the controls. Serum levels of all biomarkers investigated were higher in musculoskeletal infections group compared with the control group (CD177 227 [155 to 432] versus 54 [10 to 100], difference of medians 173, p < 0.01; MYBL2 255 [231 to 314] versus 180 [148 to 214], difference of medians 75, p < 0.01; RRM2 250 [216 to 305] versus 190 [148 to 255], difference of medians 60, p < 0.01). Similarly, PBMC levels of all biomarkers were higher in the musculoskeletal infections group (CD177 55.3 [39.1 to 80.5] versus 17.5 [10.5 to 27.5], difference of medians 37.8, p < 0.01; MYBL2 144 [114 to 190] versus 91 [70 to 105], difference of medians 53, p < 0.01; RRM2 168 [143 to 202] versus 100 [77.5 to 133], difference of medians 68, p < 0.01). Additionally, serum levels of all biomarkers decreased in seven patients with musculoskeletal infections after infection treatment (CD177 3080 [2690 to 3320] versus 4250 [3100 to 8640], difference of medians 1170, p < 0.01; MYBL2 4340 [4120 to 4750] versus 5010 [4460 to 5880], difference of medians 670, p < 0.01; RRM2 4350 [3980 to 5000] versus 5025 [4430 to 6280], difference of medians 675, p = 0.01). Similarly, PBMC levels of all biomarkers were lower after infection treatment (CD177 805 [680 to 980] versus 1025 [750 to 1610], difference of medians 220, p < 0.01; MYBL2 2300 [2100 to 2550] versus 2680 [2220 to 3400], difference of medians 380, p = 0.02; RRM2 2720 [2500 to 3200] versus 3350 [2825 to 4030], difference of medians 630, p < 0.01). The area under the ROC curve for diagnosing musculoskeletal infections in the serum and PBMC was as follows: CD177 95% confidence interval [CI] > 0.99 and > 0.99, MYBL2 95% CI > 0.99 and > 0.99, and RRM2 95% CI = 0.96 and > 0.99, respectively. CONCLUSION We may utilize blood-based tests for CD177, MYBL2, and RRM2 to aid in the diagnosis of musculoskeletal infections, particularly when arthrocentesis or obtaining tissue culture is challenging. They may also assist in monitoring treatment response. As some of these biomarkers may also be elevated in other inflammatory conditions, a large-scale clinical study is needed to confirm their reliability in differentiating musculoskeletal infections from other inflammatory conditions. CLINICAL RELEVANCE CD177, MYBL2, and RRM2 proteins in blood samples may serve as novel biomarkers for diagnosing and monitoring treatment response in musculoskeletal infections.
Collapse
Affiliation(s)
- Taiwo Samuel Agidigbi
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, USA
| | - Brianna Fram
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, USA
| | - Ilda Molloy
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, USA
| | - Matthew Riedel
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, USA
| | - Daniel Wiznia
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, USA
| | - Irvin Oh
- Department of Orthopedics and Rehabilitation, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
14
|
Saqib M, Das S, Nafiz TN, McDonough E, Sankar P, Mishra LK, Zhang X, Cai Y, Subbian S, Mishra BB. Pathogenic role for CD101-negative neutrophils in the type I interferon-mediated immunopathogenesis of tuberculosis. Cell Rep 2025; 44:115072. [PMID: 39693225 PMCID: PMC11829800 DOI: 10.1016/j.celrep.2024.115072] [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: 05/01/2024] [Revised: 09/13/2024] [Accepted: 11/25/2024] [Indexed: 12/20/2024] Open
Abstract
Neutrophils are vital for immunity against Mycobacterium tuberculosis (Mtb), the causative agent of tuberculosis (TB), yet their heterogeneous nature suggests a complex role in TB pathogenesis. Here, we identify two distinct neutrophil populations based on CD101 expression, highlighting their divergent roles in TB. CD101-negative (CD101-ve) neutrophils, which resemble immature, pro-inflammatory granulocytes, exhibit reduced Mtb phagocytosis compared to their mature, CD101-positive (CD101+ve) counterparts. Our findings reveal that type I interferons (IFN-Is) suppress neutrophil Mtb uptake and drive the recruitment of CD101-ve neutrophils to the lungs. Infiltration of these cells promotes Mtb extracellular persistence, exacerbates epithelial damage, and impairs surfactant production. Furthermore, we demonstrate that granulocyte colony-stimulating factor (G-CSF) and chemokine receptor CXCR2 are essential for the pulmonary accumulation of CD101-ve neutrophils. Our study uncovers a pathogenic role for CD101-ve neutrophils in TB and highlights the IFN-I-dependent recruitment of this functionally compromised immature neutrophil as a driver of TB immunopathogenesis.
Collapse
Affiliation(s)
- Mohd Saqib
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Shreya Das
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Tanvir N Nafiz
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Elizabeth McDonough
- GE Healthcare Technology and Innovation Center, GE Research, Niskayuna, NY, USA
| | - Poornima Sankar
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Lokesh K Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA
| | - Ximeng Zhang
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Yi Cai
- Guangdong Key Laboratory of Regional Immunity and Diseases, Department of Pathogen Biology, Shenzhen University Medical School, Shenzhen, China
| | - Selvakumar Subbian
- Public Health Research Institute, New Jersey Medical School, Rutgers University, Newark, NJ, USA
| | - Bibhuti B Mishra
- Department of Immunology and Microbial Disease, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
15
|
Desale H, Tu W, Goff K, Marx PA, Herrera C, Dumonteil E. PBMC transcriptomic signatures reflect Trypanosoma cruzi strain diversity and trained immunity in chronically infected macaques. JCI Insight 2025; 10:e186003. [PMID: 39774119 PMCID: PMC11949070 DOI: 10.1172/jci.insight.186003] [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: 08/16/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
Chagas disease is a tropical disease caused by Trypanosoma cruzi with clinical presentations ranging from asymptomatic to cardiac and/or gastrointestinal complications. The mechanisms of pathogenesis are still poorly understood, but T. cruzi strain diversity may be associated with disease progression. Therefore, we evaluated the transcriptomic response of PBMCs from macaques with natural chronic infections and tested for heterogeneity in their gene signatures. Remarkably, transcriptomic response to T. cruzi infection matched parasite strain profiles, indicating that parasite diversity is a key determinant of host response. While differences in adaptive immune responses were identified, more striking alterations of innate immune processes were detected. Thus, initial innate response to T. cruzi infection may be conditioned by parasite strain diversity, resulting in different profiles of trained immunity modulating subsequent adaptive responses, allowing parasite control or its persistence during the chronic phase. These results call for further characterization of the cross-talk between innate and adaptive immunity according to parasite diversity as well as how altered trained immunity contributes to pathogenesis, as this may lead to better treatments and vaccines.
Collapse
Affiliation(s)
- Hans Desale
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Weihong Tu
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Kelly Goff
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Preston A. Marx
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and
- Division of Microbiology, Tulane National Primate Research Center, Tulane University, Covington, Louisiana, USA
| | - Claudia Herrera
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| | - Eric Dumonteil
- Department of Tropical Medicine and Infectious Disease, School of Public Health and Tropical Medicine, and
- Vector-Borne and Infectious Disease Research Center, Tulane University, New Orleans, Louisiana, USA
| |
Collapse
|
16
|
Valcenko A, Zwick A, Schneider L, Linxweiler M, Lohse S. The tumor cell killing capacity of head and neck cancer patient-derived neutrophils depends on tumor stage, gender and the antibody isotype. Oral Oncol 2024; 159:107042. [PMID: 39306937 DOI: 10.1016/j.oraloncology.2024.107042] [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: 07/31/2024] [Revised: 09/13/2024] [Accepted: 09/15/2024] [Indexed: 11/30/2024]
Abstract
Neutrophils play a crucial role in the tumor microenvironment (TME) of head and neck squamous cell carcinomas (HNSCC) and significantly influence treatment outcomes. Phenotypic and functional properties of neutrophils adapt to the TME with distinct subsets modulating disease progression and therapeutic interventions. Here, we evaluated phenotypic and functional differences of neutrophils derived from HNSCC patients and healthy donors. We observed significant phenotypic differences between neutrophils from healthy donors and HNSCC patient-derived neutrophils. Gender and tumor stage influenced neutrophil phenotypes and their ability to lyse tumor cells through antibody-dependent cell-mediated cytotoxicity (ADCC). Patients with advanced HNSCC and males may benefit less from neutrophil-centered immunotherapy. An engineered IgA2 antibody specific for the epidermal growth factor receptor (EGFR) demonstrated superior efficacy in activating neutrophils for ADCC compared to Panitumumab using healthy and patient-derived neutrophils, underscoring the potential of the IgA isotype as a therapeutic alternative. The distinct behavior and antibody-isotype dependent ADCC competence of CD177+/- neutrophils of healthy but not HNSCC donors warrants further exploration. Our study emphasizes the importance of personalized immunotherapy treatments that consider the characteristics of neutrophils, patient demographics, and the type of antibody to improve ADCC and ultimately enhance treatment outcomes for HNSCC.
Collapse
Affiliation(s)
- Alexander Valcenko
- Institute for Virology, Saarland University Medical Center, Homburg, Germany
| | - Anabel Zwick
- Institute for Virology, Saarland University Medical Center, Homburg, Germany
| | - Lissy Schneider
- Institute for Virology, Saarland University Medical Center, Homburg, Germany
| | - Maximilian Linxweiler
- Department of Otorhinolaryngology, Head and Neck Surgery, Saarland University Medical Center, 66421, Homburg/Saar, Germany
| | - Stefan Lohse
- Institute for Virology, Saarland University Medical Center, Homburg, Germany.
| |
Collapse
|
17
|
Tsakiroglou M, Evans A, Doce-Carracedo A, Little M, Hornby R, Roberts P, Zhang E, Miyajima F, Pirmohamed M. Gene Expression Dysregulation in Whole Blood of Patients with Clostridioides difficile Infection. Int J Mol Sci 2024; 25:12653. [PMID: 39684365 DOI: 10.3390/ijms252312653] [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: 09/21/2024] [Revised: 11/19/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
Clostridioides difficile (C. difficile) is a global threat and has significant implications for individuals and health care systems. Little is known about host molecular mechanisms and transcriptional changes in peripheral immune cells. This is the first gene expression study in whole blood from patients with C. difficile infection. We took blood and stool samples from patients with toxigenic C. difficile infection (CDI), non-toxigenic C. difficile infection (GDH), inflammatory bowel disease (IBD), diarrhea from other causes (DC), and healthy controls (HC). We performed transcriptome-wide RNA profiling on peripheral blood to identify diarrhea common and CDI unique gene sets. Diarrhea groups upregulated innate immune responses with neutrophils at the epicenter. The common signature associated with diarrhea was non-specific and shared by various other inflammatory conditions. CDI had a unique 45 gene set reflecting the downregulation of humoral and T cell memory functions. Dysregulation of immunometabolic genes was also abundant and linked to immune cell fate during differentiation. Whole transcriptome analysis of white cells in blood from patients with toxigenic C. difficile infection showed that there is an impairment of adaptive immunity and immunometabolism.
Collapse
Affiliation(s)
- Maria Tsakiroglou
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Anthony Evans
- Computational Biology Facility, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool L69 7BE, UK
| | - Alejandra Doce-Carracedo
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Clinical Directorate, GCP Laboratories, University of Liverpool, Liverpool L7 8TX, UK
| | - Margaret Little
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Rachel Hornby
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Paul Roberts
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Faculty of Science and Engineering, School of Biomedical Science and Physiology, University of Wolverhampton, Wolverhampton WV1 1LZ, UK
| | - Eunice Zhang
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| | - Fabio Miyajima
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
- Oswaldo Cruz Foundation (Fiocruz), Branch Ceara, Eusebio 61773-270, Brazil
| | - Munir Pirmohamed
- Department of Pharmacology and Therapeutics, Institute of Systems Molecular and Integrative Biology, University of Liverpool, Liverpool L69 3GL, UK
| |
Collapse
|
18
|
Guo MMH, Kuo HC. Promising biomarkers of Kawasaki disease: markers that aid in diagnosis. Expert Rev Mol Diagn 2024:1-13. [PMID: 39556196 DOI: 10.1080/14737159.2024.2432025] [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: 07/06/2024] [Revised: 09/29/2024] [Accepted: 11/17/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION Currently the diagnosis of Kawasaki disease is still heavily reliant on clinical criteria which may be subject to interpretation or mimic other common febrile diseases of childhood. Biomarkers that can aid in the accurate and timely diagnosis of KD are of great clinical utility. AREAS COVERED A literature search of PubMed was performed using the key words: Kawasaki disease, diagnosis, biomarkers, proteomics and transcriptomics. In this article we review biomarkers that are widely clinically available including NT-ProBNP and ferritin. We also include promising novel biomarkers that have been identified through newer transcriptomic and proteomic techniques. EXPERT OPINION While the identification of biomarkers that can accurately assist in diagnosing patients with KD is a promising field of research, more still remains to be done to in order to validate new biomarkers in larger cohorts, and to set standardized cutoff values for potential biomarkers that are currently clinically available. Further research is needed before KD biomarkers that are consistent, readily available, and cost-effective can be a clinical reality.
Collapse
Affiliation(s)
- Mindy Ming-Huey Guo
- Department of Pediatrics and Kawasaki Disease Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- School of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Ho-Chang Kuo
- Department of Pediatrics and Kawasaki Disease Center, Kaohsiung Chang Gung Memorial Hospital and Chang Gung University College of Medicine, Kaohsiung, Taiwan
- Department of Respiratory Therapy, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| |
Collapse
|
19
|
Shirvani P, Shirvani A, Holick MF. Decoding the Genetic Basis of Mast Cell Hypersensitivity and Infection Risk in Hypermobile Ehlers-Danlos Syndrome. Curr Issues Mol Biol 2024; 46:11613-11629. [PMID: 39451569 PMCID: PMC11506785 DOI: 10.3390/cimb46100689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 10/15/2024] [Accepted: 10/16/2024] [Indexed: 10/26/2024] Open
Abstract
Hypermobile Ehlers-Danlos syndrome (hEDS) is a connective tissue disorder marked by joint hypermobility, skin hyperextensibility, and tissue fragility. Recent studies have linked hEDS with mast cell activation syndrome (MCAS), suggesting a genetic interplay affecting immune regulation and infection susceptibility. This study aims to decode the genetic basis of mast cell hypersensitivity and increased infection risk in hEDS by identifying specific genetic variants associated with these conditions. We conducted whole-genome sequencing (WGS) on 18 hEDS participants and 7 first-degree relatives as controls, focusing on identifying genetic variants associated with mast cell dysregulation. Participants underwent clinical assessments to document hEDS symptoms and mast cell hypersensitivity, with particular attention to past infections and antihistamine response. Our analysis identified specific genetic variants in MT-CYB, HTT, MUC3A, HLA-B and HLA-DRB1, which are implicated in hEDS and MCAS. Protein-protein interaction (PPI) network analysis revealed significant interactions among identified variants, highlighting their involvement in pathways related to antigen processing, mucosal protection, and collagen synthesis. Notably, 61.1% of the hEDS cohort reported recurrent infections compared to 28.5% in controls, and 72.2% had documented mast cell hypersensitivity versus 14.2% in controls. These findings provide a plausible explanation for the complex interplay between connective tissue abnormalities and immune dysregulation in hEDS. The identified genetic variants offer insights into potential therapeutic targets for modulating mast cell activity and improving patient outcomes. Future research should validate these findings in larger cohorts and explore the functional implications of these variants to develop effective treatment strategies for hEDS and related mast cell disorders.
Collapse
Affiliation(s)
| | - Arash Shirvani
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| | - Michael F. Holick
- Section of Endocrinology, Diabetes, Nutrition and Weight Management, Department of Medicine, Boston University School of Medicine, Boston, MA 02118, USA
| |
Collapse
|
20
|
Friedrich VD, Pennitz P, Wyler E, Adler JM, Postmus D, Müller K, Teixeira Alves LG, Prigann J, Pott F, Vladimirova D, Hoefler T, Goekeri C, Landthaler M, Goffinet C, Saliba AE, Scholz M, Witzenrath M, Trimpert J, Kirsten H, Nouailles G. Neural network-assisted humanisation of COVID-19 hamster transcriptomic data reveals matching severity states in human disease. EBioMedicine 2024; 108:105312. [PMID: 39317638 DOI: 10.1016/j.ebiom.2024.105312] [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: 04/05/2024] [Revised: 08/12/2024] [Accepted: 08/13/2024] [Indexed: 09/26/2024] Open
Abstract
BACKGROUND Translating findings from animal models to human disease is essential for dissecting disease mechanisms, developing and testing precise therapeutic strategies. The coronavirus disease 2019 (COVID-19) pandemic has highlighted this need, particularly for models showing disease severity-dependent immune responses. METHODS Single-cell transcriptomics (scRNAseq) is well poised to reveal similarities and differences between species at the molecular and cellular level with unprecedented resolution. However, computational methods enabling detailed matching are still scarce. Here, we provide a structured scRNAseq-based approach that we applied to scRNAseq from blood leukocytes originating from humans and hamsters affected with moderate or severe COVID-19. FINDINGS Integration of data from patients with COVID-19 with two hamster models that develop moderate (Syrian hamster, Mesocricetus auratus) or severe (Roborovski hamster, Phodopus roborovskii) disease revealed that most cellular states are shared across species. A neural network-based analysis using variational autoencoders quantified the overall transcriptomic similarity across species and severity levels, showing highest similarity between neutrophils of Roborovski hamsters and patients with severe COVID-19, while Syrian hamsters better matched patients with moderate disease, particularly in classical monocytes. We further used transcriptome-wide differential expression analysis to identify which disease stages and cell types display strongest transcriptional changes. INTERPRETATION Consistently, hamsters' response to COVID-19 was most similar to humans in monocytes and neutrophils. Disease-linked pathways found in all species specifically related to interferon response or inhibition of viral replication. Analysis of candidate genes and signatures supported the results. Our structured neural network-supported workflow could be applied to other diseases, allowing better identification of suitable animal models with similar pathomechanisms across species. FUNDING This work was supported by German Federal Ministry of Education and Research, (BMBF) grant IDs: 01ZX1304B, 01ZX1604B, 01ZX1906A, 01ZX1906B, 01KI2124, 01IS18026B and German Research Foundation (DFG) grant IDs: 14933180, 431232613.
Collapse
Affiliation(s)
- Vincent D Friedrich
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany; Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Leipzig, Germany
| | - Peter Pennitz
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany
| | - Emanuel Wyler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Julia M Adler
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Dylan Postmus
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Liverpool School of Tropical Medicine, Department of Tropical Disease Biology, Liverpool, United Kingdom
| | - Kristina Müller
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany
| | - Luiz Gustavo Teixeira Alves
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany
| | - Julia Prigann
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Gladstone Institutes, San Francisco, USA
| | - Fabian Pott
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Thomas Hoefler
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Cengiz Goekeri
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany; Cyprus International University, Faculty of Medicine, Nicosia, Cyprus
| | - Markus Landthaler
- Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin Institute for Medical Systems Biology (BIMSB), Berlin, Germany; Humboldt-Universität zu Berlin, Institut fuer Biologie, Berlin, Germany
| | - Christine Goffinet
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Institute of Virology, Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany; Liverpool School of Tropical Medicine, Department of Tropical Disease Biology, Liverpool, United Kingdom
| | - Antoine-Emmanuel Saliba
- Faculty of Medicine, Institute of Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany; Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), Würzburg, Germany
| | - Markus Scholz
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany; University of Leipzig, Faculty of Mathematics and Computer Science, Leipzig, Germany
| | - Martin Witzenrath
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany; German Center for Lung Research (DZL), Berlin, Germany
| | - Jakob Trimpert
- Freie Universität Berlin, Institut für Virologie, Berlin, Germany
| | - Holger Kirsten
- University of Leipzig, Institute for Medical Informatics, Statistics, and Epidemiology, Leipzig, Germany.
| | - Geraldine Nouailles
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Infectious Diseases, Respiratory Medicine and Critical Care, Berlin, Germany.
| |
Collapse
|
21
|
Koss KM, Son T, Li C, Hao Y, Cao J, Churchward MA, Zhang ZJ, Wertheim JA, Derda R, Todd KG. Toward discovering a novel family of peptides targeting neuroinflammatory states of brain microglia and astrocytes. J Neurochem 2024; 168:3386-3414. [PMID: 37171455 PMCID: PMC10640667 DOI: 10.1111/jnc.15840] [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: 12/16/2022] [Revised: 04/17/2023] [Accepted: 04/19/2023] [Indexed: 05/13/2023]
Abstract
Microglia are immune-derived cells critical to the development and healthy function of the brain and spinal cord, yet are implicated in the active pathology of many neuropsychiatric disorders. A range of functional phenotypes associated with the healthy brain or disease states has been suggested from in vivo work and were modeled in vitro as surveying, reactive, and primed sub-types of primary rat microglia and mixed microglia/astrocytes. It was hypothesized that the biomolecular profile of these cells undergoes a phenotypical change as well, and these functional phenotypes were explored for potential novel peptide binders using a custom 7 amino acid-presenting M13 phage library (SX7) to identify unique peptides that bind differentially to these respective cell types. Surveying glia were untreated, reactive were induced with a lipopolysaccharide treatment, recovery was modeled with a potent anti-inflammatory treatment dexamethasone, and priming was determined by subsequently challenging the cells with interferon gamma. Microglial function was profiled by determining the secretion of cytokines and nitric oxide, and expression of inducible nitric oxide synthase. After incubation with the SX7 phage library, populations of SX7-positive microglia and/or astrocytes were collected using fluorescence-activated cell sorting, SX7 phage was amplified in Escherichia coli culture, and phage DNA was sequenced via next-generation sequencing. Binding validation was done with synthesized peptides via in-cell westerns. Fifty-eight unique peptides were discovered, and their potential functions were assessed using a basic local alignment search tool. Peptides potentially originated from proteins ranging in function from a variety of supportive glial roles, including synapse support and pruning, to inflammatory incitement including cytokine and interleukin activation, and potential regulation in neurodegenerative and neuropsychiatric disorders.
Collapse
Affiliation(s)
- K M Koss
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - T Son
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - C Li
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - Y Hao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
| | - J Cao
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - M A Churchward
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biology and Environmental Sciences, Concordia University of Edmonton, Alberta, Edmonton, Canada
| | - Z J Zhang
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
| | - J A Wertheim
- Comprehensive Transplant Center and Department of Surgery, Feinberg School of Medicine, Northwestern University, Illinois, Chicago, USA
- Department of Surgery, University of Arizona College of Medicine, Arizona, Tucson, USA
| | - R Derda
- Department of Chemistry, University of Alberta, 11227 Saskatchewan Dr NW, Edmonton, AB T6G 2G2, Canada
- 48Hour Discovery Inc, 11421 Saskatchewan Dr NW, Edmonton, AB T6G 2M9, Canada
| | - K G Todd
- Neurochemical Research Unit, Department of Psychiatry, University of Alberta, Alberta, Edmonton, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Alberta, Edmonton, Canada
- Department of Biomedical Engineering, University of Alberta, Alberta, Edmonton, Canada
| |
Collapse
|
22
|
Mao Y, Suryawanshi A, Patial S, Saini Y. Airspaces-derived exosomes contain disease-relevant protein signatures in a mouse model of cystic fibrosis (CF)-like mucoinflammatory lung disease. Front Pharmacol 2024; 15:1460692. [PMID: 39386033 PMCID: PMC11461968 DOI: 10.3389/fphar.2024.1460692] [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: 07/06/2024] [Accepted: 08/26/2024] [Indexed: 10/12/2024] Open
Abstract
Exosomes, membrane-bound extracellular vesicles, ranging from approximately 30-200 nm in diameter, are released by almost all cell types and play critical roles in intercellular communication. In response to the prevailing stress, the exosome-bound protein signatures vary in abundance and composition. To identify the bronchoalveolar lavage fluid (BALF) exosome-bound proteins associated with mucoinflammatory lung disease and to gain insights into their functional implications, we compared BALF exosomes-derived proteins from adult Scnn1b transgenic (Scnn1b-Tg+) and wild type (WT) mice. A total of 3,144 and 3,119 proteins were identified in BALF exosomes from Scnn1b-Tg+ and WT mice, respectively. Using cutoff criteria (Log2 fold-change > 1 and adjusted p-value < 0.05), the comparison of identified proteins revealed 127 and 30 proteins that were significantly upregulated and downregulated, respectively, in Scnn1b-Tg+ versus WT mice. In addition, 52 and 27 proteins were exclusively enriched in Scnn1b-Tg+ and WT mice, respectively. The identified exosome-bound proteins from the homeostatic airspaces of WT mice were mostly relevant to the normal physiological processes. The protein signatures enriched in the BALF exosomes of Scnn1b-Tg+ mice were relevant to macrophage activation and mucoinflammatory processes. Ingenuity pathway analyses revealed that the enriched proteins in Scnn1b-Tg+ mice contributed to the inflammatory responses and antimicrobial defense pathways. Selective proteins including, RETNLA/FIZZ1, LGALS3/Galectin-3, S100A8/MRP8, and CHIL3/YM1 were immunolocalized to specific cell types. The comparative analysis between enriched BALF exosome proteins and previously identified differentially upregulated genes in Scnn1b-Tg+ versus WT mice suggested that the compartment-/cell-specific upregulation in gene expression dictates the enrichment of their respective proteins in the lung airspaces. Taken together, this study demonstrates that the BALF exosome-bound protein signatures reflect disease-relevant disturbances. Our findings suggest that the exosomes carry disease-relevant protein signatures that can be used as a diagnostic as well as predictive biomarkers for mucoinflammatory lung disease.
Collapse
Affiliation(s)
- Yun Mao
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Amol Suryawanshi
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| | - Sonika Patial
- Comparative and Molecular Pathogenesis Branch, Division of Translational Toxicology, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC, United States
| | - Yogesh Saini
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, Raleigh, NC, United States
| |
Collapse
|
23
|
Zheng C, Li J, Chen H, Ma X, Si T, Zhu W. Dual role of CD177 + neutrophils in inflammatory bowel disease: a review. J Transl Med 2024; 22:813. [PMID: 39223577 PMCID: PMC11370282 DOI: 10.1186/s12967-024-05539-3] [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: 04/12/2024] [Accepted: 07/25/2024] [Indexed: 09/04/2024] Open
Abstract
Inflammatory bowel disease (IBD) represents a group of recurrent chronic inflammatory disorders associated with autoimmune dysregulation, typically characterized by neutrophil infiltration and mucosal inflammatory lesions. Neutrophils, as the earliest immune cells to arrive at inflamed tissues, play a dual role in the onset and progression of mucosal inflammation in IBD. Most of these cells specifically express CD177, a molecule increasingly recognized for its critical role in the pathogenesis of IBD. Under IBD-related inflammatory stimuli, CD177 is highly expressed on neutrophils and promotes their migration. CD177 + neutrophils activate bactericidal and barrier-protective functions at IBD mucosal inflammation sites and regulate the release of inflammatory mediators highly correlated with the severity of inflammation in IBD patients, thus playing a dual role. However, mitigating the detrimental effects of neutrophils in inflammatory bowel disease remains a challenge. Based on these data, we have summarized recent articles on the role of neutrophils in intestinal inflammation, with a particular emphasis on CD177, which mediates the recruitment, transepithelial migration, and activation of neutrophils, as well as their functional consequences. A better understanding of CD177 + neutrophils may contribute to the development of novel therapeutic targets to selectively modulate the protective role of this class of cells in IBD.
Collapse
Affiliation(s)
- Chengli Zheng
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jiekai Li
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hailin Chen
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiaolin Ma
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tianyu Si
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenwei Zhu
- Department of Hematology, Yueyang Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
24
|
Tomonaga T, Higashi H, Izumi H, Nishida C, Kawai N, Sato K, Morimoto T, Higashi Y, Yatera K, Morimoto Y. Investigation of pulmonary inflammatory responses following intratracheal instillation of and inhalation exposure to polypropylene microplastics. Part Fibre Toxicol 2024; 21:29. [PMID: 39107780 PMCID: PMC11301944 DOI: 10.1186/s12989-024-00592-8] [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: 03/01/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Microplastics have been detected in the atmosphere as well as in the ocean, and there is concern about their biological effects in the lungs. We conducted a short-term inhalation exposure and intratracheal instillation using rats to evaluate lung disorders related to microplastics. We conducted an inhalation exposure of polypropylene fine powder at a low concentration of 2 mg/m3 and a high concentration of 10 mg/m3 on 8-week-old male Fischer 344 rats for 6 h a day, 5 days a week for 4 weeks. We also conducted an intratracheal instillation of polypropylene at a low dose of 0.2 mg/rat and a high dose of 1.0 mg/rat on 12-week-old male Fischer 344 rats. Rats were dissected from 3 days to 6 months after both exposures, and bronchoalveolar lavage fluid (BALF) and lung tissue were collected to analyze lung inflammation and lung injury. RESULTS Both exposures to polypropylene induced a persistent influx of inflammatory cells and expression of CINC-1, CINC-2, and MPO in BALF from 1 month after exposure. Genetic analysis showed a significant increase in inflammation-related factors for up to 6 months. The low concentration in the inhalation exposure of polypropylene also induced mild lung inflammation. CONCLUSION These findings suggest that inhaled polypropylene, which is a microplastic, induces persistent lung inflammation and has the potential for lung disorder. Exposure to 2 mg/m3 induced inflammatory changes and was thought to be the Lowest Observed Adverse Effect Level (LOAEL) for acute effects of polypropylene. However, considering the concentration of microplastics in a real general environment, the risk of environmental hazards to humans may be low.
Collapse
Affiliation(s)
- Taisuke Tomonaga
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan.
| | - Hidenori Higashi
- Department of Environmental Health Engineering, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Hiroto Izumi
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Chinatsu Nishida
- Department of Environmental Health Engineering, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Naoki Kawai
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Kazuma Sato
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Toshiki Morimoto
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yasuyuki Higashi
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Kazuhiro Yatera
- Department of Respiratory Medicine, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| | - Yasuo Morimoto
- Department of Occupational Pneumology, Institute of Industrial Ecological Sciences, University of Occupational and Environmental Health, Japan, 1-1 Iseigaoka, Yahata-nishi-ku, Kitakyushu, Fukuoka, 807-8555, Japan
| |
Collapse
|
25
|
Qing L, Wu W. The mechanism of geniposide in patients with COVID-19 and atherosclerosis: A pharmacological and bioinformatics analysis. Medicine (Baltimore) 2024; 103:e39065. [PMID: 39093733 PMCID: PMC11296471 DOI: 10.1097/md.0000000000039065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/01/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024] Open
Abstract
In patients with severe acute respiratory syndrome coronavirus 2 (which causes coronavirus disease 2019 [COVID-19]), oxidative stress (OS) is associated with disease severity and death. OS is also involved in the pathogenesis of atherosclerosis (AS). Previous studies have shown that geniposide has anti-inflammatory and anti-viral properties, and can protect cells against OS. However, the potential target(s) of geniposide in patients with COVID-19 and AS, as well as the mechanism it uses, are unclear. We combined pharmacology and bioinformatics analysis to obtain geniposide against COVID-19/AS targets, and build protein-protein interaction network to filter hub genes. The hub genes were performed an enrichment analysis by ClueGO, including Gene Ontology and KEGG. The Enrichr database and the target microRNAs (miRNAs) of hub genes were predicted through the MiRTarBase via Enrichr. The common miRNAs were used to construct the miRNAs-mRNAs regulated network, and the miRNAs' function was evaluated by mirPath v3.0 software. Two hundred forty-seven targets of geniposide were identified in patients with COVID-19/AS comorbidity by observing the overlap between the genes modulated by geniposide, COVID-19, and AS. A protein-protein interaction network of geniposide in patients with COVID-19/AS was constructed, and 27 hub genes were identified. The results of enrichment analysis suggested that geniposide may be involved in regulating the OS via the FoxO signaling pathway. MiRNA-mRNA network revealed that hsa-miR-34a-5p may play an important role in the therapeutic mechanism of geniposide in COVID-19/AS patients. Our study found that geniposide represents a promising therapy for patients with COVID-19 and AS comorbidity. Furthermore, the target genes and miRNAs that we identified may aid the development of new treatment strategies against COVID-19/AS.
Collapse
Affiliation(s)
- Lijin Qing
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| | - Wei Wu
- First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong 510405, China
| |
Collapse
|
26
|
Zhang L, Li Y, Hu W, Gao S, Tang Y, Sun L, Jiang N, Xiao Z, Han L, Zhou W. Computational identification of mitochondrial dysfunction biomarkers in severe SARS-CoV-2 infection: Facilitating therapeutic applications of phytomedicine. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 131:155784. [PMID: 38878325 DOI: 10.1016/j.phymed.2024.155784] [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: 12/11/2023] [Revised: 03/18/2024] [Accepted: 04/13/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Currently, SARS-CoV-2 has not disappeared and continues to prevail worldwide, with the ongoing risk of mutations and the potential for severe COVID-19. The impairment of monocyte mitochondrial function caused by SARS-CoV-2, leading to a metabolic and immune dysregulation, is a crucial factor in the development of severe COVID-19. PURPOSE Discover effective phytomedicines based on mitochondrial-related biomarkers in severe SARS-CoV-2 infection. METHODS Firstly, differential gene analysis and gene set enrichment analysis (GSEA) were conducted on monocytes datasets to identify genes and pathways distinguishing severe patients from uninfected individuals. Then, GO and KEGG enrichment analysis on the differentially expressed genes (DEGs) obtained. Take the DEGs and intersect them with the MitoCarta 3.0 gene set to obtain the differentially expressed mitochondrial-related genes (DE-MRGs). Subsequently, machine learning algorithms were employed to screen potential mitochondrial dysfunction biomarkers for severe COVID-19 based on score values. ROC curves were then plotted to assess the distinguish capability of the biomarkers, followed by validation using two additional independent datasets. Next, the effects of the identified biomarkers on metabolic pathways and immune cells were explored through Gene Set Variation Analysis (GSVA) and CIBERSORT. Finally, potential nature products for severe COVID-19 were screened from the expression profile dataset based on dysregulated mitochondrial-related genes, followed by in vitro experimental validation. RESULTS There are 1812 DEGs and 17 dysregulated mitochondrial processes between severe COVID-19 patients and uninfected individuals. A total of 77 DE-MRGs were identified, and the potential biomarkers were identified as RECQL4, PYCR1, PIF1, POLQ, and GLDC. In both the training and validation sets, the area under the ROC curve (AUC) for these five biomarkers was greater than 0.9. And they did not show significant changes in mild to moderate patients (p > 0.05), indicating their ability to effectively distinguish severe COVID-19. These biomarkers exhibit a highly significant correlation with the dysregulated metabolic processes (p < 0.05) and immune cell imbalance (p < 0.05) in severe patients, as demonstrated by GSVA and CIBERSORT algorithms. Curcumin has the highest score in the predictive model based on transcriptomic data from 496 natural compounds (p = 0.02; ES = 0.90). Pre-treatment with curcumin for 8 h has been shown to alleviate mitochondrial membrane potential damage caused by the SARS-CoV-2 S1 protein (p < 0.05) and reduce elevated levels of reactive oxygen species (ROS) (p < 0.01). CONCLUSION The results of this study indicate a significant correlation between severe SARS-CoV-2 infection and mitochondrial dysfunction. The proposed mitochondrial dysfunction biomarkers identified in this study are associated with the disease progression, metabolic and immune changes in severe SARS-CoV-2 infected patients. Curcumin has a potential role in preventing severe COVID-19 by protecting mitochondrial function. Our findings provide new strategies for predicting the prognosis and enabling early intervention in SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Lihui Zhang
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Yuehan Li
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Wanting Hu
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Shengqiao Gao
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Yiran Tang
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Lei Sun
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Ning Jiang
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Zhiyong Xiao
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China
| | - Lu Han
- Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China.
| | - Wenxia Zhou
- Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China; State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology & Toxicology, Beijing 100850, China.
| |
Collapse
|
27
|
Meng L, Zhou M, Wang Y, Pan Y, Chen Z, Wu B, Zhao Y. CD177 on neutrophils engages stress-related behavioral changes in male mice. Brain Behav Immun 2024; 120:403-412. [PMID: 38871062 DOI: 10.1016/j.bbi.2024.06.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Revised: 05/14/2024] [Accepted: 06/10/2024] [Indexed: 06/15/2024] Open
Abstract
Persistent psychological stress can affect immune homeostasis and is a key factor in the development of depression. Many efforts are focused on the identifcation of pathways that link the immune system and mood disorders. Here, we found that psychological stress caused an increase in the frequency of brain-associated neutrophils and the level of neutrophil-specific antigen CD177 on peripheral neutrophils in male mice. Upregulated levels of blood CD177 are associated with depression in humans. Neutrophil depletion or Cd177 deficiency protected mice from stress-induced behavioral deficits. Importantly, adoptive transfer of CD177+ neutrophils from stressed mice increased the frequency of brain-associated leukocytes, including neutrophils, and caused behavioral defects in naive mice. These effects may be related to the endothelial adhesion advantage of CD177+ neutrophils and the interference of serine protease on endothelial junction. Our findings suggest a critical link between circulating CD177+ neutrophils and psychological stress-driven behavioral disorder.
Collapse
Affiliation(s)
- Ling Meng
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Mi Zhou
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yunpeng Wang
- Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China
| | - Yiming Pan
- Laboratory of Developmental Biology, Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China
| | - Zheng Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bin Wu
- Growth, Development, and Mental Health of Children and Adolescence Center, Pediatric Research Institute, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Chongqing Key Laboratory of Child Neurodevelopment and Cognitive Disorders, Children's Hospital of Chongqing Medical University, Chongqing, China.
| | - Yan Zhao
- Department of Respiratory Medicine, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China.
| |
Collapse
|
28
|
Johnson E, Long MB, Chalmers JD. Biomarkers in bronchiectasis. Eur Respir Rev 2024; 33:230234. [PMID: 38960612 PMCID: PMC11220624 DOI: 10.1183/16000617.0234-2023] [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: 11/13/2023] [Accepted: 03/09/2024] [Indexed: 07/05/2024] Open
Abstract
Bronchiectasis is a heterogeneous disease with multiple aetiologies and diverse clinical features. There is a general consensus that optimal treatment requires precision medicine approaches focused on specific treatable disease characteristics, known as treatable traits. Identifying subtypes of conditions with distinct underlying biology (endotypes) depends on the identification of biomarkers that are associated with disease features, prognosis or treatment response and which can be applied in clinical practice. Bronchiectasis is a disease characterised by inflammation, infection, structural lung damage and impaired mucociliary clearance. Increasingly there are available methods to measure each of these components of the disease, revealing heterogeneous inflammatory profiles, microbiota, radiology and mucus and epithelial biology in patients with bronchiectasis. Using emerging biomarkers and omics technologies to guide treatment in bronchiectasis is a promising field of research. Here we review the most recent data on biomarkers in bronchiectasis.
Collapse
Affiliation(s)
- Emma Johnson
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - Merete B Long
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| | - James D Chalmers
- Division of Molecular and Clinical Medicine, University of Dundee, Ninewells Hospital and Medical School, Dundee, UK
| |
Collapse
|
29
|
Li Y, Tao X, Ye S, Tai Q, You YA, Huang X, Liang M, Wang K, Wen H, You C, Zhang Y, Zhou X. A T-Cell-Derived 3-Gene Signature Distinguishes SARS-CoV-2 from Common Respiratory Viruses. Viruses 2024; 16:1029. [PMID: 39066192 PMCID: PMC11281602 DOI: 10.3390/v16071029] [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: 04/28/2024] [Revised: 06/06/2024] [Accepted: 06/21/2024] [Indexed: 07/28/2024] Open
Abstract
Research on the host responses to respiratory viruses could help develop effective interventions and therapies against the current and future pandemics from the host perspective. To explore the pathogenesis that distinguishes SARS-CoV-2 infections from other respiratory viruses, we performed a multi-cohort analysis with integrated bioinformatics and machine learning. We collected 3730 blood samples from both asymptomatic and symptomatic individuals infected with SARS-CoV-2, seasonal human coronavirus (sHCoVs), influenza virus (IFV), respiratory syncytial virus (RSV), or human rhinovirus (HRV) across 15 cohorts. First, we identified an enhanced cellular immune response but limited interferon activities in SARS-CoV-2 infection, especially in asymptomatic cases. Second, we identified a SARS-CoV-2-specific 3-gene signature (CLSPN, RBBP6, CCDC91) that was predominantly expressed by T cells, could distinguish SARS-CoV-2 infection, including Omicron, from other common respiratory viruses regardless of symptoms, and was predictive of SARS-CoV-2 infection before detectable viral RNA on RT-PCR testing in a longitude follow-up study. Thereafter, a user-friendly online tool, based on datasets collected here, was developed for querying a gene of interest across multiple viral infections. Our results not only identify a unique host response to the viral pathogenesis in SARS-CoV-2 but also provide insights into developing effective tools against viral pandemics from the host perspective.
Collapse
Affiliation(s)
- Yang Li
- Beijing International Center for Mathematical Research, Peking University, Beijing 100871, China;
- Chongqing Research Institute of Big Data, Peking University, Chongqing 400041, China; (X.T.); (X.H.)
| | - Xinya Tao
- Chongqing Research Institute of Big Data, Peking University, Chongqing 400041, China; (X.T.); (X.H.)
| | - Sheng Ye
- Chongqing Center for Disease Control and Prevention, Chongqing 400707, China;
| | - Qianchen Tai
- Department of Probability and Statistics, School of Mathematical Sciences, Peking University, Beijing 100091, China;
| | - Yu-Ang You
- Institute of Pharmaceutical Science, King’s College London, London WC2R 2LS, UK;
| | - Xinting Huang
- Chongqing Research Institute of Big Data, Peking University, Chongqing 400041, China; (X.T.); (X.H.)
| | - Mifang Liang
- NHC Key Laboratory of Medical Virology and Viral Diseases, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing 102206, China;
| | - Kai Wang
- Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Department of Infectious Diseases, Institute for Viral Hepatitis, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China;
| | - Haiyan Wen
- Chongqing International Travel Health Care Center, Chongqing 401120, China;
| | - Chong You
- Beijing International Center for Mathematical Research, Peking University, Beijing 100871, China;
- Chongqing Research Institute of Big Data, Peking University, Chongqing 400041, China; (X.T.); (X.H.)
- Shanghai Institute for Mathematics and Interdisciplinary Sciences, Fudan University, Shanghai 200433, China
| | - Yan Zhang
- Sports & Medicine Integration Research Center (SMIRC), Capital University of Physical Education and Sports, Beijing 100088, China
| | - Xiaohua Zhou
- Beijing International Center for Mathematical Research, Peking University, Beijing 100871, China;
- Chongqing Research Institute of Big Data, Peking University, Chongqing 400041, China; (X.T.); (X.H.)
- Department of Probability and Statistics, School of Mathematical Sciences, Peking University, Beijing 100091, China;
| |
Collapse
|
30
|
Zhong J, Yuan H, Yang J, Du Y, Li Z, Liu X, Yang H, Wang Z, Wang Z, Jiang L, Ren Z, Li H, Li Z, Liu Y. Bioinformatics and system biology approach to identify potential common pathogenesis for COVID-19 infection and sarcopenia. Front Med (Lausanne) 2024; 11:1378846. [PMID: 38978778 PMCID: PMC11228343 DOI: 10.3389/fmed.2024.1378846] [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: 01/30/2024] [Accepted: 06/13/2024] [Indexed: 07/10/2024] Open
Abstract
Sarcopenia is a condition characterized by age-related loss of muscle mass and strength. Increasing evidence suggests that patients with sarcopenia have higher rates of coronavirus 2019 (COVID-19) infection and poorer post-infection outcomes. However, the exact mechanism and connections between the two is unknown. In this study, we used high-throughput data from the GEO database for sarcopenia (GSE111016) and COVID-19 (GSE171110) to identify common differentially expressed genes (DEGs). We conducted GO and KEGG pathway analyses, as well as PPI network analysis on these DEGs. Using seven algorithms from the Cytoscape plug-in cytoHubba, we identified 15 common hub genes. Further analyses included enrichment, PPI interaction, TF-gene and miRNA-gene regulatory networks, gene-disease associations, and drug prediction. Additionally, we evaluated immune cell infiltration with CIBERSORT and assessed the diagnostic accuracy of hub genes for sarcopenia and COVID-19 using ROC curves. In total, we identified 66 DEGs (34 up-regulated and 32 down-regulated) and 15 hub genes associated with sarcopenia and COVID-19. GO and KEGG analyses revealed functions and pathways between the two diseases. TF-genes and TF-miRNA regulatory network suggest that FOXOC1 and hsa-mir-155-5p may be identified as key regulators, while gene-disease analysis showed strong correlations with hub genes in schizophrenia and bipolar disorder. Immune infiltration showed a correlation between the degree of immune infiltration and the level of infiltration of different immune cell subpopulations of hub genes in different datasets. The ROC curves for ALDH1L2 and KLF5 genes demonstrated their potential as diagnostic markers for both sarcopenia and COVID-19. This study suggests that sarcopenia and COVID-19 may share pathogenic pathways, and these pathways and hub genes offer new targets and strategies for early diagnosis, effective treatment, and tailored therapies for sarcopenia patients with COVID-19.
Collapse
Affiliation(s)
- Jun Zhong
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Hui Yuan
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Jinghong Yang
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Yimin Du
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Zheng Li
- Department of Orthopedics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Xu Liu
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Haibo Yang
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhaojun Wang
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Zi Wang
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Lujun Jiang
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhiqiang Ren
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Hongliang Li
- School of Clinical Medicine, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhong Li
- Department of Orthopedics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| | - Yanshi Liu
- Department of Orthopedics, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan, China
| |
Collapse
|
31
|
Geng S, Zhang Y, Lu R, Irimia D, Li L. Resolving neutrophils through genetic deletion of TRAM attenuate atherosclerosis pathogenesis. iScience 2024; 27:110097. [PMID: 38883832 PMCID: PMC11179630 DOI: 10.1016/j.isci.2024.110097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 04/23/2024] [Accepted: 05/21/2024] [Indexed: 06/18/2024] Open
Abstract
Systemic neutrophil dysregulation contributes to atherosclerosis pathogenesis, and restoring neutrophil homeostasis may be beneficial for treating atherosclerosis. Herein, we report that a homeostatic resolving subset of neutrophils exists in mice and humans characterized by the low expression of TRAM, correlated with reduced expression of inflammatory mediators (leukotriene B4 [LTB4] and elastase) and elevated expression of anti-inflammatory resolving mediators (resolvin D1 [RvD1] and CD200R). TRAM-deficient neutrophils can potently improve vascular integrity and suppress atherosclerosis pathogenesis when adoptively transfused into recipient atherosclerotic animals. Mechanistically, we show that TRAM deficiency correlates with reduced expression of 5-lipoxygenase (LOX5) activating protein (LOX5AP), dislodges nuclear localization of LOX5, and switches the lipid mediator secretion from pro-inflammatory LTB4 to pro-resolving RvD1. TRAM also serves as a stress sensor of oxidized low-density lipoprotein (oxLDL) and/or free cholesterol and triggers inflammatory signaling processes that facilitate elastase release. Together, our study defines a unique neutrophil population characterized by reduced TRAM, capable of homeostatic resolution and treatment of atherosclerosis.
Collapse
Affiliation(s)
- Shuo Geng
- Department of Biological Sciences, Virginia Tech, Blacksburg VA 24061, USA
| | - Yao Zhang
- Department of Biological Sciences, Virginia Tech, Blacksburg VA 24061, USA
| | - Ran Lu
- Department of Biological Sciences, Virginia Tech, Blacksburg VA 24061, USA
| | - Daniel Irimia
- Center for Engineering in Medicine & Surgery, Massachusetts General Hospital, Harvard Medical School, Shriners Burns Hospital, Boston, MA 02114, USA
| | - Liwu Li
- Department of Biological Sciences, Virginia Tech, Blacksburg VA 24061, USA
| |
Collapse
|
32
|
Nhamoyebonde S, Chambers M, Ndlovu L, Karim F, Mazibuko M, Mhlane Z, Madziwa L, Moosa Y, Moodley S, Hoque M, Leslie A. Detailed phenotyping reveals diverse and highly skewed neutrophil subsets in both the blood and airways during active tuberculosis infection. Front Immunol 2024; 15:1422836. [PMID: 38947330 PMCID: PMC11212598 DOI: 10.3389/fimmu.2024.1422836] [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: 04/24/2024] [Accepted: 05/21/2024] [Indexed: 07/02/2024] Open
Abstract
Introduction Neutrophils play a complex and important role in the immunopathology of TB. Data suggest they are protective during early infection but become a main driver of immunopathology if infection progresses to active disease. Neutrophils are now recognized to exist in functionally diverse states, but little work has been done on how neutrophil states or subsets are skewed in TB disease. Methods To address this, we carried out comprehensive phenotyping by flow cytometry of neutrophils in the blood and airways of individuals with active pulmonary TB with and without HIV co-infection recruited in Durban, South Africa. Results Active TB was associated with a profound skewing of neutrophils in the blood toward phenotypes associated with activation and apoptosis, reduced phagocytosis, reverse transmigration, and immune regulation. This skewing was also apparently in airway neutrophils, particularly the regulatory subsets expressing PDL-1 and LOX-1. HIV co-infection did not impact neutrophil subsets in the blood but was associated with a phenotypic change in the airways and a reduction in key neutrophil functional proteins cathelicidin and arginase 1. Discussion Active TB is associated with profound skewing of blood and airway neutrophils and suggests multiple mechanisms by which neutrophils may exacerbate the immunopathology of TB. These data indicate potential avenues for reducing neutrophil-mediated lung pathology at the point of diagnosis.
Collapse
Affiliation(s)
| | - Mark Chambers
- Africa Health Research Institute, Durban, South Africa
| | - Lerato Ndlovu
- Africa Health Research Institute, Durban, South Africa
| | - Farina Karim
- Africa Health Research Institute, Durban, South Africa
| | | | - Zoey Mhlane
- Africa Health Research Institute, Durban, South Africa
| | | | - Yunus Moosa
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | | | - Monjurul Hoque
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Alasdair Leslie
- Africa Health Research Institute, Durban, South Africa
- Department of Infectious Diseases, Nelson R. Mandela School of Clinical Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Infection and Immunity, University College London, London, United Kingdom
| |
Collapse
|
33
|
Chen X, Yang F, Luo G. Identification of key regulatory genes in the pathogenesis of COVID-19 and sepsis: An observational study. Medicine (Baltimore) 2024; 103:e38378. [PMID: 39259097 PMCID: PMC11142772 DOI: 10.1097/md.0000000000038378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Revised: 03/05/2024] [Accepted: 05/06/2024] [Indexed: 09/12/2024] Open
Abstract
Patients with severe COVID-19 and those with sepsis have similar clinical manifestations. We used bioinformatics methods to identify the common hub genes in these 2 diseases. Two RNA-seq datasets from the Gene Expression Omnibus were used to identify common differentially expressed genes (DEGs) in COVID-19 and sepsis. These common genes were used for analysis of functional enrichment; pathway analysis; identification of associated transcription factors, metabolites, and miRNAs; and mapping of protein-protein interaction networks. The major hub genes of COVID-19 and sepsis were identified, and validation datasets were used to assess the value of these hub genes using receiver operating characteristic (ROC) curves. Analysis of the 800 common DEGs for COVID-19 and sepsis, as well as common transcription factors, miRNAs, and metabolites, demonstrated that the immune response had a key role in both diseases. DLGAP5, BUB1, CDK1, CCNB1, and BUB1B were the most important common hub genes. Analysis of a validation cohort indicated these 5 genes had significantly higher expression in COVID-19 patients and sepsis patients than in corresponding controls, and the area under the ROC curves ranged from 0.832 to 0.981 for COVID-19 and 0.840 to 0.930 for sepsis. We used bioinformatics tools to identify common DEGs, miRNAs, and transcription factors for COVID-19 and sepsis. The 5 identified hub genes had higher expression in validation cohorts of COVID-19 and sepsis. These genes had good or excellent diagnostic performance based on ROC analysis, and therefore have potential use as novel markers or therapeutic targets.
Collapse
Affiliation(s)
- Xing Chen
- Department of Infection, Nanchong Central Hospital, Nanchong, Sichuan, China
| | - Fengbo Yang
- Department of Otolaryngology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, China
| | - Guoping Luo
- Department of Infection, Nanchong Central Hospital, Nanchong, Sichuan, China
| |
Collapse
|
34
|
Armignacco R, Carlier N, Jouinot A, Birtolo MF, de Murat D, Tubach F, Hausfater P, Simon T, Gorochov G, Pourcher V, Beurton A, Goulet H, Manivet P, Bertherat J, Assié G. Whole blood transcriptome signature predicts severe forms of COVID-19: Results from the COVIDeF cohort study. Funct Integr Genomics 2024; 24:107. [PMID: 38772950 PMCID: PMC11108918 DOI: 10.1007/s10142-024-01359-2] [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: 02/26/2024] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/23/2024]
Abstract
COVID-19 is associated with heterogeneous outcome. Early identification of a severe progression of the disease is essential to properly manage the patients and improve their outcome. Biomarkers reflecting an increased inflammatory response, as well as individual features including advanced age, male gender, and pre-existing comorbidities, are risk factors of severe COVID-19. Yet, these features show limited accuracy for outcome prediction. The aim was to evaluate the prognostic value of whole blood transcriptome at an early stage of the disease. Blood transcriptome of patients with mild pneumonia was profiled. Patients with subsequent severe COVID-19 were compared to those with favourable outcome, and a molecular predictor based on gene expression was built. Unsupervised classification discriminated patients who would later develop a COVID-19-related severe pneumonia. The corresponding gene expression signature reflected the immune response to the viral infection dominated by a prominent type I interferon, with IFI27 among the most over-expressed genes. A 48-genes transcriptome signature predicting the risk of severe COVID-19 was built on a training cohort, then validated on an external independent cohort, showing an accuracy of 81% for predicting severe outcome. These results identify an early transcriptome signature of severe COVID-19 pneumonia, with a possible relevance to improve COVID-19 patient management.
Collapse
Affiliation(s)
- Roberta Armignacco
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France.
| | - Nicolas Carlier
- Service de Pneumologie, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - Anne Jouinot
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, 75014, Paris, France
| | | | - Daniel de Murat
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France
| | - Florence Tubach
- Sorbonne Université, INSERM, Institut Pierre Louis d'Epidémiologie Et de Santé Publique, AP-HP, 1901, F-75013, Paris, France
| | - Pierre Hausfater
- Emergency Department, APHP-Sorbonne Université, Hôpital Pitié-Salpêtrière, GRC-14 BIOSFAST, CIMI, UMR 1135, Sorbonne Université, Paris, France
| | - Tabassome Simon
- Service de Pharmacologie, Plateforme de Recherche Clinique URC-CRC-CRB de L'Est Parisien, Assistance Publique-Hôpitaux de Paris, Hôpital Saint Antoine, Sorbonne Université, Paris, France
| | - Guy Gorochov
- Centre d'Immunologie Et Des Maladies Infectieuses (CIMI), Department of Immunology, Sorbonne Université, Inserm, Hôpital Pitié Salpêtrière, Groupe Hospitalo-Universitaire Assistance Publique - Hôpitaux de Paris, Paris, France
| | - Valérie Pourcher
- Department of Infectious Diseases, Hôpital Pitié Salpêtrière, Groupe Hospitalo-Universitaire Assistance Publique - Hôpitaux de Paris, Sorbonne Université, Paris, France
| | - Alexandra Beurton
- Service de Médecine Intensive Réanimation EOLE - Département R3S - Sorbonne, Université - Hôpital Universitaire Pitié - Salpêtrière - Assistance Publique Hôpitaux de Paris - 83 Boulevard de L'Hôpital, 75013, Paris, France
- UMRS 1158 Inserm-Sorbonne Université "Neurophysiologie Respiratoire Expérimentale Et Clinique'' Intensive Care Unit, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Hélène Goulet
- Emergency Department, Hôpital Tenon, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Philippe Manivet
- INSERM UMR 1141 "NeuroDiderot", Université Paris Cité, FHU I2-D2, Paris, France
- AP-HP, DMU BioGem, Centre de Ressources Biologiques Biobank Lariboisière/Saint Louis (BB-0033-00064), Hôpital Lariboisière, Paris, France
| | - Jérôme Bertherat
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France
- Service d'Endocrinologie, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, 75014, Paris, France
| | - Guillaume Assié
- Université Paris Cité, CNRS UMR8104, INSERM U1016, Institut Cochin, F-75014, Paris, France.
- Service d'Endocrinologie, Center for Rare Adrenal Diseases, AP-HP, Hôpital Cochin, 75014, Paris, France.
| |
Collapse
|
35
|
Peronnet E, Terraz G, Cerrato E, Imhoff K, Blein S, Brengel-Pesce K, Bodinier M, Fleurie A, Rimmelé T, Lukaszewicz AC, Monneret G, Llitjos JF. Use of Immune Profiling Panel to assess the immune response of septic patients for prediction of worsening as a composite endpoint. Sci Rep 2024; 14:11305. [PMID: 38760488 PMCID: PMC11101454 DOI: 10.1038/s41598-024-62202-z] [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: 12/22/2023] [Accepted: 05/14/2024] [Indexed: 05/19/2024] Open
Abstract
Sepsis induces intense, dynamic and heterogeneous host response modulations. Despite improvement of patient management, the risk of mortality and healthcare-associated infections remains high. Treatments to counterbalance immune response are under evaluation, but effective biomarkers are still lacking to perform patient stratification. The design of the present study was defined to alleviate the limitations of existing literature: we selected patients who survived the initial hyperinflammatory response and are still hospitalized at day 5-7 after ICU admission. Using the Immune Profiling Panel (IPP), a fully automated RT-qPCR multiplex prototype, we optimized a machine learning model combining the IPP gene expression levels for the identification of patients at high risk of worsening, a composite endpoint defined as death or secondary infection, within one week after sampling. This was done on 332 sepsis patients selected from two retrospective studies. The IPP model identified a high-risk group comprising 30% of patients, with a significant increased proportion of worsening events at day 28 compared to the low-risk group (49% vs. 28%, respectively). These preliminary results underline the potential clinical application of IPP for sepsis patient stratification in a personalized medicine perspective, that will be confirmed in a larger prospective multicenter study.
Collapse
Affiliation(s)
- Estelle Peronnet
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France.
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy-l'Etoile, France.
| | - Gabriel Terraz
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- EFOR, Champagne-au-Mont-d'Or, France
| | - Elisabeth Cerrato
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy-l'Etoile, France
| | - Katia Imhoff
- Data Science, bioMérieux S.A., Marcy l'Etoile, France
| | - Sophie Blein
- Data Science, bioMérieux S.A., Marcy l'Etoile, France
| | - Karen Brengel-Pesce
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy-l'Etoile, France
| | - Maxime Bodinier
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy-l'Etoile, France
| | - Aurore Fleurie
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy-l'Etoile, France
| | - Thomas Rimmelé
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Anaesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Anne-Claire Lukaszewicz
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Anaesthesia and Critical Care Medicine Department, Hospices Civils de Lyon, Edouard Herriot Hospital, Lyon, France
| | - Guillaume Monneret
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Immunology Laboratory, Edouard Herriot Hospital - Hospices Civils de Lyon, Lyon, France
| | - Jean-François Llitjos
- Joint Research Unit HCL-bioMérieux, EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1 - Hospices Civils de Lyon, bioMérieux), Lyon, France
- Open Innovation and Partnerships (OI&P), bioMérieux S.A., Marcy-l'Etoile, France
| |
Collapse
|
36
|
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
|
37
|
Tomasicchio M, Jaumdally S, Wilson L, Kotze A, Semple L, Meier S, Pooran A, Esmail A, Pillay K, Roberts R, Kriel R, Meldau R, Oelofse S, Mandviwala C, Burns J, Londt R, Davids M, van der Merwe C, Roomaney A, Kühn L, Perumal T, Scott AJ, Hale MJ, Baillie V, Mahtab S, Williamson C, Joseph R, Sigal A, Joubert I, Piercy J, Thomson D, Fredericks DL, Miller MGA, Nunes MC, Madhi SA, Dheda K. SARS-CoV-2 Viral Replication Persists in the Human Lung for Several Weeks after Symptom Onset. Am J Respir Crit Care Med 2024; 209:840-851. [PMID: 38226855 PMCID: PMC10995573 DOI: 10.1164/rccm.202308-1438oc] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/12/2024] [Indexed: 01/17/2024] Open
Abstract
Rationale: In the upper respiratory tract, replicating (culturable) severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is recoverable for ∼4-8 days after symptom onset, but there is a paucity of data about the frequency and duration of replicating virus in the lower respiratory tract (i.e., the human lung).Objectives: We undertook lung tissue sampling (needle biopsy) shortly after death in 42 mechanically ventilated decedents during the Beta and Delta waves. An independent group of 18 ambulatory patients served as a control group.Methods: Lung biopsy cores from decedents underwent viral culture, histopathological analysis, electron microscopy, transcriptomic profiling, and immunohistochemistry.Measurements and Main Results: Thirty-eight percent (16 of 42) of mechanically ventilated decedents had culturable virus in the lung for a median of 15 days (persisting for up to 4 wk) after symptom onset. Lung viral culture positivity was not associated with comorbidities or steroid use. Delta but not Beta variant lung culture positivity was associated with accelerated death and secondary bacterial infection (P < 0.05). Nasopharyngeal culture was negative in 23.1% (6 of 26) of decedents despite lung culture positivity. This hitherto undescribed biophenotype of lung-specific persisting viral replication was associated with an enhanced transcriptomic pulmonary proinflammatory response but with concurrent viral culture positivity.Conclusions: Concurrent rather than sequential active viral replication continues to drive a heightened proinflammatory response in the human lung beyond the second week of illness and was associated with variant-specific increased mortality and morbidity. These findings have potential implications for the design of interventional strategies and clinical management of patients with severe coronavirus disease (COVID-19).
Collapse
Affiliation(s)
- Michele Tomasicchio
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Shameem Jaumdally
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Lindsay Wilson
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Andrea Kotze
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Lynn Semple
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Stuart Meier
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Anil Pooran
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Aliasgar Esmail
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Komala Pillay
- Division of Anatomical Pathology, Department of Pathology, and
| | - Riyaadh Roberts
- Division of Anatomical Pathology, Department of Pathology, and
| | - Raymond Kriel
- Division of Anatomical Pathology, Department of Pathology, and
| | - Richard Meldau
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Suzette Oelofse
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Carley Mandviwala
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Jessica Burns
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Rolanda Londt
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Malika Davids
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Charnay van der Merwe
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Aqeedah Roomaney
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Louié Kühn
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Tahlia Perumal
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | - Alex J. Scott
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
| | | | - Vicky Baillie
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Sana Mahtab
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | | | | | - Alex Sigal
- Africa Health Research Institute, Durban, South Africa; and
| | - Ivan Joubert
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - Jenna Piercy
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - David Thomson
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - David L. Fredericks
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - Malcolm G. A. Miller
- Division of Critical Care, Department of Anaesthesia and Perioperative Medicine, University of Cape Town, Cape Town, South Africa
| | - Marta C. Nunes
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Centre of Excellence in Respiratory Pathogens, Hospices Civils de Lyon and Centre International de Recherche en Infectiologie, Équipe Santé Publique, Épidémiologie et Écologie Évolutive des Maladies Infectieuses, Inserm U1111, CNRS UMR5308, ENS de Lyon, Université Claude Bernard – Lyon 1, Lyon, France
| | - Shabir A. Madhi
- South African Medical Research Council, Vaccines and Infectious Diseases Analytics Research Unit, and
- Department of Science and Technology/National Research Foundation South African Research Chair Initiative in Vaccine Preventable Diseases, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Keertan Dheda
- Centre for Lung Infection and Immunity, Division of Pulmonology, Department of Medicine, University of Cape Town and UCT Lung Institute, Cape Town, South Africa
- South African MRC Centre for the Study of Antimicrobial Resistance
- Institute of Infectious Disease and Molecular Medicine
- Faculty of Infectious and Tropical Diseases, Department of Immunology and Infection, London School of Hygiene & Tropical Medicine, London, United Kingdom
| |
Collapse
|
38
|
Ullah MA, Moin AT, Nipa JF, Islam NN, Johora FT, Chowdhury RH, Islam S. Exploring risk factors and molecular targets in leukemia patients with COVID-19: a bioinformatics analysis of differential gene expression. J Leukoc Biol 2024; 115:723-737. [PMID: 38323674 DOI: 10.1093/jleuko/qiae002] [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: 09/24/2023] [Revised: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 02/08/2024] Open
Abstract
The molecular mechanism of COVID-19's pathogenic effects in leukemia patients is still poorly known. Our study investigated the possible disease mechanism of COVID-19 and its associated risk factors in patients with leukemia utilizing differential gene expression analysis. We also employed network-based approaches to identify molecular targets that could potentially diagnose and treat COVID-19-infected leukemia patients. Our study demonstrated a shared set of 60 genes that are expressed differentially among patients with leukemia and COVID-19. Most of these genes are expressed in blood and bone marrow tissues and are predominantly implicated in the pathogenesis of different hematologic malignancies, increasingly imperiling COVID-19 morbidity and mortality among the affected patients. Additionally, we also found that COVID-19 may influence the expression of several cancer-associated genes in leukemia patients, such as CCR7, LEF1, and 13 candidate cancer-driver genes. Furthermore, our findings reveal that COVID-19 may predispose leukemia patients to altered blood homeostasis, increase the risk of COVID-19-related liver injury, and deteriorate leukemia-associated injury and patient prognosis. Our findings imply that molecular signatures, like transcription factors, proteins such as TOP21, and 25 different microRNAs, may be potential targets for diagnosing and treating COVID-19-infected leukemia patients. Nevertheless, additional experimental studies will contribute to further validating the study's findings.
Collapse
Affiliation(s)
- Md Asad Ullah
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Hathazari, Chattogram-4331, Bangladesh
| | - Jannatul Ferdous Nipa
- Department of Genetic Engineering and Biotechnology, East West University, Aftabnagar, Dhaka-1212, Bangladesh
| | - Nafisa Nawal Islam
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Fatema Tuz Johora
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Rahee Hasan Chowdhury
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Hathazari, Chattogram-4331, Bangladesh
| | - Saiful Islam
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram Laboratories, Chittagong Cantonment, Chattogram-4220, Bangladesh
| |
Collapse
|
39
|
Kaiser R, Gold C, Joppich M, Loew Q, Akhalkatsi A, Mueller TT, Offensperger F, Droste Zu Senden A, Popp O, di Fina L, Knottenberg V, Martinez-Navarro A, Eivers L, Anjum A, Escaig R, Bruns N, Briem E, Dewender R, Muraly A, Akgöl S, Ferraro B, Hoeflinger JKL, Polewka V, Khaled NB, Allgeier J, Tiedt S, Dichgans M, Engelmann B, Enard W, Mertins P, Hubner N, Weckbach L, Zimmer R, Massberg S, Stark K, Nicolai L, Pekayvaz K. Peripheral priming induces plastic transcriptomic and proteomic responses in circulating neutrophils required for pathogen containment. SCIENCE ADVANCES 2024; 10:eadl1710. [PMID: 38517968 DOI: 10.1126/sciadv.adl1710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 02/16/2024] [Indexed: 03/24/2024]
Abstract
Neutrophils rapidly respond to inflammation and infection, but to which degree their functional trajectories after mobilization from the bone marrow are shaped within the circulation remains vague. Experimental limitations have so far hampered neutrophil research in human disease. Here, using innovative fixation and single-cell-based toolsets, we profile human and murine neutrophil transcriptomes and proteomes during steady state and bacterial infection. We find that peripheral priming of circulating neutrophils leads to dynamic shifts dominated by conserved up-regulation of antimicrobial genes across neutrophil substates, facilitating pathogen containment. We show the TLR4/NF-κB signaling-dependent up-regulation of canonical neutrophil activation markers like CD177/NB-1 during acute inflammation, resulting in functional shifts in vivo. Blocking de novo RNA synthesis in circulating neutrophils abrogates these plastic shifts and prevents the adaptation of antibacterial neutrophil programs by up-regulation of distinct effector molecules upon infection. These data underline transcriptional plasticity as a relevant mechanism of functional neutrophil reprogramming during acute infection to foster bacterial containment within the circulation.
Collapse
Affiliation(s)
- Rainer Kaiser
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Christoph Gold
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Markus Joppich
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Quentin Loew
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
| | | | - Tonina T Mueller
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Vascular Biology and Pathology, Institute of Laboratory Medicine, University Hospital Ludwig-Maximilians University, Munich, Germany
| | - Felix Offensperger
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | | | - Oliver Popp
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Lea di Fina
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | | | | | - Luke Eivers
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
| | - Afra Anjum
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Raphael Escaig
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Nils Bruns
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Eva Briem
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Robin Dewender
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
| | - Abhinaya Muraly
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
| | - Sezer Akgöl
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Bartolo Ferraro
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig Maximilian University Munich, Planegg-Martinsried, Germany
| | - Jonathan K L Hoeflinger
- Vascular Biology and Pathology, Institute of Laboratory Medicine, University Hospital Ludwig-Maximilians University, Munich, Germany
| | - Vivien Polewka
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
| | - Najib Ben Khaled
- Medizinische Klinik und Poliklinik II, University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Julian Allgeier
- Medizinische Klinik und Poliklinik II, University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Steffen Tiedt
- Institute for Stroke and Dementia Research, University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Martin Dichgans
- Institute for Stroke and Dementia Research, University Hospital Ludwig-Maximilian University, Munich, Germany
| | - Bernd Engelmann
- Vascular Biology and Pathology, Institute of Laboratory Medicine, University Hospital Ludwig-Maximilians University, Munich, Germany
| | - Wolfgang Enard
- Anthropology and Human Genomics, Faculty of Biology, Ludwig-Maximilians-Universität, Munich, Germany
| | - Philipp Mertins
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
| | - Norbert Hubner
- Max Delbrück Center for Molecular Medicine (MDC) and Berlin Institute of Health (BIH), Berlin, Germany
- Charite-Universitätsmedizin Berlin, Berlin, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Ludwig Weckbach
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
- Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, Ludwig Maximilian University Munich, Planegg-Martinsried, Germany
| | - Ralf Zimmer
- LFE Bioinformatik, Department of Informatics, Ludwig-Maximilians-Universität München, Munich, Germany
| | - Steffen Massberg
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Konstantin Stark
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Leo Nicolai
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| | - Kami Pekayvaz
- Department of Medicine I, LMU University Hospital, LMU Munich, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany
| |
Collapse
|
40
|
Huang Z, Cheng Z, Deng X, Yang Y, Sun N, Hou P, Fan R, Liu S. Integrated Bioinformatics Exploration and Preliminary Clinical Verification for the Identification of Crucial Biomarkers in Severe Cases of COVID-19. J Inflamm Res 2024; 17:1561-1576. [PMID: 38495341 PMCID: PMC10942013 DOI: 10.2147/jir.s454284] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/29/2024] [Indexed: 03/19/2024] Open
Abstract
Background Coronavirus disease 2019 (COVID-19) is a respiratory infectious illness caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The objective of this study is to identify reliable and accurate biomarkers for the early stratification of disease severity, a crucial aspect that is currently lacking for the impending phases of the next COVID-19 pandemic. Methods In this study, we identified important module and hub genes related to clinical severe COVID-19 using differentially expressed genes (DEGs) screening combing weighted gene co-expression network analysis (WGCNA) in dataset GSE213313. We further screened and confirmed these hub genes in another two new independent datasets (GSE172114 and GSE157103). In order to evaluate these key genes' stability and robustness for diagnosing or predicting the progression of illness, we used RT-PCR validation of selected genes in blood samples obtained from hospitalized COVID-19 patients. Results A total of 968 and 52 DEGs were identified between COVID-19 patients and normal people, critical and non-critical patients, respectively. Then, using WGCNA, 10 modules were constructed. Among them, the blue module positively associated with clinic disease severity of COVID-19. From overlapped section between DEGs and blue module, 12 intersected common differential genes were obtained. Subsequently, these hub genes were validated in another two new independent datasets as well and 9 genes that overlapped showed a highly correlation with disease severity. Finally, the mRNA expression levels of these hub genes were tested in blood samples from COVID-19 patients. In severe cases, there was increased expression of MCEMP1, ANXA3, CD177, and SCN9A. In particular, MCEMP1 increased with disease severity, which suggested an unfavorable development and a frustrating prognosis. Conclusion Using comprehensive bioinformatical analysis and the validation of clinical samples, we identified four major candidate genes, MCEMP1, ANXA3, CD177, and SCN9A, which are essential for diagnosis or development of COVID-19.
Collapse
Affiliation(s)
- Zhisheng Huang
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, People’s Republic of China
- Department of Pulmonary and Critical Care Medicine, National Regional Center for Respiratory Medicine, Jiangxi Hospital of China-Japan Friendship Hospital, Nanchang, Jiangxi, People’s Republic of China
| | - Zuowang Cheng
- Department of Clinical Laboratory, Zhangqiu District People’s Hospital Affiliated to Jining Medical University, Jinan, Shandong, People’s Republic of China
| | - Xia Deng
- School of Public Health, Shandong Second Medical University, Weifang, Shandong, People’s Republic of China
| | - Ying Yang
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, People’s Republic of China
| | - Na Sun
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, People’s Republic of China
| | - Peibin Hou
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, People’s Republic of China
| | - Ruyue Fan
- Shandong Center for Disease Control and Prevention, Jinan, Shandong, People’s Republic of China
| | - Shuai Liu
- Department of Respiratory and Critical Care Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, People’s Republic of China
- Shandong Key Laboratory of Infectious Respiratory Disease, Jinan, Shandong, People’s Republic of China
| |
Collapse
|
41
|
Karabekmez ME, Yarıcı M. Parameterization of asymmetric sigmoid functions in weighted gene co-expression network analysis. Comput Biol Chem 2024; 108:107998. [PMID: 38071762 DOI: 10.1016/j.compbiolchem.2023.107998] [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: 02/22/2023] [Revised: 11/22/2023] [Accepted: 12/03/2023] [Indexed: 01/22/2024]
Abstract
In most the biological contexts, examining gene expressions at the genomic level gives more accurate results than examining genes individually. It can improve understanding of the molecular mechanisms that cause molecular alterations. Weighted gene co-expression network analysis (WGCNA), which has recently been widely used to cluster transcriptomic datasets, implements a soft thresholding procedure using power function. However, these functions may sometimes exaggerate minor differences in expression correlations. We have previously proposed to use asymmetric sigmoid functions in soft thresholding as an alternative solution. However, the number of variables in asymmetric sigmoid functions may vary and parameterization can be problematic. In this study, we have introduced a systematic procedure for parameterizing asymmetric sigmoid function to ease using it as an alternative soft-thresholding solution in WGCNA. The efficiency of the employment was shown on four different COVID-19 datasets, on a yeast dataset, and on an E.Coli dataset. The results indicate that this approach provides biologically plausible associations for the resulting modules.
Collapse
Affiliation(s)
| | - Merve Yarıcı
- Istanbul Medeniyet University, Department of Bioengineering, Istanbul, Turkey
| |
Collapse
|
42
|
Zhuang Q, Zhang R, Li X, Ma D, Wang Y. Identification of the shared molecular mechanisms between major depressive disorder and COVID-19 from postmortem brain transcriptome analysis. J Affect Disord 2024; 346:273-284. [PMID: 37956829 DOI: 10.1016/j.jad.2023.11.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/21/2023] [Accepted: 11/09/2023] [Indexed: 11/15/2023]
Abstract
OBJECTIVES This study aims to investigate the molecular mechanisms underlying the interaction of major depressive disorder (MDD) and COVID-19, and on this basis, diagnostic biomarkers and potential therapeutic drugs are further explored. METHODS Differential gene expression analysis and weighted gene co-expression network analysis (WGCNA) were employed to identify common key genes involved in the pathogenesis of COVID-19 and MDD. Correlations with clinical features were explored. Detailed mechanisms were further investigated through protein interaction networks, GSEA, and immune cell infiltration analysis. Finally, Enrichr's Drug Signature Database and Coremine Medical were used to predict the potential drugs associated with key genes. RESULTS The study identified 18 genes involved in both COVID-19 and MDD. Four key genes (MBP, CYP4B1, ERMN, and SLC26A7) were selected based on clinical relevance. A multi-gene prediction model showed good diagnostic efficiency for the two diseases: AUC of 0.852 for COVID-19 and 0.915 for MDD. GO and GSEA analyses identified specific biological functions and pathways associated with key genes in COVID-19 (axon guidance, metabolism, stress response) and MDD (neuron ensheathment, biosynthesis, glutamatergic neuron differentiation). The key genes also affected immune infiltration. Potential therapeutic drugs, including small molecules and traditional Chinese medicines, targeting these genes were identified. CONCLUSION This study provides insights into the complex biological mechanisms underlying COVID-19 and MDD, develops an effective diagnostic model, and predicts potential therapeutic drugs, which may contribute to the prevention and treatment of these two prevalent diseases.
Collapse
Affiliation(s)
- Qishuai Zhuang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China
| | - Rongqing Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, China
| | - Xiaobing Li
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, China
| | - Dapeng Ma
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, China
| | - Yue Wang
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Neurosurgery, Jinan 250014, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, China.
| |
Collapse
|
43
|
Lei H. Hypoxia and Activation of Neutrophil Degranulation-Related Genes in the Peripheral Blood of COVID-19 Patients. Viruses 2024; 16:201. [PMID: 38399976 PMCID: PMC10891603 DOI: 10.3390/v16020201] [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: 01/17/2024] [Revised: 01/20/2024] [Accepted: 01/21/2024] [Indexed: 02/25/2024] Open
Abstract
Severe COVID-19 is characterized by systematic hyper-inflammation and subsequent damage to various organs. Therefore, it is critical to trace this cascade of hyper-inflammation. Blood transcriptome has been routinely utilized in the interrogation of host immune response in COVID-19 and other infectious conditions. In this study, consensus gene dysregulation in the blood was obtained from 13 independent transcriptome studies on COVID-19. Among the up-regulated genes, the most prominent functional categories were neutrophil degranulation and cell cycle, which is clearly different from the classical activation of interferon signaling pathway in seasonal flu. As for the potential upstream causal factors of the atypical gene dysregulation, systemic hypoxia was further examined because it is much more widely reported in COVID-19 than that in seasonal flu. It was found that both physiological and pathological hypoxia can induce activation of neutrophil degranulation-related genes in the blood. Furthermore, COVID-19 patients with different requirement for oxygen intervention showed distinctive levels of gene expression related to neutrophil degranulation in the whole blood, which was validated in isolated neutrophils. Thus, activation of neutrophil degranulation-related genes in the blood of COVID-19 could be partially attributed to hypoxia. Interestingly, similar pattern was also observed in H1N1 infection (the cause of Spanish flu) and several other severe respiratory viral infections. As for the molecular mechanism, both HIF-dependent and HIF-independent pathways have been examined. Since the activation of neutrophil degranulation-related genes is highly correlated with disease severity in COVID-19, early detection of hypoxia and active intervention may prevent further activation of neutrophil degranulation-related genes and other harmful downstream hyper-inflammation. This common mechanism is applicable to current and future pandemic as well as the severe form of common respiratory infection.
Collapse
Affiliation(s)
- Hongxing Lei
- CAS Key Laboratory of Genome Sciences and Information, Beijing Institute of Genomics, Chinese Academy of Sciences, and China National Center for Bioinformation, Beijing 100101, China; ; Tel.: +86-010-84097276
- Cunji Medical School, University of Chinese Academy of Sciences, Beijing 101408, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing 100069, China
| |
Collapse
|
44
|
Huang B, Huang J, Chiang NH, Chen Z, Lui G, Ling L, Kwan MYW, Wong JSC, Mak PQ, Ling JWH, Lam ICS, Ng RWY, Wang X, Gao R, Hui DSC, Ma SL, Chan PKS, Tang NLS. Interferon response and profiling of interferon response genes in peripheral blood of vaccine-naive COVID-19 patients. Front Immunol 2024; 14:1315602. [PMID: 38268924 PMCID: PMC10806211 DOI: 10.3389/fimmu.2023.1315602] [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: 10/10/2023] [Accepted: 12/18/2023] [Indexed: 01/26/2024] Open
Abstract
Introduction There is insufficient understanding on systemic interferon (IFN) responses during COVID-19 infection. Early reports indicated that interferon responses were suppressed by the coronavirus (SARS-CoV-2) and clinical trials of administration of various kinds of interferons had been disappointing. Expression of interferon-stimulated genes (ISGs) in peripheral blood (better known as interferon score) has been a well-established bioassay marker of systemic IFN responses in autoimmune diseases. Therefore, with archival samples of a cohort of COVID-19 patients collected before the availability of vaccination, we aimed to better understand this innate immune response by studying the IFN score and related ISGs expression in bulk and single cell RNAs sequencing expression datasets. Methods In this study, we recruited 105 patients with COVID-19 and 30 healthy controls in Hong Kong. Clinical risk factors, disease course, and blood sampling times were recovered. Based on a set of five commonly used ISGs (IFIT1, IFIT2, IFI27, SIGLEC1, IFI44L), the IFN score was determined in blood leukocytes collected within 10 days after onset. The analysis was confined to those blood samples collected within 10 days after disease onset. Additional public datasets of bulk gene and single cell RNA sequencing of blood samples were used for the validation of IFN score results. Results Compared to the healthy controls, we showed that ISGs expression and IFN score were significantly increased during the first 10 days after COVID infection in majority of patients (71%). Among those low IFN responders, they were more commonly asymptomatic patients (71% vs 25%). 22 patients did not mount an overall significant IFN response and were classified as low IFN responders (IFN score < 1). However, early IFN score or ISGs level was not a prognostic biomarker and could not predict subsequent disease severity. Both IFI27 and SIGLEC1 were monocyte-predominant expressing ISGs and IFI27 were activated even among those low IFN responders as defined by IFN score. In conclusion, a substantial IFN response was documented in this cohort of COVID-19 patients who experience a natural infection before the vaccination era. Like innate immunity towards other virus, the ISGs activation was observed largely during the early course of infection (before day 10). Single-cell RNA sequencing data suggested monocytes were the cell-type that primarily accounted for the activation of two highly responsive ISGs (IFI44L and IFI27). Discussion As sampling time and age were two major confounders of ISG expression, they may account for contradicting observations among previous studies. On the other hand, the IFN score was not associated with the severity of the disease.
Collapse
Affiliation(s)
- Baozhen Huang
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Jinghan Huang
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nim Hang Chiang
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Zigui Chen
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Grace Lui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Lowell Ling
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Mike Yat Wah Kwan
- Paediatric Infectious Disease Unit, Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Joshua Sung Chih Wong
- Paediatric Infectious Disease Unit, Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Phoebe Qiaozhen Mak
- Paediatric Infectious Disease Unit, Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Janet Wan Hei Ling
- Paediatric Infectious Disease Unit, Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Ivan Cheuk San Lam
- Paediatric Infectious Disease Unit, Department of Paediatrics and Adolescent Medicine, Princess Margaret Hospital, Hong Kong, Hong Kong SAR, China
| | - Rita Wai Yin Ng
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Xingyan Wang
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Ruonan Gao
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - David Shu-Cheong Hui
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Suk Ling Ma
- Department of Psychiatry, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Paul K. S. Chan
- Department of Microbiology, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Nelson Leung Sang Tang
- Department of Chemical Pathology, and Li Ka Shing Institute of Health Science, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
- Hong Kong Branch of CAS Center for Excellence in Animal Evolution and Genetics and KIZ/CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, Hong Kong, Hong Kong SAR, China
- Functional Genomics and Biostatistical Computing Laboratory, CUHK Shenzhen Research Institute, Shenzhen, China
| |
Collapse
|
45
|
Liu H, Wang J, Li S, Sun Y, Zhang P, Ma J. The unfolded protein response pathway as a possible link in the pathogenesis of COVID-19 and sepsis. Arch Virol 2024; 169:20. [PMID: 38191819 DOI: 10.1007/s00705-023-05948-7] [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: 09/22/2023] [Accepted: 11/10/2023] [Indexed: 01/10/2024]
Abstract
The global impact of the COVID-19 pandemic has been substantial. Emerging evidence underscores a strong clinical connection between COVID-19 and sepsis. Numerous studies have identified the unfolded protein response (UPR) pathway as a crucial pathogenic pathway for both COVID-19 and sepsis, but it remains to be investigated whether this signaling pathway operates as a common pathogenic mechanism for both COVID-19 and sepsis. In this study, single-cell RNA-seq data and transcriptome data for COVID-19 and sepsis cases were downloaded from GEO (Gene Expression Omnibus). By analyzing the single-cell transcriptome data, we identified B cells as the critical cell subset and the UPR pathway as the critical signaling pathway. Based on the transcriptome data, a machine learning diagnostic model was then constructed using the interleaved genes of B-cell-related and UPR-pathway-related genes. We validated the diagnostic model using both internal and external datasets and found the accuracy and stability of this model to be extremely strong. Even after integrating our algorithmic model with the patient's clinical status, it continued to yield identical results, further emphasizing the reliability of this model. This study provides a novel molecular perspective on the pathogenesis of sepsis and COVID-19 at the single-cell level and suggests that these two diseases may share a common mechanism.
Collapse
Affiliation(s)
- Hong Liu
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Junyi Wang
- Advanced Medical Research Institute, Shandong University, Jinan, Shandong, China
| | - Shaofeng Li
- School of Pharmacy, Key Laboratory of Nano-carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, China
| | - Yanmei Sun
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Peng Zhang
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jiahao Ma
- School of Pharmacy, Key Laboratory of Nano-carbon Modified Film Technology of Henan Province, Diagnostic Laboratory of Animal Diseases, Xinxiang University, Xinxiang, China.
| |
Collapse
|
46
|
Diddens J, Lepennetier G, Friedrich V, Schmidt M, Brand RM, Georgieva T, Hemmer B, Lehmann-Horn K. Single-Cell Profiling Indicates a Proinflammatory Role of Meningeal Ectopic Lymphoid Tissue in Experimental Autoimmune Encephalomyelitis. NEUROLOGY(R) NEUROIMMUNOLOGY & NEUROINFLAMMATION 2024; 11:e200185. [PMID: 38100739 PMCID: PMC10723639 DOI: 10.1212/nxi.0000000000200185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Accepted: 09/28/2023] [Indexed: 12/17/2023]
Abstract
BACKGROUND AND OBJECTIVES The factors that drive progression in multiple sclerosis (MS) remain obscure. Identification of key properties of meningeal inflammation will contribute to a better understanding of the mechanisms of progression and how to prevent it. METHODS Applying single-cell RNA sequencing, we compared gene expression profiles in immune cells from meningeal ectopic lymphoid tissue (mELT) with those from secondary lymphoid organs (SLOs) in spontaneous chronic experimental autoimmune encephalomyelitis (EAE), an animal model of MS. RESULTS Generally, mELT contained the same immune cell types as SLOs, suggesting a close relationship. Preponderance of B cells over T cells, an increase in regulatory T cells and granulocytes, and a decrease in naïve CD4+ T cells characterize mELT compared with SLOs. Differential gene expression analysis revealed that immune cells in mELT show a more activated and proinflammatory phenotype compared with their counterparts in SLOs. However, the increase in regulatory T cells and upregulation of immunosuppressive genes in most immune cell types indicate that there are mechanisms in place to counter-regulate the inflammatory events, keeping the immune response emanating from mELT in check. DISCUSSION Common features in immune cell composition and gene expression indicate that mELT resembles SLOs and may be regarded as a tertiary lymphoid tissue. Distinct differences in expression profiles suggest that mELT rather than SLOs is a key driver of CNS inflammation in spontaneous EAE. Our data provide a starting point for further exploration of molecules or pathways that could be targeted to disrupt mELT formation.
Collapse
Affiliation(s)
- Jolien Diddens
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Gildas Lepennetier
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Verena Friedrich
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Monika Schmidt
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Rosa M Brand
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Tanya Georgieva
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Bernhard Hemmer
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| | - Klaus Lehmann-Horn
- From the Department of Neurology (J.D., G.L., V.F., M.S., R.M.B., T.G., B.H., K.L.-H.), School of Medicine, Technical University of Munich; and Munich Cluster of Systems Neurology (SyNergy) (B.H.), Germany
| |
Collapse
|
47
|
Hafkamp FMJ, Taanman-Kueter EWM, van Capel TMM, Wynberg E, van Willigen HDG, Verveen A, Kootstra NA, Nieuwkerk P, de Jong MD, de Bree GJ, Prins M, Hazenberg MD, Groot Kormelink T, de Jong EC. Aberrant neutrophil degranulation in hospitalized patients with COVID-19 partially remains for 6 months. Eur J Immunol 2024; 54:e2350404. [PMID: 37853954 DOI: 10.1002/eji.202350404] [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: 01/24/2023] [Revised: 10/14/2023] [Accepted: 10/17/2023] [Indexed: 10/20/2023]
Abstract
Neutrophils are important players in COVID-19, contributing to tissue damage by release of inflammatory mediators, including ROS and neutrophil elastase. Longitudinal studies on the effects of COVID-19 on neutrophil phenotype and function are scarce. Here, we longitudinally investigated the phenotype and degranulation of neutrophils in COVID-19 patients (28 nonhospitalized and 35 hospitalized patients) compared with 17 healthy donors (HDs). We assessed phenotype, degranulation, CXCL8 (IL-8) release, and ROS generation within 8 days, at one or 6 month(s) after COVID-19 diagnosis. For degranulation and ROS production, we stimulated neutrophils, either with ssRNA and TNF or granulocyte-macrophage colony-stimulating factor and N-Formylmethionyl-leucyl-phenylalanine. During active COVID-19, neutrophils from hospitalized patients were more immature than from HDs and were impaired in degranulation and ROS generation, while neutrophils from nonhospitalized patients only demonstrated reduced CD66b+ granule release and ROS production. Baseline CD63 expression, indicative of primary granule release, and CXCL8 production by neutrophils from hospitalized patients were elevated for up to 6 months. These findings show that patients hospitalized due to COVID-19, but not nonhospitalized patients, demonstrated an aberrant neutrophil phenotype, degranulation, CXCL8 release, and ROS generation that partially persists up to 6 months after infection.
Collapse
Affiliation(s)
- Florianne M J Hafkamp
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Esther W M Taanman-Kueter
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Toni M M van Capel
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Elke Wynberg
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Hugo D G van Willigen
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anouk Verveen
- Department of Medical Psychology, Amsterdam UMC, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, the Netherlands
| | - Neeltje A Kootstra
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Pythia Nieuwkerk
- Department of Medical Psychology, Amsterdam UMC, Amsterdam Public Health Research Institute, University of Amsterdam, Amsterdam, the Netherlands
| | - Menno D de Jong
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Medical Microbiology & Infection Prevention, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Godelieve J de Bree
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Maria Prins
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Department of Infectious Diseases, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Mette D Hazenberg
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
- Department of Hematology, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Department of Hematopoiesis, Sanquin Research, Amsterdam, the Netherlands
| | - Tom Groot Kormelink
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| | - Esther C de Jong
- Department of Experimental Immunology, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, the Netherlands
- Amsterdam Institute for Infection and Immunity, Amsterdam, the Netherlands
| |
Collapse
|
48
|
Fan L, Hu Y, Gao L, Yu A, Zhang D, Wu Y, Yu F, Li L, Li B, Zhao H, Ma P. Association between increased CD177 + neutrophils and chronic activation in people living with HIV. Chin Med J (Engl) 2023; 136:2996-2998. [PMID: 38013464 PMCID: PMC10752480 DOI: 10.1097/cm9.0000000000002958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Indexed: 11/29/2023] Open
Affiliation(s)
- Lina Fan
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Yue Hu
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Liying Gao
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Aiping Yu
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Defa Zhang
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Yue Wu
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Fangfang Yu
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Lei Li
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| | - Bei Li
- Department of Infectious Diseases, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100015, China
| | - Hongxin Zhao
- Clinical and Research Center of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Ping Ma
- Department of Infectious Diseases, Tianjin Second People’s Hospital, Tianjin 300192, China
| |
Collapse
|
49
|
Merdler-Rabinowicz R, Gorelik D, Park J, Meydan C, Foox J, Karmon M, Roth H, Cohen-Fultheim R, Shohat-ophir G, Eisenberg E, Ruppin E, Mason C, Levanon E. Elevated A-to-I RNA editing in COVID-19 infected individuals. NAR Genom Bioinform 2023; 5:lqad092. [PMID: 37859800 PMCID: PMC10583280 DOI: 10.1093/nargab/lqad092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Revised: 08/29/2023] [Accepted: 09/29/2023] [Indexed: 10/21/2023] Open
Abstract
Given the current status of coronavirus disease 2019 (COVID-19) as a global pandemic, it is of high priority to gain a deeper understanding of the disease's development and how the virus impacts its host. Adenosine (A)-to-Inosine (I) RNA editing is a post-transcriptional modification, catalyzed by the ADAR family of enzymes, that can be considered part of the inherent cellular defense mechanism as it affects the innate immune response in a complex manner. It was previously reported that various viruses could interact with the host's ADAR enzymes, resulting in epigenetic changes both to the virus and the host. Here, we analyze RNA-seq of nasopharyngeal swab specimens as well as whole-blood samples of COVID-19 infected individuals and show a significant elevation in the global RNA editing activity in COVID-19 compared to healthy controls. We also detect specific coding sites that exhibit higher editing activity. We further show that the increment in editing activity during the disease is temporary and returns to baseline shortly after the symptomatic period. These significant epigenetic changes may contribute to the immune system response and affect adverse outcomes seen in post-viral cases.
Collapse
Affiliation(s)
- Rona Merdler-Rabinowicz
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, Israel
| | - David Gorelik
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, Israel
| | - Jiwoon Park
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY, USA
| | - Cem Meydan
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Jonathan Foox
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
| | - Miriam Karmon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, Israel
| | - Hillel S Roth
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, Israel
| | - Roni Cohen-Fultheim
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, Israel
| | - Galit Shohat-ophir
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- Leslie and Susan Gonda Multidisciplinary Brain Research Center and The Nanotechnology Institute, Bar-Ilan University, Ramat Gan, Israel
| | - Eli Eisenberg
- Raymond and Beverly Sackler School of Physics and Astronomy, Tel-Aviv University, Tel Aviv, Israel
| | - Eytan Ruppin
- Cancer Data Science Lab, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Christopher E Mason
- Department of Physiology, Biophysics and Systems Biology, Weill Cornell Medicine, New York, NY, USA
- The HRH Prince Alwaleed Bin Talal Bin Abdulaziz Alsaud Institute for Computational Biomedicine, Weill Cornell Medicine, New York, NY, USA
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY, USA
- The WorldQuant Initiative for Quantitative Prediction, Weill Cornell Medicine, New York, NY, USA
| | - Erez Y Levanon
- Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
- The Institute of Nanotechnology and Advanced Materials, Bar‐Ilan University, Ramat Gan, Israel
| |
Collapse
|
50
|
He W, Yu S, Li H, He P, Xiong T, Yan C, Zhang J, Chen S, Guo M, Tan X, Zhong D, Sun J, Xu Z, Cheng W, Li J. Comparison and Evaluation of Two Combination Modes of Angiotensin for Establishing Murine Aortic Dissection Models. J Cardiovasc Transl Res 2023; 16:1392-1407. [PMID: 37749480 DOI: 10.1007/s12265-023-10408-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 06/22/2023] [Indexed: 09/27/2023]
Abstract
Aortic dissection (AD) is a potentially fatal cardiovascular emergency caused by separation of different layers of aortic wall. However, because of limited time window available for clinical research, there is an urgent need for an ideal animal research model. In recent years, the incidence of AD complicated by atherosclerosis has increased with improvements of living standards and changes of eating habits. Accordingly, considering multiple risk factors, we successfully and efficiently established a novel AD model through a high-fat diet combined with chronic angiotensin II (AngII) infusion. Compared with traditional chemical induction model using AngII and β-aminopropionitrile, our model is more clinically relevant for atherosclerosis-related AD. Moreover, infiltration of neutrophils and apoptosis of vascular smooth muscle cells in AD tissues were more significant. In addition to enriching the existing models, the novel model may be a long-term useful tool for more in-depth investigation of AD mechanisms and preclinical therapeutic developments.
Collapse
Affiliation(s)
- Wenhui He
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Sanjiu Yu
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Haoyang Li
- Brigade 5 of Medical Undergraduate, Third Military Medical University (Army Medical University), Chongqing, China
| | - Ping He
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Tiantian Xiong
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Chaojun Yan
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jingyu Zhang
- Army Medical Center of PLA, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shan Chen
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Mei Guo
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Xu Tan
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Dan Zhong
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jianbin Sun
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Zhizhen Xu
- Department of Biochemistry and Molecular Biology, Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Wei Cheng
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| | - Jun Li
- Department of Cardiac Surgery, The First Affiliated Hospital of Third Military Medical University (Army Medical University), Chongqing, 400038, China.
| |
Collapse
|