1
|
Gao Y, Cai C, Adamo S, Biteus E, Kamal H, Dager L, Miners KL, Llewellyn-Lacey S, Ladell K, Amratia PS, Bentley K, Kollnberger S, Wu J, Akhirunnesa M, Jones SA, Julin P, Lidman C, Stanton RJ, Goepfert PA, Peluso MJ, Deeks SG, Davies HE, Aleman S, Buggert M, Price DA. Identification of soluble biomarkers that associate with distinct manifestations of long COVID. Nat Immunol 2025; 26:692-705. [PMID: 40307449 PMCID: PMC12043503 DOI: 10.1038/s41590-025-02135-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 03/14/2025] [Indexed: 05/02/2025]
Abstract
Long coronavirus disease (COVID) is a heterogeneous clinical condition of uncertain etiology triggered by infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we used ultrasensitive approaches to profile the immune system and the plasma proteome in healthy convalescent individuals and individuals with long COVID, spanning geographically independent cohorts from Sweden and the United Kingdom. Symptomatic disease was not consistently associated with quantitative differences in immune cell lineage composition or antiviral T cell immunity. Healthy convalescent individuals nonetheless exhibited higher titers of neutralizing antibodies against SARS-CoV-2 than individuals with long COVID, and extensive phenotypic analyses revealed a subtle increase in the expression of some co-inhibitory receptors, most notably PD-1 and TIM-3, among SARS-CoV-2 nonspike-specific CD8+ T cells in individuals with long COVID. We further identified a shared plasma biomarker signature of disease linking breathlessness with apoptotic inflammatory networks centered on various proteins, including CCL3, CD40, IKBKG, IL-18 and IRAK1, and dysregulated pathways associated with cell cycle progression, lung injury and platelet activation, which could potentially inform the diagnosis and treatment of long COVID.
Collapse
Affiliation(s)
- Yu Gao
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Curtis Cai
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Sarah Adamo
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Laboratory of Translational Immuno-Oncology, Department of Biomedicine, University and University Hospital Basel, Basel, Switzerland
| | - Elsa Biteus
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Center for Clinical Research Sörmland, Uppsala University, Uppsala, Sweden
| | - Habiba Kamal
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Lena Dager
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Kelly L Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Sian Llewellyn-Lacey
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Pragati S Amratia
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Kirsten Bentley
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Simon Kollnberger
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Jinghua Wu
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Mily Akhirunnesa
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Samantha A Jones
- Department of Respiratory Medicine, University Hospital Llandough, Penarth, UK
| | - Per Julin
- Post-COVID Policlinic, Karolinska University Hospital, Stockholm, Sweden
- Department of Neurobiology, Care Sciences and Society, Karolinska Institutet, Stockholm, Sweden
| | - Christer Lidman
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Richard J Stanton
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK
| | - Paul A Goepfert
- Division of Infectious Diseases, Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Michael J Peluso
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Steven G Deeks
- Department of Medicine, University of California, San Francisco, San Francisco, CA, USA
| | - Helen E Davies
- Department of Respiratory Medicine, University Hospital Llandough, Penarth, UK
| | - Soo Aleman
- Division of Infectious Diseases and Dermatology, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
| | - Marcus Buggert
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| | - David A Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK.
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, UK.
| |
Collapse
|
2
|
Nishimura H, Araki K, Mitsuoka C, Toriumi W, Kitajima S, Takahashi E. Possible involvement of neuropeptide Y sub-receptor 1 (NPY-Y1) in the anti-viral response of SARS-CoV-2 infection in Syrian hamster. Biomed Res 2025; 46:37-50. [PMID: 40189329 DOI: 10.2220/biomedres.46.37] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Co-administration of Molnupiravir and Remdesivir, treatments for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), inhibits viral replication and infectivity. Previous studies indicate that the neuropeptide Y sub-receptor 1 (NPY-Y1) is involved in influenza virus aggravation in mouse pulmonary phagocytes, but the exact mechanisms remain unclear. Understanding the NPY-Y1 receptor's involvement in influenza and SARS-CoV-2 in both mice and hamsters may help explore its potential as an indicator of viral infections and support the development of preventive care. This study examined the effects of Molnupiravir and Remdesivir on infected Syrian hamsters and the NPY pathway during SARS-CoV-2 infection. SARS-CoV-2 infection increased mRNA expression of NPY, NPY-Y1 receptors, and inflammatory cytokines and chemokines in hamster lungs. Co-administration of the drugs significantly reduced these expressions. Changes in NPY-Y1 receptor expression were correlated with NPY, IL-10, IL-12, and IFN-γ, implying a role in the antiviral response pathway. These findings highlight that changes in the mRNA expression levels of NPY and NPY-Y1 receptor are influenced by SARS-CoV-2 infection and that the antiviral drugs impact the NPY-NPY-Y1 receptor cascade. This implies the pathway's involvement in inflammatory responses during viral infection and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Haruka Nishimura
- Department of Biomedicine, Graduate School of Medical Sciences, Kyusyu University
| | - Kohei Araki
- Infectivity and Immunology Laboratory, FUKUOKA Research Institute, Trans Genic Inc
| | - Chihomi Mitsuoka
- Infectivity and Immunology Laboratory, FUKUOKA Research Institute, Trans Genic Inc
| | | | - Shunichi Kitajima
- Infectivity and Immunology Laboratory, FUKUOKA Research Institute, Trans Genic Inc
| | - Eiki Takahashi
- Department of Biomedicine, Graduate School of Medical Sciences, Kyusyu University
| |
Collapse
|
3
|
Drozd M, Hamilton F, Cheng CW, Lillie PJ, Brown OI, Chaddock N, Savic S, Naseem K, Iles MM, Morgan AW, Kearney MT, Cubbon RM. Plasma MERTK is causally associated with infection mortality. J Infect 2024; 89:106262. [PMID: 39241967 DOI: 10.1016/j.jinf.2024.106262] [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: 07/10/2024] [Revised: 08/26/2024] [Accepted: 08/28/2024] [Indexed: 09/09/2024]
Abstract
BACKGROUND Infectious diseases are a major cause of mortality in spite of existing public health, anti-microbial and vaccine interventions. We aimed to define plasma proteomic associates of infection mortality and then apply Mendelian randomisation (MR) to yield biomarkers that may be causally associated. METHODS We used UK Biobank plasma proteomic data to associate 2923 plasma proteins with infection mortality before 31st December 2019 (240 events in 52,520 participants). Since many plasma proteins also predict non-infection mortality, we focussed on those associated with >1.5-fold risk of infection mortality in an analysis excluding survivors. Protein quantitative trait scores (pQTS) were then used to identify whether genetically predicted protein levels also associated with infection mortality. To conduct Two Sample MR, we performed a genome-wide association study (GWAS) of infection mortality using UK Biobank participants without plasma proteomic data (n = 363,953 including 984 infection deaths). FINDINGS After adjusting for clinical risk factors, 1142 plasma proteins were associated with risk of infection mortality (false discovery rate <0.05). 259 proteins were associated with >1.5-fold increased risk of infection versus non-infection mortality. Of these, we identified genetically predicted increasing MERTK concentration was associated with increased risk of infection mortality. MR supported a causal association between increasing plasma MERTK protein and infection mortality (odds ratio 1.46 per unit; 95% CI 1.15- 1.85; p = 0.002). CONCLUSION Plasma MERTK is causally associated with infection mortality and warrants exploration as a potential therapeutic target.
Collapse
Affiliation(s)
- Michael Drozd
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK.
| | - Fergus Hamilton
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
| | - Chew W Cheng
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Patrick J Lillie
- Department of Infection, Castle Hill Hospital, Hull University Hospitals NHS Trust, Kingston Upon Hull, UK
| | - Oliver I Brown
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Natalie Chaddock
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Sinisa Savic
- Leeds Institute of Rheumatic and Musculoskeletal Medicine, School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Khalid Naseem
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK
| | - Mark M Iles
- NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Leeds, UK; Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Ann W Morgan
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Mark T Kearney
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Leeds, UK
| | - Richard M Cubbon
- Leeds Institute of Cardiovascular and Metabolic Medicine, School of Medicine, University of Leeds, Leeds, UK; NIHR Leeds Biomedical Research Centre, Leeds Teaching Hospitals NHS Trust, Chapel Allerton Hospital, Leeds, UK.
| |
Collapse
|
4
|
Bochnia-Bueno L, Coelho GM, Cataneo AHD, Zanluca C, Ferreira LH, Cavalcanti LPDG, Clementino MADF, Yaochite JNU, Dos Santos HG, Nogueira MB, Duarte Dos Santos CN, Raboni SM. Assessment of immune responses to a Comirnaty® booster following CoronaVac® vaccination in healthcare workers. Mem Inst Oswaldo Cruz 2024; 119:e230239. [PMID: 39258622 PMCID: PMC11385826 DOI: 10.1590/0074-02760230239] [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: 12/20/2023] [Accepted: 06/13/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND The immunological response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection and immunisation is variable. OBJECTIVES To describe the humoral immune response by correlating IgA and IgG antibodies with NAbs titration following CoronaVac® immunisation and an mRNA (Comirnaty®) booster among healthcare workers (HCWs) and to compare the cytokine and interleukin profiles between HCWs vaccinated with CoronaVac and coronavirus disease 2019 (COVID-19) infected patients. METHODS Samples from 133 HCWs collected at 20 (T1) and 90 (T2) days after CoronaVac immunisation and 15 (T3) days after a booster dose with the Comirnaty vaccine were analysed for IgA and IgG EIA and neutralisation assay. Cytokine levels from vaccinated individuals at T1 day and COVID-19 patients were compared. FINDINGS Neutralising antibodies (NAbs) were observed in 81.7% of participants at T1, but only 49.2% maintained detectable NAbs after 90 days. The booster dose increased NAbs response in all participants. The cytokines with the highest levels post-vaccination were IL-6 and MCP-1. The MCP-1, IL-18, and IFN- γ levels were higher in COVID-19 patients than in vaccinated HCWs, while IL-22 levels increased in the vaccinated HCWs group. MAIN CONCLUSIONS The neutralisation titres in the T2 samples decreased, and antibody levels detected at T2 showed a more significant reduction than the neutralisation. The higher IL-22 expression in immunised individuals compared to those with COVID-19 suggests that IL-22 may be beneficial in protecting against severe disease.
Collapse
Affiliation(s)
- Lucas Bochnia-Bueno
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | - Gabriela Mattoso Coelho
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Virologia Molecular, Curitiba, PR, Brasil
| | | | - Camila Zanluca
- Fundação Oswaldo Cruz-Fiocruz, Instituto Carlos Chagas, Laboratório de Virologia Molecular, Curitiba, PR, Brasil
| | - Laura Holtman Ferreira
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | | | | | - Juliana Navarro Ueda Yaochite
- Universidade Federal do Ceará, Faculdade de Farmácia, Odontologia e Enfermagem, Departamento de Análises Clínicas e Toxicologia, Fortaleza, CE, Brasil
| | | | - Meri Bordignon Nogueira
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| | | | - Sonia Mara Raboni
- Universidade Federal do Paraná, Laboratório de Virologia, Curitiba, PR, Brasil
- Universidade Federal do Paraná, Programa de Pós-Graduação em Microbiologia, Parasitologia e Patologia, Curitiba, PR, Brasil
| |
Collapse
|
5
|
Wang H, Zhao T, Zeng J, Zhang R, Pu L, Qian S, Xu S, Jiang Y, Pan L, Dai X, Guo X, Han L. Methods and clinical biomarker discovery for targeted proteomics using Olink technology. Proteomics Clin Appl 2024; 18:e2300233. [PMID: 38726756 DOI: 10.1002/prca.202300233] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/12/2024] [Accepted: 04/09/2024] [Indexed: 11/18/2024]
Abstract
PURPOSE This paper is to offer insights for designing research utilizing Olink technology to identify biomarkers and potential therapeutic targets for disease treatment. EXPERIMENTAL DESIGN We discusses the application of Olink technology in oncology, cardiovascular, respiratory and immune-related diseases, and Outlines the advantages and limitations of Olink technology. RESULTS Olink technology simplifies the search for therapeutic targets, advances proteomics research, reveals the pathogenesis of diseases, and ultimately helps patients develop precision treatments. CONCLUSIONS Although proteomics technology has been rapidly developed in recent years, each method has its own disadvantages, so in the future research, more methods should be selected for combined application to verify each other.
Collapse
Affiliation(s)
- Han Wang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Tian Zhao
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Jingjing Zeng
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Ruijie Zhang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Liyuan Pu
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Suying Qian
- Key Laboratory of Diagnosis and Treatment of Digestive System Tumors of Zhejiang Province, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Shan Xu
- Shen zhen Nanshan Center for Chronic Disease Control, Shenzhen, Guangdong, China
| | - Yannan Jiang
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Lifang Pan
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| | - Xiaoyu Dai
- Department of Anus & Intestine Surgery, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Xu Guo
- Department of Rehabilitation Medicine, Ningbo No.2 Hospital, Ningbo, Zhejiang, China
| | - Liyuan Han
- Department of Clinical Epidemiology, Ningbo No. 2 Hospital, Ningbo, Zhejiang, China
- Center for Cardiovascular and Cerebrovascular Epidemiology and Translational Medicine, Ningbo Institute of Life and Health Industry, University of Chinese Academy of Sciences, Ningbo, Zhejiang, China
| |
Collapse
|
6
|
Agamah FE, Ederveen THA, Skelton M, Martin DP, Chimusa ER, ’t Hoen PAC. Network-based integrative multi-omics approach reveals biosignatures specific to COVID-19 disease phases. Front Mol Biosci 2024; 11:1393240. [PMID: 39040605 PMCID: PMC11260748 DOI: 10.3389/fmolb.2024.1393240] [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: 02/28/2024] [Accepted: 05/22/2024] [Indexed: 07/24/2024] Open
Abstract
Background COVID-19 disease is characterized by a spectrum of disease phases (mild, moderate, and severe). Each disease phase is marked by changes in omics profiles with corresponding changes in the expression of features (biosignatures). However, integrative analysis of multiple omics data from different experiments across studies to investigate biosignatures at various disease phases is limited. Exploring an integrative multi-omics profile analysis through a network approach could be used to determine biosignatures associated with specific disease phases and enable the examination of the relationships between the biosignatures. Aim To identify and characterize biosignatures underlying various COVID-19 disease phases in an integrative multi-omics data analysis. Method We leveraged a multi-omics network-based approach to integrate transcriptomics, metabolomics, proteomics, and lipidomics data. The World Health Organization Ordinal Scale WHO Ordinal Scale was used as a disease severity reference to harmonize COVID-19 patient metadata across two studies with independent data. A unified COVID-19 knowledge graph was constructed by assembling a disease-specific interactome from the literature and databases. Disease-state specific omics-graphs were constructed by integrating multi-omics data with the unified COVID-19 knowledge graph. We expanded on the network layers of multiXrank, a random walk with restart on multilayer network algorithm, to explore disease state omics-specific graphs and perform enrichment analysis. Results Network analysis revealed the biosignatures involved in inducing chemokines and inflammatory responses as hubs in the severe and moderate disease phases. We observed distinct biosignatures between severe and moderate disease phases as compared to mild-moderate and mild-severe disease phases. Mild COVID-19 cases were characterized by a unique biosignature comprising C-C Motif Chemokine Ligand 4 (CCL4), and Interferon Regulatory Factor 1 (IRF1). Hepatocyte Growth Factor (HGF), Matrix Metallopeptidase 12 (MMP12), Interleukin 10 (IL10), Nuclear Factor Kappa B Subunit 1 (NFKB1), and suberoylcarnitine form hubs in the omics network that characterizes the moderate disease state. The severe cases were marked by biosignatures such as Signal Transducer and Activator of Transcription 1 (STAT1), Superoxide Dismutase 2 (SOD2), HGF, taurine, lysophosphatidylcholine, diacylglycerol, triglycerides, and sphingomyelin that characterize the disease state. Conclusion This study identified both biosignatures of different omics types enriched in disease-related pathways and their associated interactions (such as protein-protein, protein-transcript, protein-metabolite, transcript-metabolite, and lipid-lipid interactions) that are unique to mild, moderate, and severe COVID-19 disease states. These biosignatures include molecular features that underlie the observed clinical heterogeneity of COVID-19 and emphasize the need for disease-phase-specific treatment strategies. The approach implemented here can be used to find associations between transcripts, proteins, lipids, and metabolites in other diseases.
Collapse
Affiliation(s)
- Francis E. Agamah
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Thomas H. A. Ederveen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| | - Michelle Skelton
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Darren P. Martin
- Computational Biology Division, Department of Integrative Biomedical Sciences, Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Emile R. Chimusa
- Department of Applied Science, Faculty of Health and Life Sciences, Northumbria University, Newcastle, United Kingdom
| | - Peter A. C. ’t Hoen
- Department of Medical BioSciences, Radboud University Medical Center Nijmegen, Nijmegen, Netherlands
| |
Collapse
|
7
|
Fredolini C, Dodig-Crnković T, Bendes A, Dahl L, Dale M, Albrecht V, Mattsson C, Thomas CE, Torinsson Naluai Å, Gisslen M, Beck O, Roxhed N, Schwenk JM. Proteome profiling of home-sampled dried blood spots reveals proteins of SARS-CoV-2 infections. COMMUNICATIONS MEDICINE 2024; 4:55. [PMID: 38565620 PMCID: PMC10987641 DOI: 10.1038/s43856-024-00480-4] [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: 10/12/2022] [Accepted: 03/07/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Self-sampling of dried blood spots (DBS) offers new routes to gather valuable health-related information from the general population. Yet, the utility of using deep proteome profiling from home-sampled DBS to obtain clinically relevant insights about SARS-CoV-2 infections remains largely unexplored. METHODS Our study involved 228 individuals from the general Swedish population who used a volumetric DBS sampling device and completed questionnaires at home during spring 2020 and summer 2021. Using multi-analyte COVID-19 serology, we stratified the donors by their response phenotypes, divided them into three study sets, and analyzed 276 proteins by proximity extension assays (PEA). After normalizing the data to account for variances in layman-collected samples, we investigated the association of DBS proteomes with serology and self-reported information. RESULTS Our three studies display highly consistent variance of protein levels and share associations of proteins with sex (e.g., MMP3) and age (e.g., GDF-15). Studying seropositive (IgG+) and seronegative (IgG-) donors from the first pandemic wave reveals a network of proteins reflecting immunity, inflammation, coagulation, and stress response. A comparison of the early-infection phase (IgM+IgG-) with the post-infection phase (IgM-IgG+) indicates several proteins from the respiratory system. In DBS from the later pandemic wave, we find that levels of a virus receptor on B-cells differ between seropositive (IgG+) and seronegative (IgG-) donors. CONCLUSIONS Proteome analysis of volumetric self-sampled DBS facilitates precise analysis of clinically relevant proteins, including those secreted into the circulation or found on blood cells, augmenting previous COVID-19 reports with clinical blood collections. Our population surveys support the usefulness of DBS, underscoring the role of timing the sample collection to complement clinical and precision health monitoring initiatives.
Collapse
Affiliation(s)
- Claudia Fredolini
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
- Affinity Proteomics Unit, SciLifeLab Infrastructure, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Tea Dodig-Crnković
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Annika Bendes
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Leo Dahl
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Matilda Dale
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
- Affinity Proteomics Unit, SciLifeLab Infrastructure, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Vincent Albrecht
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Cecilia Mattsson
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
- Affinity Proteomics Unit, SciLifeLab Infrastructure, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Cecilia E Thomas
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden
| | - Åsa Torinsson Naluai
- Institute of Biomedicine, Sahlgrenska Academy at the University of Gothenburg, 405 30, Gothenburg, Sweden
| | - Magnus Gisslen
- Department of Infectious Diseases, The Sahlgrenska Academy at University of Gothenburg, 405 30, Gothenburg, Sweden
- Sahlgrenska University Hospital, 413 45, Gothenburg, Sweden
- Public Health Agency of Sweden, 171 65, Solna, Sweden
| | - Olof Beck
- Department of Clinical Neuroscience, Karolinska Institutet, 171 77, Stockholm, Sweden
| | - Niclas Roxhed
- MedTechLabs, BioClinicum, Karolinska University Hospital, 171 64, Solna, Sweden.
- Department of Micro and Nanosystems, School of Electrical Engineering and Computer Science, KTH Royal Institute of Technology Stockholm, 100 44, Stockholm, Sweden.
| | - Jochen M Schwenk
- Department of Protein Science, SciLifeLab, KTH Royal Institute of Technology, 171 65, Solna, Sweden.
| |
Collapse
|
8
|
Sundling C, Yman V, Mousavian Z, Angenendt S, Foroogh F, von Horn E, Lautenbach MJ, Grunewald J, Färnert A, Sondén K. Disease-specific plasma protein profiles in patients with fever after traveling to tropical areas. Eur J Immunol 2024; 54:e2350784. [PMID: 38308504 DOI: 10.1002/eji.202350784] [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: 09/21/2023] [Revised: 01/16/2024] [Accepted: 01/17/2024] [Indexed: 02/04/2024]
Abstract
Fever is common among individuals seeking healthcare after traveling to tropical regions. Despite the association with potentially severe disease, the etiology is often not determined. Plasma protein patterns can be informative to understand the host response to infection and can potentially indicate the pathogen causing the disease. In this study, we measured 49 proteins in the plasma of 124 patients with fever after travel to tropical or subtropical regions. The patients had confirmed diagnoses of either malaria, dengue fever, influenza, bacterial respiratory tract infection, or bacterial gastroenteritis, representing the most common etiologies. We used multivariate and machine learning methods to identify combinations of proteins that contributed to distinguishing infected patients from healthy controls, and each other. Malaria displayed the most unique protein signature, indicating a strong immunoregulatory response with high levels of IL10, sTNFRI and II, and sCD25 but low levels of sCD40L. In contrast, bacterial gastroenteritis had high levels of sCD40L, APRIL, and IFN-γ, while dengue was the only infection with elevated IFN-α2. These results suggest that characterization of the inflammatory profile of individuals with fever can help to identify disease-specific host responses, which in turn can be used to guide future research on diagnostic strategies and therapeutic interventions.
Collapse
Affiliation(s)
- Christopher Sundling
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Victor Yman
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Stockholm South Hospital, Stockholm, Sweden
| | - Zaynab Mousavian
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Sina Angenendt
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Fariba Foroogh
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Ellen von Horn
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| | - Maximilian Julius Lautenbach
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Johan Grunewald
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
- Respiratory Medicine Unit, Department of Medicine, Solna, Karolinska Institutet and Karolinska University Hospital Solna, Stockholm, Sweden
| | - Anna Färnert
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
- Department of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Klara Sondén
- Division of Infectious Diseases, Department of Medicine Solna, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
9
|
Abe K, Beer JC, Nguyen T, Ariyapala IS, Holmes TH, Feng W, Zhang B, Kuo D, Luo Y, Ma XJ, Maecker HT. Cross-Platform Comparison of Highly Sensitive Immunoassays for Inflammatory Markers in a COVID-19 Cohort. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 212:1244-1253. [PMID: 38334457 PMCID: PMC10948291 DOI: 10.4049/jimmunol.2300729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/19/2024] [Indexed: 02/10/2024]
Abstract
A variety of commercial platforms are available for the simultaneous detection of multiple cytokines and associated proteins, often employing Ab pairs to capture and detect target proteins. In this study, we comprehensively evaluated the performance of three distinct platforms: the fluorescent bead-based Luminex assay, the proximity extension-based Olink assay, and a novel proximity ligation assay platform known as Alamar NULISAseq. These assessments were conducted on human serum samples from the National Institutes of Health IMPACC study, with a focus on three essential performance metrics: detectability, correlation, and differential expression. Our results reveal several key findings. First, the Alamar platform demonstrated the highest overall detectability, followed by Olink and then Luminex. Second, the correlation of protein measurements between the Alamar and Olink platforms tended to be stronger than the correlation of either of these platforms with Luminex. Third, we observed that detectability differences across the platforms often translated to differences in differential expression findings, although high detectability did not guarantee the ability to identify meaningful biological differences. Our study provides valuable insights into the comparative performance of these assays, enhancing our understanding of their strengths and limitations when assessing complex biological samples, as exemplified by the sera from this COVID-19 cohort.
Collapse
Affiliation(s)
- Koji Abe
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Tran Nguyen
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | | | - Tyson H. Holmes
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| | - Wei Feng
- Alamar Biosciences, Inc., Fremont, CA 94538
| | | | - Dwight Kuo
- Alamar Biosciences, Inc., Fremont, CA 94538
| | - Yuling Luo
- Alamar Biosciences, Inc., Fremont, CA 94538
| | | | - Holden T. Maecker
- Institute for Immunity, Transplantation, and Infection, Stanford University School of Medicine, Stanford, CA 94305
| |
Collapse
|
10
|
Harriott NC, Ryan AL. Proteomic profiling identifies biomarkers of COVID-19 severity. Heliyon 2024; 10:e23320. [PMID: 38163173 PMCID: PMC10755324 DOI: 10.1016/j.heliyon.2023.e23320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/27/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
SARS-CoV-2 infection remains a major public health concern, particularly for the aged and those individuals with co-morbidities at risk for developing severe COVID-19. Understanding the pathogenesis and biomarkers associated with responses to SARS-CoV-2 infection remain critical components in developing effective therapeutic approaches, especially in cases of severe and long-COVID-19. In this study blood plasma protein expression was compared in subjects with mild, moderate, and severe COVID-19 disease. Evaluation of an inflammatory protein panel confirms upregulation of proteins including TNFβ, IL-6, IL-8, IL-12, already associated with severe cytokine storm and progression to severe COVID-19. Importantly, we identify several proteins not yet associated with COVID-19 disease, including mesothelin (MSLN), that are expressed at significantly higher levels in severe COVID-19 subjects. In addition, we find a subset of markers associated with T-cell and dendritic cell responses to viral infection that are significantly higher in mild cases and decrease in expression as severity of COVID-19 increases, suggesting that an immediate and effective activation of T-cells is critical in modulating disease progression. Together, our findings identify new targets for further investigation as therapeutic approaches for the treatment of SARS-CoV-2 infection and prevention of complications of severe COVID-19.
Collapse
Affiliation(s)
- Noa C. Harriott
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| | - Amy L. Ryan
- Hastings Center for Pulmonary Research, Division of Pulmonary, Critical Care and Sleep Medicine, Department of Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Stem Cell Biology and Regenerative Medicine, University of Southern California, Los Angeles CA 90033, USA
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City IA 52240, USA
| |
Collapse
|
11
|
Chatham WW. Macrophage Activation Syndrome in the Setting of Rheumatic Diseases. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2024; 1448:399-408. [PMID: 39117829 DOI: 10.1007/978-3-031-59815-9_27] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
Patients with established rheumatic disorders may develop complications of macrophage activation syndrome due to severe flares of the underlying disease (adult-onset Still's disease, SLE); however, in most other rheumatic disorders, MAS develops in association with identified viral or other infectious triggers. It is therefore important to pursue appropriate studies to identify potential infectious triggers in rheumatic disease patients who develop MAS. Management is best directed toward treatment of the triggering infections and combinations of high-dose corticosteroids, calcineurin inhibitors, and biologic therapies targeting IL-1 and/or IL-6 to suppress the associated cytokine storm.
Collapse
Affiliation(s)
- W Winn Chatham
- Department of Internal Medicine, University of Nevada, Las Vegas, Las Vegas, NV, USA.
| |
Collapse
|
12
|
Feng W, Beer JC, Hao Q, Ariyapala IS, Sahajan A, Komarov A, Cha K, Moua M, Qiu X, Xu X, Iyengar S, Yoshimura T, Nagaraj R, Wang L, Yu M, Engel K, Zhen L, Xue W, Lee CJ, Park CH, Peng C, Zhang K, Grzybowski A, Hahm J, Schmidt SV, Odainic A, Spitzer J, Buddika K, Kuo D, Fang L, Zhang B, Chen S, Latz E, Yin Y, Luo Y, Ma XJ. NULISA: a proteomic liquid biopsy platform with attomolar sensitivity and high multiplexing. Nat Commun 2023; 14:7238. [PMID: 37945559 PMCID: PMC10636041 DOI: 10.1038/s41467-023-42834-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 10/23/2023] [Indexed: 11/12/2023] Open
Abstract
The blood proteome holds great promise for precision medicine but poses substantial challenges due to the low abundance of most plasma proteins and the vast dynamic range of the plasma proteome. Here we address these challenges with NUcleic acid Linked Immuno-Sandwich Assay (NULISA™), which improves the sensitivity of traditional proximity ligation assays by ~10,000-fold to attomolar level, by suppressing assay background via a dual capture and release mechanism built into oligonucleotide-conjugated antibodies. Highly multiplexed quantification of both low- and high-abundance proteins spanning a wide dynamic range is achieved by attenuating signals from abundant targets with unconjugated antibodies and next-generation sequencing of barcoded reporter DNA. A 200-plex NULISA containing 124 cytokines and chemokines and other proteins demonstrates superior sensitivity to a proximity extension assay in detecting biologically important low-abundance biomarkers in patients with autoimmune diseases and COVID-19. Fully automated NULISA makes broad and in-depth proteomic analysis easily accessible for research and diagnostic applications.
Collapse
Affiliation(s)
- Wei Feng
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | - Qinyu Hao
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | | | | | - Katie Cha
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Mason Moua
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | - Xiaomei Xu
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | | | | | - Li Wang
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Ming Yu
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Kate Engel
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Lucas Zhen
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Wen Xue
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | | | - Cheng Peng
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | | | | | - Susanne V Schmidt
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | - Alexandru Odainic
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Australia
| | - Jasper Spitzer
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
| | | | - Dwight Kuo
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Lei Fang
- Alamar Biosciences, Inc, Fremont, CA, USA
| | | | - Steve Chen
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Eicke Latz
- Institute of Innate Immunity, Medical Faculty, University of Bonn, Bonn, Germany
- Deutsches Rheuma-Forschungszentrum Berlin (DRFZ), Berlin, Germany
| | - Yiyuan Yin
- Alamar Biosciences, Inc, Fremont, CA, USA
| | - Yuling Luo
- Alamar Biosciences, Inc, Fremont, CA, USA.
| | | |
Collapse
|
13
|
Abe K, Beer JC, Nguyen T, Ariyapala IS, Holmes TH, Feng W, Zhang B, Kuo D, Luo Y, Ma XJ, Maecker HT. Cross-platform comparison of highly-sensitive immunoassays for inflammatory markers in a COVID-19 cohort 1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.24.563866. [PMID: 37961126 PMCID: PMC10634816 DOI: 10.1101/2023.10.24.563866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
A variety of commercial platforms are available for the simultaneous detection of multiple cytokines and associated proteins, often employing antibody pairs to capture and detect target proteins. In this study, we comprehensively evaluated the performance of three distinct platforms: the fluorescent bead-based Luminex assay, the proximity extension-based Olink assay, and a novel proximity ligation assay platform known as Alamar NULISAseq. These assessments were conducted on serum samples from the NIH IMPACC study, with a focus on three essential performance metrics: detectability, correlation, and differential expression. Our results reveal several key findings. Firstly, the Alamar platform demonstrated the highest overall detectability, followed by Olink and then Luminex. Secondly, the correlation of protein measurements between the Alamar and Olink platforms tended to be stronger than the correlation of either of these platforms with Luminex. Thirdly, we observed that detectability differences across the platforms often translated to differences in differential expression findings, although high detectability did not guarantee the ability to identify meaningful biological differences. Our study provides valuable insights into the comparative performance of these assays, enhancing our understanding of their strengths and limitations when assessing complex biological samples, as exemplified by the sera from this COVID-19 cohort.
Collapse
|
14
|
Keur N, Saridaki M, Ricaño-Ponce I, Netea MG, Giamarellos-Bourboulis EJ, Kumar V. Analysis of inflammatory protein profiles in the circulation of COVID-19 patients identifies patients with severe disease phenotypes. Respir Med 2023; 217:107331. [PMID: 37364721 PMCID: PMC10290733 DOI: 10.1016/j.rmed.2023.107331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 06/14/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
BACKGROUND The coronavirus disease (COVID-19) caused by the severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) can present with a broad range of clinical manifestations, ranging from asymptomatic to severe multiple organ failure. The severity of the disease can vary depending on factors such as age, sex, ethnicity, and pre-existing medical conditions. Despite multiple efforts to identify reliable prognostic factors and biomarkers, the predictive capacity of these markers for clinical outcomes remains poor. Circulating proteins, which reflect the active mechanisms in an individual, can be easily measured in clinical practice and therefore may be useful as biomarkers for COVID-19 disease severity. In this study, we sought to identify protein biomarkers and endotypes for COVID-19 severity and evaluate their reproducibility in an independent cohort. METHODS We investigated a cohort of 153 Greek patients with confirmed SARS-CoV-2 infection in which plasma protein levels were measured using the Olink Explore 1536 panel, which consists of 1472 proteins. We compared the protein profiles from severe and moderate COVID-19 patients to identify proteins associated with disease severity. To evaluate the reproducibility of our findings, we compared the protein profiles of 174 patients with comparable COVID-19 severities in a US COVID-19 cohort to identify proteins consistently correlated with COVID-19 severity in both groups. RESULTS We identified 218 differentially regulated proteins associated with severity, 20 proteins were also replicated in an external cohort which we used for validation. Moreover, we performed unsupervised clustering of patients based on 97 proteins with the highest log2 fold changes in order to identify COVID-19 endotypes. Clustering of patients based on differentially regulated proteins revealed the presence of three clinical endotypes. While endotypes 2 and 3 were enriched for severe COVID-19 patients, endotypes 3 represented the most severe form of the disease. CONCLUSIONS These results suggest that identified circulating proteins may be useful for identifying COVID-19 patients with worse outcomes, and this potential utility may extend to other populations. TRIAL REGISTRATION NCT04357366.
Collapse
Affiliation(s)
- Nick Keur
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Maria Saridaki
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| | - Isis Ricaño-Ponce
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania.
| | | | - Vinod Kumar
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands; University of Groningen, University Medical Center Groningen, Department of Genetics, the Netherlands; Nitte (Deemed to Be University), Nitte University Centre for Science Education and Research (NUCSER), Deralakatte, Mangalore, India.
| |
Collapse
|
15
|
Schmidt F, Abdesselem HB, Suhre K, Vaikath NN, Sohail MU, Al-Nesf M, Bensmail I, Mashod F, Sarwath H, Bernhardt J, Schaefer-Ramadan S, Tan TM, Morris PE, Schenck EJ, Price D, Mohamed-Ali V, Al-Maadheed M, Arredouani A, Decock J, Blackburn JM, Choi AMK, El-Agnaf OM. Auto-immunoproteomics analysis of COVID-19 ICU patients revealed increased levels of autoantibodies related to the male reproductive system. Front Physiol 2023; 14:1203723. [PMID: 37520825 PMCID: PMC10374950 DOI: 10.3389/fphys.2023.1203723] [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: 04/11/2023] [Accepted: 06/28/2023] [Indexed: 08/01/2023] Open
Abstract
Background: Coronavirus disease (COVID-19) manifests many clinical symptoms, including an exacerbated immune response and cytokine storm. Autoantibodies in COVID-19 may have severe prodromal effects that are poorly understood. The interaction between these autoantibodies and self-antigens can result in systemic inflammation and organ dysfunction. However, the role of autoantibodies in COVID-19 complications has yet to be fully understood. Methods: The current investigation screened two independent cohorts of 97 COVID-19 patients [discovery (Disc) cohort from Qatar (case = 49 vs. control = 48) and replication (Rep) cohort from New York (case = 48 vs. control = 28)] utilizing high-throughput KoRectly Expressed (KREX) Immunome protein-array technology. Total IgG autoantibody responses were evaluated against 1,318 correctly folded and full-length human proteins. Samples were randomly applied on the precoated microarray slides for 2 h. Cy3-labeled secondary antibodies were used to detect IgG autoantibody response. Slides were scanned at a fixed gain setting using the Agilent fluorescence microarray scanner, generating a 16-bit TIFF file. Group comparisons were performed using a linear model and Fisher's exact test. Differentially expressed proteins were used for KEGG and WIKIpathway annotation to determine pathways in which the proteins of interest were significantly over-represented. Results and conclusion: Autoantibody responses to 57 proteins were significantly altered in the COVID-19 Disc cohort compared to healthy controls (p ≤ 0.05). The Rep cohort had altered autoantibody responses against 26 proteins compared to non-COVID-19 ICU patients who served as controls. Both cohorts showed substantial similarities (r 2 = 0.73) and exhibited higher autoantibody responses to numerous transcription factors, immunomodulatory proteins, and human disease markers. Analysis of the combined cohorts revealed elevated autoantibody responses against SPANXN4, STK25, ATF4, PRKD2, and CHMP3 proteins in COVID-19 patients. The sequences for SPANXN4 and STK25 were cross-validated using sequence alignment tools. ELISA and Western blot further verified the autoantigen-autoantibody response of SPANXN4. SPANXN4 is essential for spermiogenesis and male fertility, which may predict a potential role for this protein in COVID-19-associated male reproductive tract complications, and warrants further research.
Collapse
Affiliation(s)
- Frank Schmidt
- Proteomics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Houari B. Abdesselem
- Proteomics Core Facility, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Hamad Bin Khalifa University (HBKU), Doha, Qatar
- Neurological Disorders Research Center, QBRI, HBKU, Qatar Foundation, Doha, Qatar
| | - Karsten Suhre
- Bioinformatics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Nishant N. Vaikath
- Neurological Disorders Research Center, QBRI, HBKU, Qatar Foundation, Doha, Qatar
| | | | - Maryam Al-Nesf
- Hamad General Hospital, Hamad Medical Corporation, Doha, Qatar
- Center of Metabolism and Inflammation, Division of Medicine, University College London, London, United Kingdom
| | - Ilham Bensmail
- Proteomics Core Facility, Qatar Biomedical Research Institute (QBRI), Qatar Foundation, Hamad Bin Khalifa University (HBKU), Doha, Qatar
| | - Fathima Mashod
- Proteomics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Hina Sarwath
- Proteomics Core, Weill Cornell Medicine—Qatar, Doha, Qatar
| | - Joerg Bernhardt
- Institute for Microbiology, University of Greifswald, Greifswald, Germany
| | | | - Ti-Myen Tan
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Damansara Heights, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Priscilla E. Morris
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Damansara Heights, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Edward J. Schenck
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York Presbyterian Hospital—Weill Cornell Medical Center, Weill Cornell Medicine, New York, NY, United States
| | - David Price
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York Presbyterian Hospital—Weill Cornell Medical Center, Weill Cornell Medicine, New York, NY, United States
| | - Vidya Mohamed-Ali
- Center of Metabolism and Inflammation, Division of Medicine, University College London, London, United Kingdom
- Anti-Doping Laboratory Qatar, Doha, Qatar
| | - Mohammed Al-Maadheed
- Center of Metabolism and Inflammation, Division of Medicine, University College London, London, United Kingdom
- Anti-Doping Laboratory Qatar, Doha, Qatar
| | - Abdelilah Arredouani
- Diabetes Research Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Julie Decock
- College of Health and Life Sciences, Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
- Translational Cancer and Immunity Center, Qatar Biomedical Research Institute (QBRI), Hamad Bin Khalifa University (HBKU), Qatar Foundation, Doha, Qatar
| | - Jonathan M. Blackburn
- Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
- Sengenics Corporation, Damansara Heights, Kuala Lumpur, Malaysia
- Institute of Infectious Disease and Molecular Medicine, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Augustine M. K. Choi
- Department of Medicine, Division of Pulmonary and Critical Care Medicine, New York Presbyterian Hospital—Weill Cornell Medical Center, Weill Cornell Medicine, New York, NY, United States
| | - Omar M. El-Agnaf
- Neurological Disorders Research Center, QBRI, HBKU, Qatar Foundation, Doha, Qatar
| |
Collapse
|
16
|
Feng W, Beer J, Hao Q, Ariyapala IS, Sahajan A, Komarov A, Cha K, Moua M, Qiu X, Xu X, Iyengar S, Yoshimura T, Nagaraj R, Wang L, Yu M, Engel K, Zhen L, Xue W, Lee CJ, Park CH, Peng C, Zhang K, Grzybowski A, Hahm J, Schmidt SV, Odainic A, Spitzer J, Buddika K, Kuo D, Fang L, Zhang B, Chen S, Latz E, Yin Y, Luo Y, Ma XJ. NULISA: a novel proteomic liquid biopsy platform with attomolar sensitivity and high multiplexing. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.09.536130. [PMID: 37090549 PMCID: PMC10120728 DOI: 10.1101/2023.04.09.536130] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
The blood proteome holds great promise for precision medicine but poses substantial challenges due to the low abundance of most plasma proteins and the vast dynamic range across the proteome. We report a novel proteomic technology - NUcleic acid Linked Immuno-Sandwich Assay (NULISA™) - that incorporates a dual capture and release mechanism to suppress the assay background and improves the sensitivity of the proximity ligation assay by over 10,000-fold to the attomolar level. It utilizes pairs of antibodies conjugated to DNA oligonucleotides that enable immunocomplex purification and generate reporter DNA containing target- and sample-specific barcodes for a next-generation sequencing-based, highly multiplexed readout. A 200-plex NULISA targeting 124 cytokines and chemokines and 80 other immune response-related proteins demonstrated superior sensitivity for detecting low-abundance proteins and high concordance with other immunoassays. The ultrahigh sensitivity allowed the detection of previously difficult-to-detect, but biologically important, low-abundance biomarkers in patients with autoimmune diseases and COVID-19. Fully automated NULISA addresses longstanding challenges in proteomic analysis of liquid biopsies and makes broad and in-depth proteomic analysis accessible to the general research community and future diagnostic applications.
Collapse
|
17
|
Roquilly A, Francois B, Huet O, Launey Y, Lasocki S, Weiss E, Petrier M, Hourmant Y, Bouras M, Lakhal K, Le Bel C, Flattres Duchaussoy D, Fernández-Barat L, Ceccato A, Flet L, Jobert A, Poschmann J, Sebille V, Feuillet F, Koulenti D, Torres A. Interferon gamma-1b for the prevention of hospital-acquired pneumonia in critically ill patients: a phase 2, placebo-controlled randomized clinical trial. Intensive Care Med 2023; 49:530-544. [PMID: 37072597 PMCID: PMC10112824 DOI: 10.1007/s00134-023-07065-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/31/2023] [Indexed: 04/20/2023]
Abstract
PURPOSE We aimed to determine whether interferon gamma-1b prevents hospital-acquired pneumonia in mechanically ventilated patients. METHODS In a multicenter, placebo-controlled, randomized trial conducted in 11 European hospitals, we randomly assigned critically ill adults, with one or more acute organ failures, under mechanical ventilation to receive interferon gamma-1b (100 µg every 48 h from day 1 to 9) or placebo (following the same regimen). The primary outcome was a composite of hospital-acquired pneumonia or all-cause mortality on day 28. The planned sample size was 200 with interim safety analyses after enrolling 50 and 100 patients. RESULTS The study was discontinued after the second safety analysis for potential harm with interferon gamma-1b, and the follow-up was completed in June 2022. Among 109 randomized patients (median age, 57 (41-66) years; 37 (33.9%) women; all included in France), 108 (99%) completed the trial. Twenty-eight days after inclusion, 26 of 55 participants (47.3%) in the interferon-gamma group and 16 of 53 (30.2%) in the placebo group had hospital-acquired pneumonia or died (adjusted hazard ratio (HR) 1.76, 95% confidence interval (CI) 0.94-3.29; P = 0.08). Serious adverse events were reported in 24 of 55 participants (43.6%) in the interferon-gamma group and 17 of 54 (31.5%) in the placebo group (P = 0.19). In an exploratory analysis, we found that hospital-acquired pneumonia developed in a subgroup of patients with decreased CCL17 response to interferon-gamma treatment. CONCLUSIONS Among mechanically ventilated patients with acute organ failure, treatment with interferon gamma-1b compared with placebo did not significantly reduce the incidence of hospital-acquired pneumonia or death on day 28. Furthermore, the trial was discontinued early due to safety concerns about interferon gamma-1b treatment.
Collapse
Affiliation(s)
- Antoine Roquilly
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 1413, 44000, Nantes, France.
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France.
| | - Bruno Francois
- ICU Department and Inserm CIC 1435 & UMR 1092, University Hospital of Limoges, Limoges, France
| | - Olivier Huet
- Département d'anesthésie réanimation et medecine peri-operatoire, CHRU de Brest, Université de Bretagne Occidentale, 29000, Brest, France
| | - Yoann Launey
- Department of Anaesthesia, Critical Care and Perioperative Medicine, Univ Rennes, CHU Rennes, 35000, Rennes, France
| | - Sigismond Lasocki
- Department of Anesthesiology and Critical Care Medicine, University Hospital of Angers, 49000, Angers, France
| | - Emmanuel Weiss
- Department of Anesthesiology and Critical Care, Université Paris Cité, INSERM UMR_S1149, and AP-HP Nord, Hôpital Beaujon, Clichy, France
| | - Melanie Petrier
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | - Yannick Hourmant
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 1413, 44000, Nantes, France
| | - Marwan Bouras
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 1413, 44000, Nantes, France
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | - Karim Lakhal
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 1413, 44000, Nantes, France
| | - Cecilia Le Bel
- Nantes Université, CHU Nantes, INSERM, Anesthesie Réanimation, CIC 1413, 44000, Nantes, France
| | | | - Laia Fernández-Barat
- CELLEX research laboratories, CibeRes (Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, 06/06/0028), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
- Servei de Pneumologia, Hospital Clinic, Barcelona, Universitat de Barcelona, CIBERES, Icrea, IDIBAPS, Barcelona, Spain
| | - Adrian Ceccato
- Servei de Pneumologia, Hospital Clinic, Barcelona, Universitat de Barcelona, CIBERES, Icrea, IDIBAPS, Barcelona, Spain
| | - Laurent Flet
- Nantes Université, CHU Nantes, Pharmacie, 44000, Nantes, France
| | - Alexandra Jobert
- Nantes Université, CHU Nantes, DRI, Département promotion, cellule vigilances recherche, Nantes, France
- Nantes Université, Université de Tours, CHU Nantes, CHU Tours, INSERM, SPHERE U1246, 44000, Nantes, France
| | - Jeremie Poschmann
- Nantes Université, CHU Nantes, INSERM, Center for Research in Transplantation and Translational Immunology, UMR 1064, 44000, Nantes, France
| | - Veronique Sebille
- Nantes Université, CHU Nantes, DRI, Plateforme de Méthodologie et de Biostatistique, 44000, Nantes, France
- Nantes Université, Université de Tours, CHU Nantes, CHU Tours, INSERM, SPHERE U1246, 44000, Nantes, France
| | - Fanny Feuillet
- Nantes Université, CHU Nantes, DRI, Plateforme de Méthodologie et de Biostatistique, 44000, Nantes, France
- Nantes Université, Université de Tours, CHU Nantes, CHU Tours, INSERM, SPHERE U1246, 44000, Nantes, France
| | - Despoina Koulenti
- 2nd Critical Care Department, Attikon University Hospital, Athens, Greece
- Faculty of Medicine, UQ Centre for Clinical Research, The University of Queensland, Brisbane, Australia
| | - Antoni Torres
- CELLEX research laboratories, CibeRes (Centro de Investigación Biomédica en Red de Enfermedades Respiratorias, 06/06/0028), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| |
Collapse
|
18
|
Liu W, Han F, Wan M, Yang XZ. Integrated bioinformatics analysis identifies shared immune changes between ischemic stroke and COVID 19. Front Immunol 2023; 14:1102281. [PMID: 36969251 PMCID: PMC10030956 DOI: 10.3389/fimmu.2023.1102281] [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: 11/18/2022] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
Although COVID-19 is primarily a respiratory disease, its neurological complications, such as ischemic stroke (IS), have aroused growing concerns and reports. However, the molecular mechanisms that underlie IS and COVID-19 are not well understood. Therefore, we implemented transcriptomic analysis from eight GEO datasets consist of 1191 samples to detect common pathways and molecular biomarkers in IS and COVID-19 that help understand the linkage between them. Differentially expressed genes (DEGs) were detected for IS and COVID-19 separately for finding shared mechanisms and we found that immune-related pathways were outlined with statistical significance. JAK2, which was identified as a hub gene, was supposed to be a potential therapeutic gene targets during the immunological process of COVID-19 and IS. Besides, we found a decrease in the proportion of CD8+ T and T helper 2 cells in the peripheral circulation of both COVID and IS patients, and NCR3 expression was significantly correlated with this change. In conclusion, we demonstrated that transcriptomic analyses reported in this study could make a deeper understanding of the common mechanism and might be promising for effective therapeutic for IS and COVID-19.
Collapse
Affiliation(s)
- Wenhao Liu
- Eight-year program of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Han
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mengyao Wan
- Eight-year program of Clinical Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
- Department of Neurology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xin-Zhuang Yang
- Medical Research Center, State Key laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- *Correspondence: Xin-Zhuang Yang,
| |
Collapse
|
19
|
Divergent Cytokine and Chemokine Responses at Early Acute Simian Immunodeficiency Virus Infection Correlated with Virus Replication and CD4 T Cell Loss in a Rhesus Macaque Model. Vaccines (Basel) 2023; 11:vaccines11020264. [PMID: 36851142 PMCID: PMC9963901 DOI: 10.3390/vaccines11020264] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/16/2023] [Accepted: 01/21/2023] [Indexed: 01/27/2023] Open
Abstract
Cytokine and chemokine levels remain one of the significant predictive factors of HIV pathogenesis and disease outcome. Understanding the impact of cytokines and chemokines during early acute infection will help to recognize critical changes during HIV pathogenesis and might assist in establishing improved HIV treatment and prevention methods. Sixty-one cytokines and chemokines were evaluated in the plasma of an SIV-infected rhesus macaque model. A substantial change in 11 cytokines/growth factors and 9 chemokines were observed during acute infection. Almost all the cytokines/chemokines were below the baseline values for an initial couple of days of infection. We detected six important cytokines/chemokines, such as IL-18, IP-10, FLT3L, MCP-1, MCP-2, and MIP-3β, that can be used as biomarkers to predict the peripheral CD4+ T cell loss and increased viral replication during the acute SIV/HIV infection. Hence, regulating IL-18, IP-10, FLT3L, MCP-1, MCP-2, and MIP-3β expression might provide an antiviral response to combat acute SIV/HIV infection.
Collapse
|
20
|
Li C, Yue L, Ju Y, Wang J, Chen M, Lu H, Liu S, Liu T, Wang J, Hu X, Tuohetaerbaike B, Wen H, Zhang W, Xu S, Jiang C, Chen F. Serum Proteomic Analysis for New Types of Long-Term Persistent COVID-19 Patients in Wuhan. Microbiol Spectr 2022; 10:e0127022. [PMID: 36314975 PMCID: PMC9784772 DOI: 10.1128/spectrum.01270-22] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 10/07/2022] [Indexed: 12/24/2022] Open
Abstract
The emergence of a new type of COVID-19 patients, who were retested positive after hospital discharge with long-term persistent SARS-CoV-2 infection but without COVID-19 clinical symptoms (hereinafter, LTPPs), poses novel challenges to COVID-19 treatment and prevention. Why was there such a contradictory phenomenon in LTPPs? To explore the mechanism underlying this phenomenon, we performed quantitative proteomic analyses using the sera of 12 LTPPs (Wuhan Pulmonary Hospital), with the longest carrying history of 132 days, and mainly focused on 7 LTPPs without hypertension (LTPPs-NH). The results showed differential serum protein profiles between LTPPs/LTPPs-NH and health controls. Further analysis identified 174 differentially-expressed-proteins (DEPs) for LTPPs, and 165 DEPs for LTPPs-NH, most of which were shared. GO and KEGG analyses for these DEPs revealed significant enrichment of "coagulation" and "immune response" in both LTPPs and LTPPs-NH. A unity of contradictory genotypes in the 2 aspects were then observed: some DEPs showed the same dysregulated expressed trend as that previously reported for patients in the acute phase of COVID-19, which might be caused by long-term stimulation of persistent SARS-CoV-2 infection in LTPPs, further preventing them from complete elimination; in contrast, some DEPs showed the opposite expression trend in expression, so as to retain control of COVID-19 clinical symptoms in LTPPs. Overall, the contrary effects of these DEPs worked together to maintain the balance of LTPPs, further endowing their contradictory steady-state with long-term persistent SARS-CoV-2 infection but without symptoms. Additionally, our study revealed some potential therapeutic targets of COVID-19. Further studies on these are warranted. IMPORTANCE This study reported a new type of COVID-19 patients and explored the underlying molecular mechanism by quantitative proteomic analyses. DEPs were significantly enriched in "coagulation" and "immune response". Importantly, we identified 7 "coagulation system"- and 9 "immune response"-related DEPs, the expression levels of which were consistent with those previously reported for patients in the acute phase of COVID-19, which appeared to play a role in avoiding the complete elimination of SARS-CoV-2 in LTPPs. On the contrary, 6 "coagulation system"- and 5 "immune response"-related DEPs showed the opposite trend in expression. The 11 inconsistent serum proteins seem to play a key role in the fight against long-term persistent SARS-CoV-2 infection, further retaining control of COVID-19 clinical symptom of LTPPs. The 26 proteins can serve as potential therapeutic targets and are thus valuable for the treatment of LTPPs; further studies on them are warranted.
Collapse
Affiliation(s)
- Cuidan Li
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
| | - Liya Yue
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
| | - Yingjiao Ju
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jie Wang
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Mengfan Chen
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Hao Lu
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Sitong Liu
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Tao Liu
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Jing Wang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang, China
| | - Xin Hu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang, China
| | - Bahetibieke Tuohetaerbaike
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang, China
| | - Hao Wen
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang, China
| | - Wenbao Zhang
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang, China
| | - Sihong Xu
- Division II of In Vitro Diagnostics for Infectious Diseases, Institute for In Vitro Diagnostics Control, National Institutes for Food and Drug Control, Beijing, China
| | - Chunlai Jiang
- National Engineering Laboratory for AIDS Vaccine, School of Life Science, Jilin University, Changchun, China
| | - Fei Chen
- Beijing Institute of Genomics, Chinese Academy of Sciences, China National Center for Bioinformation, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Urumqi, Xinjiang, China
- Beijing Key Laboratory of Genome and Precision Medicine Technologies, Beijing, China
| |
Collapse
|
21
|
Maurer DJ, Barletta E, Heider A, Stocker N, Wallimann A, Villiger M, Villiger B, Bärenfaller K, Akdis CA, Kistler W. Immune-inflammatory proteome of elite ice hockey players before and after SARS-CoV-2 infection. Allergy 2022; 77:3144-3147. [PMID: 35690999 PMCID: PMC9347679 DOI: 10.1111/all.15405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Revised: 06/03/2022] [Accepted: 06/07/2022] [Indexed: 01/28/2023]
Affiliation(s)
- Debbie J. Maurer
- Swiss Research Institute for Sports Medicine (SRISM)DavosSwitzerland
- Division of Sports and Exercise MedicineDepartment of Sport, Exercise and HealthUniversity of BaselBaselSwitzerland
| | - Elena Barletta
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Swiss Institute of Bioinformatics (SIB)LausanneSwitzerland
| | - Anja Heider
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Nino Stocker
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | | | - Michael Villiger
- Swiss Research Institute for Sports Medicine (SRISM)DavosSwitzerland
- Department of Sports MedicineHospital DavosDavosSwitzerland
| | - Beat Villiger
- Swiss Research Institute for Sports Medicine (SRISM)DavosSwitzerland
- Medical Center Bad RagazBad RagazSwitzerland
| | - Katja Bärenfaller
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
- Swiss Institute of Bioinformatics (SIB)LausanneSwitzerland
| | - Cezmi A. Akdis
- Swiss Research Institute for Sports Medicine (SRISM)DavosSwitzerland
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| | - Walter Kistler
- Swiss Research Institute for Sports Medicine (SRISM)DavosSwitzerland
- Swiss Institute of Allergy and Asthma Research (SIAF)University of ZurichDavosSwitzerland
| |
Collapse
|
22
|
Increased mRNA Levels of ADAM17, IFITM3, and IFNE in Peripheral Blood Cells Are Present in Patients with Obesity and May Predict Severe COVID-19 Evolution. Biomedicines 2022; 10:biomedicines10082007. [PMID: 36009555 PMCID: PMC9406212 DOI: 10.3390/biomedicines10082007] [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: 06/10/2022] [Revised: 08/12/2022] [Accepted: 08/15/2022] [Indexed: 11/21/2022] Open
Abstract
Gene expression patterns in blood cells from SARS-CoV-2 infected individuals with different clinical phenotypes and body mass index (BMI) could help to identify possible early prognosis factors for COVID-19. We recruited patients with COVID-19 admitted in Hospital Universitari Son Espases (HUSE) between March 2020 and November 2021, and control subjects. Peripheral blood cells (PBCs) and plasma samples were obtained on hospital admission. Gene expression of candidate transcriptomic biomarkers in PBCs were compared based on the patients’ clinical status (mild, severe and critical) and BMI range (normal weight, overweight, and obesity). mRNA levels of ADAM17, IFITM3, IL6, CXCL10, CXCL11, IFNG and TYK2 were increased in PBCs of COVID-19 patients (n = 73) compared with controls (n = 47), independently of sex. Increased expression of IFNE was observed in the male patients only. PBC mRNA levels of ADAM17, IFITM3, CXCL11, and CCR2 were higher in those patients that experienced a more serious evolution during hospitalization. ADAM17, IFITM3, IL6 and IFNE were more highly expressed in PBCs of patients with obesity. Interestingly, the expression pattern of ADAM17, IFITM3 and IFNE in PBCs was related to both the severity of COVID-19 evolution and obesity status, especially in the male patients. In conclusion, gene expression in PBCs can be useful for the prognosis of COVID-19 evolution.
Collapse
|
23
|
Torres-Sangiao E, Giddey AD, Leal Rodriguez C, Tang Z, Liu X, Soares NC. Proteomic Approaches to Unravel Mechanisms of Antibiotic Resistance and Immune Evasion of Bacterial Pathogens. Front Med (Lausanne) 2022; 9:850374. [PMID: 35586072 PMCID: PMC9108449 DOI: 10.3389/fmed.2022.850374] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
The profound effects of and distress caused by the global COVID-19 pandemic highlighted what has been known in the health sciences a long time ago: that bacteria, fungi, viruses, and parasites continue to present a major threat to human health. Infectious diseases remain the leading cause of death worldwide, with antibiotic resistance increasing exponentially due to a lack of new treatments. In addition to this, many pathogens share the common trait of having the ability to modulate, and escape from, the host immune response. The challenge in medical microbiology is to develop and apply new experimental approaches that allow for the identification of both the microbe and its drug susceptibility profile in a time-sensitive manner, as well as to elucidate their molecular mechanisms of survival and immunomodulation. Over the last three decades, proteomics has contributed to a better understanding of the underlying molecular mechanisms responsible for microbial drug resistance and pathogenicity. Proteomics has gained new momentum as a result of recent advances in mass spectrometry. Indeed, mass spectrometry-based biomedical research has been made possible thanks to technological advances in instrumentation capability and the continuous improvement of sample processing and workflows. For example, high-throughput applications such as SWATH or Trapped ion mobility enable the identification of thousands of proteins in a matter of minutes. This type of rapid, in-depth analysis, combined with other advanced, supportive applications such as data processing and artificial intelligence, presents a unique opportunity to translate knowledge-based findings into measurable impacts like new antimicrobial biomarkers and drug targets. In relation to the Research Topic “Proteomic Approaches to Unravel Mechanisms of Resistance and Immune Evasion of Bacterial Pathogens,” this review specifically seeks to highlight the synergies between the powerful fields of modern proteomics and microbiology, as well as bridging translational opportunities from biomedical research to clinical practice.
Collapse
Affiliation(s)
- Eva Torres-Sangiao
- Clinical Microbiology Lab, University Hospital Marqués de Valdecilla, Santander, Spain
- Instituto de Investigación Sanitaria Marqués de Valdecilla (IDIVAL), Santander, Spain
- *Correspondence: Eva Torres-Sangiao,
| | - Alexander Dyason Giddey
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Division of Chemical and Systems Biology, Department of Integrative Biomedical Sciences, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa
| | - Cristina Leal Rodriguez
- Copenhagen Prospectives Studies on Asthma in Childhood, COPSAC, Copenhagen University Hospital, Herlev-Gentofte, Denmark
| | - Zhiheng Tang
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Xiaoyun Liu
- Department of Microbiology, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Nelson C. Soares
- Sharjah Institute of Medical Research, University of Sharjah, Sharjah, United Arab Emirates
- Department of Medicinal Chemistry, College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
- Nelson C. Soares,
| |
Collapse
|