1
|
Li H, Liu H, Wu H, Guo C, Zuo W, Zheng Y, Deng X, Xu J, Wang Y, Wang Z, Lu B, Hou B, Cao B. Reading of human acute immune dynamics in omicron SARS-CoV-2 breakthrough infection. Emerg Microbes Infect 2025; 14:2494705. [PMID: 40231451 PMCID: PMC12064115 DOI: 10.1080/22221751.2025.2494705] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/19/2025] [Accepted: 04/13/2025] [Indexed: 04/16/2025]
Abstract
The dynamics of the immune response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) breakthrough infections remain unclear, particularly when compared to responses in naive individuals. In this longitudinal prospective cohort study, 13 participants were recruited. Peripheral blood samples were collected every other day until day 7 after symptom onset. Transcriptome sequencing, single-cell sequencing, T-cell receptor (TCR) sequencing, B-cell receptor (BCR) sequencing, Olink proteomics, and antigen-antibody binding experiments were then performed. During the incubation periods of breakthrough infections, peripheral blood exhibited type 2 cytokine response, which shifted to type 1 cytokine response upon symptom onset. Plasma cytokine levels of C-X-C motif chemokine ligand 10, monocyte chemoattractant protein-1, interferon-γ, and interleukin-6 show larger changes in breakthrough infections than naïve infections. The inflammatory response in breakthrough infections rapidly subsided, returning to homeostasis by day 5 after symptom onset. Notably, the levels of monocyte-derived S100A8/A9, previously considered a marker of severe disease, physiologically significantly increased in the early stages of mild cases and persisted until day 7, suggesting a specific biological function. Longitudinal tracking also revealed that antibodies anti-Receptor Binding Domain (anti-RBD) in breakthrough infections significantly increased by day 7 after symptom onset, whereas cytotoxic T lymphocytes appeared by day 5. This study presents a reference for interpreting the immunological response to breakthrough infectious disease in humans.
Collapse
Affiliation(s)
- Haibo Li
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
| | - Hongyu Liu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Hongping Wu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Chang Guo
- Changping National Laboratory (CPNL), Beijing, People’s Republic of China
| | - Wenting Zuo
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Ying Zheng
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyan Deng
- Tsinghua University-Peking University Joint Center for Life Sciences, Beijing, People’s Republic of China
| | - Jiuyang Xu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Yeming Wang
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Zai Wang
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Binghuai Lu
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| | - Baidong Hou
- State Key Laboratory of Epigenetic Regulation and Intervention, Institute of Biophysics, Chinese Academy of Sciences, Beijing, People’s Republic of China
- University of Chinese Academy of Sciences, Beijing, People’s Republic of China
| | - Bin Cao
- National Center for Respiratory Medicine; State Key Laboratory of Respiratory Health and Multimorbidity; National Clinical Research Center for Respiratory Diseases; Institute of Respiratory Medicine, Chinese Academy of Medical Sciences; Department of Pulmonary and Critical Care Medicine, Center of Respiratory Medicine, China-Japan Friendship Hospital, Beijing, People’s Republic of China
- New Cornerstone Science Laboratory, Beijing, People’s Republic of China
- Department of Respiratory Medicine, Capital Medical University, Beijing, People’s Republic of China
- Institute of Clinical Medical Sciences, China-Japan Friendship Hospital, Beijing, People’s Republic of China
| |
Collapse
|
2
|
Deinhardt-Emmer S, Chousterman BG, Schefold JC, Flohé SB, Skirecki T, Kox M, Winkler MS, Cossarizza A, Wiersinga WJ, van der Poll T, Weigand MA, Cajander S, Giamarellos-Bourboulis EJ, Lachmann G, Girardis M, Scicluna BP, Ferrer R, Payen D, Weis S, Torres A, Bermejo-Martín JF, Osuchowski MF, Rubio I, Bouma HR. Sepsis in patients who are immunocompromised: diagnostic challenges and future therapies. THE LANCET. RESPIRATORY MEDICINE 2025:S2213-2600(25)00124-9. [PMID: 40409328 DOI: 10.1016/s2213-2600(25)00124-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 03/31/2025] [Accepted: 04/02/2025] [Indexed: 05/25/2025]
Abstract
Sepsis is a life-threatening, dysregulated host response to infection. Immunosuppression is a risk factor for infections and sepsis. However, the specific immune derangements elevating the risk for infections and sepsis remain unclear in the individual patient, raising the question of whether a general state of immunosuppression exists. In this Review, we explore the relationship between immunosuppression and sepsis, detailing the definitions, causes, and clinical implications. We address the effect of primary immunodeficiencies, acquired conditions, and drugs on the risk of infection and the development of sepsis. Patients with sepsis who are immunocompromised often present with atypical symptoms and diagnostic test results can differ, making early recognition difficult. Future perspectives entail novel biomarkers to improve early sepsis detection and tailored treatments to modulate immune function. Including patients who are immunocompromised in clinical trials is crucial to enhance the relevance of research findings and improve treatment strategies for this vulnerable population.
Collapse
Affiliation(s)
- Stefanie Deinhardt-Emmer
- Institute of Medical Microbiology, Jena University Hospital, Friedrich-Schiller University Jena, Jena, Germany
| | - Benjamin G Chousterman
- Department of Anesthesia and Critical Care, Lariboisière Hospital, APHP, Paris, France; Université Paris Cité, Inserm UMRS 942 Mascot, Paris, France
| | - Joerg C Schefold
- Department of Intensive Care Medicine, Inselspital, University of Bern, Bern University Hospital, Bern, Switzerland
| | - Stefanie B Flohé
- Department of Trauma, Hand, and Reconstructive Surgery, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Tomasz Skirecki
- Department of Translational Immunology and Experimental Intensive Care, Centre of Postgraduate Medical Education, Warsaw, Poland
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, Netherlands
| | - Martin S Winkler
- Department of Anaesthesiology, University Medical Center Göttingen, Göttingen, Germany
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands; Department of Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Tom van der Poll
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center, Amsterdam, Netherlands; Department of Medicine, Division of Infectious Diseases, Amsterdam University Medical Center, Amsterdam, Netherlands; Amsterdam Institute for Infection and Immunity, Amsterdam University Medical Center, Amsterdam, Netherlands
| | - Markus A Weigand
- Department of Anesthesiology, Medical Faculty Heidelberg, Heidelberg University, Heidelberg, Germany
| | - Sara Cajander
- Department of Infectious Diseases, Faculty of Medicine and Health, Örebro University, Örebro, Sweden
| | - Evangelos J Giamarellos-Bourboulis
- 4th Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece; Hellenic Institute for the Study of Sepsis, Athens, Greece
| | - Gunnar Lachmann
- Department of Anesthesiology and Intensive Care Medicine Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Massimo Girardis
- Department of Anesthesiology and Intensive Care Medicine, University Hospital of Modena, University of Modena and Reggio Emilia, Modena, Italy
| | - Brendon P Scicluna
- Department of Applied Biomedical Science, Faculty of Health Sciences, Mater Dei hospital, University of Malta, Malta; Centre for Molecular Medicine and Biobanking, Biomedical Sciences, University of Malta, Malta
| | - Ricard Ferrer
- Department of Intensive Care Medicine, Vall d'Hebron University Hospital, Barcelona, Spain; SODIR, Vall d'Hebron Institute of Research, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Didier Payen
- Paris 7 University Denis Diderot, Paris Sorbonne, Cité, Paris, France; Service de Maladies Infectieuses, CHU de Nice, Nice, France
| | - Sebastian Weis
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller University Jena, Jena, Germany; Institute for Infectious Disease and Infection Control, Jena University Hospital, Friedrich-Schiller University Jena, Jena, Germany; Leibniz Institute for Natural Product Research and Infection Biology, Hans-Knöll Institute-HKI, Jena, Germany
| | - Antoni Torres
- Pulmonology Department, Hospital Clinic of Barcelona, University of Barcelona, Ciberes, IDIBAPS, ICREA, Barcelona, Spain
| | - Jesús F Bermejo-Martín
- School of Medicine, Universidad de Salamanca, Salamanca, Spain; Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain; Centro de Investigación Biomédica en Red en Enfermedades Respiratorias, Instituto de Salud Carlos III, Madrid, Spain
| | - Marcin F Osuchowski
- Ludwig Boltzmann Institute for Traumatology, the Research Center in Cooperation with AUVA, Vienna, Austria
| | - Ignacio Rubio
- Department of Anesthesiology and Intensive Care Medicine, Jena University Hospital, Friedrich-Schiller University Jena, Jena, Germany
| | - Hjalmar R Bouma
- Department of Acute Care, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; Department of Internal Medicine, University Medical Center Groningen, University of Groningen, Groningen, Netherlands; Department of Clinical Pharmacy & Pharmacology, University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| |
Collapse
|
3
|
Mezouar S, Mege J. Monitoring Macrophage Polarization in Infectious Disease, Lesson From SARS-CoV-2 Infection. Rev Med Virol 2025; 35:e70034. [PMID: 40148134 PMCID: PMC11976041 DOI: 10.1002/ird3.70006] [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: 03/11/2025] [Revised: 03/11/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025]
Abstract
The concept of macrophage polarization has been largely used in human diseases to define a typology of activation of myeloid cells reminiscent of lymphocyte functional subsets. In COVID-19, several studies have investigated myeloid compartment dysregulation and macrophage polarization as an indicator of disease prognosis and monitoring. SARS-CoV-2 induces an in vitro activation state in monocytes and macrophages that does not match the polarization categories in most studies. In COVID-19 patients, monocytes and macrophages are activated but they do not show a polarization profile. Therefore, the investigation of polarization under basic conditions was not relevant to assess monocyte and macrophage activation. The analysis of monocytes and macrophages with high-throughput methods has allowed the identification of new functional subsets in the context of COVID-19. This approach proposes an innovative stratification of myeloid cell activation. These new functional subsets of myeloid cells would be better biomarkers to assess the risk of complications in COVID-19, reserving the concept of polarization for pharmacological programme evaluation. This review reappraises the polarization of monocytes and macrophages in viral infections, particularly in COVID-19.
Collapse
Affiliation(s)
- Soraya Mezouar
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Faculty of Medical and Paramedical SciencesAix‐Marseille UniversityHIPE Human LabMarseilleFrance
| | - Jean‐Louis Mege
- Centre National de la Recherche ScientifiqueÉtablissement Français du SangAnthropologie Bio‐Culturelle, Droit, Éthique et SantéAix‐Marseille UniversityMarseilleFrance
- Department of ImmunologyLa Timone HospitalMarseilleFrance
| |
Collapse
|
4
|
Jogani S, Pol AS, Prajapati M, Samal A, Bhatia K, Parmar J, Patel U, Shah F, Vyas N, Gupta S. scaLR: a low-resource deep neural network-based platform for single cell analysis and biomarker discovery. Brief Bioinform 2025; 26:bbaf243. [PMID: 40439670 PMCID: PMC12121358 DOI: 10.1093/bib/bbaf243] [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: 12/16/2024] [Revised: 04/14/2025] [Accepted: 05/02/2025] [Indexed: 06/02/2025] Open
Abstract
Single-cell ribonucleic acid (RNA) sequencing (scRNA-seq) produces vast amounts of individual cell profiling data. Its analysis presents a significant challenge in accurately annotating cell types and their associated biomarkers. Different pipelines based on deep neural network (DNN) methods have been employed to tackle these issues. These pipelines have arisen as a promising resource and can extract meaningful and concise features from noisy, diverse, and high-dimensional data to enhance annotations and subsequent analysis. Existing tools require high computational resources to execute large sample datasets. We have developed a cutting-edge platform known as scaLR (Single-cell analysis using low resource) that efficiently processes data into feature subsets, samples in batches to reduce the required memory for processing large datasets, and running DNN models in multiple central processing units. scaLR is equipped with data processing, feature extraction, training, evaluation, and downstream analysis. Its novel feature extraction algorithm first trains the model on a feature subset and stores the importance of the features for all the features in that subset. At the end of the training of all subsets, the top-K features are selected based on their importance. The final model is trained on top-K features; its performance evaluation and associated downstream analysis provide significant biomarkers for different cell types and diseases/traits. Our findings indicate that scaLR offers comparable prediction accuracy and requires less model training time and computational resources than existing Python-based pipelines. We present scaLR, a Python-based platform, engineered to utilize minimal computational resources while maintaining comparable execution times and analysis costs to existing frameworks.
Collapse
Affiliation(s)
- Saiyam Jogani
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Anand Santosh Pol
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Mayur Prajapati
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Amit Samal
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Kriti Bhatia
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Laxman Nagar Baner, Pune 411045, Maharashtra, India
| | - Jayendra Parmar
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Urvik Patel
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Falak Shah
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Nisarg Vyas
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| | - Saurabh Gupta
- Department of Generative AI & Bioinformatics, Infocusp Innovations, Gala-hub, Bopal, Ahmedabad 380058, Gujarat, India
| |
Collapse
|
5
|
Courtemanche O, Blais-Lecours P, Lesage S, Chabot-Roy G, Coderre L, Blanchet MR, Châteauvert N, Lellouche F, Marsolais D. Exploratory analyses of leukocyte responses in hospitalized patients treated with ozanimod following a severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) infection. Immunol Cell Biol 2025; 103:433-443. [PMID: 40025871 DOI: 10.1111/imcb.70006] [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: 01/22/2025] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 03/04/2025]
Abstract
Sphingosine-1-phosphate receptor 1 (S1P1) ligands effectively reduce immunopathological damage in viral pneumonia models. Specifically, S1P1 ligands inhibit cytokine storm and help preserve lung endothelial barrier integrity. We recently showed that the S1P receptor ligand ozanimod can be safely administered to hospitalized patients with coronavirus disease 2019 (COVID-19) exhibiting severe symptoms of viral pneumonia, with potential clinical benefits. Here, we extend on this study and investigate the impact of ozanimod on key features of the immune response in patients with severe COVID-19. We quantified circulating cytokine levels, peripheral immune cell numbers, proportions and activation status; we also monitored the quality of the humoral response by assessing anti-severe acute respiratory syndrome coronavirus 2 (SARS‑CoV‑2) antibodies. Our findings reveal that patients receiving ozanimod during acute SARS-CoV-2 infection exhibit significantly reduced numbers of circulating monocytes compared with those receiving standard care. Correspondingly, in the ozanimod-treated group, circulating levels of C-C motif ligand 2 (CCL2) were decreased. While treatment with ozanimod negatively impacted the humoral response to COVID-19 in unvaccinated patients, it did not impair the development of a robust anti-SARS-CoV-2 antibody response in vaccinated patients. These findings suggest that ozanimod influences key immune mechanisms during the acute phase of SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Olivier Courtemanche
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Pascale Blais-Lecours
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - Sylvie Lesage
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montreal, QC, Canada
| | | | - Lise Coderre
- Centre de Recherche de l'Hôpital Maisonneuve-Rosemont, Montréal, QC, Canada
- Département de microbiologie, infectiologie et immunologie, Université de Montréal, Montreal, QC, Canada
| | - Marie-Renée Blanchet
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
- Département de Médecine, Université Laval, Quebec, QC, Canada
| | - Nathalie Châteauvert
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
| | - François Lellouche
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
- Département de Médecine, Université Laval, Quebec, QC, Canada
| | - David Marsolais
- Centre de recherche de l'Institut Universitaire de Cardiologie et de Pneumologie de Québec - Université Laval, Québec, QC, Canada
- Département de Médecine, Université Laval, Quebec, QC, Canada
| |
Collapse
|
6
|
Xu C, Zhao LY, Ye CS, Xu KC, Xu KY. The application of machine learning in clinical microbiology and infectious diseases. Front Cell Infect Microbiol 2025; 15:1545646. [PMID: 40375898 PMCID: PMC12078339 DOI: 10.3389/fcimb.2025.1545646] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/08/2025] [Indexed: 05/18/2025] Open
Abstract
With the development of artificial intelligence(AI) in computer science and statistics, it has been further applied to the medical field. These applications include the management of infectious diseases, in which machine learning has created inroads in clinical microbiology, radiology, genomics, and the analysis of electronic health record data. Especially, the role of machine learning in microbiology has gradually become prominent, and it is used in etiological diagnosis, prediction of antibiotic resistance, association between human microbiome characteristics and complex host diseases, prognosis judgment, and prevention and control of infectious diseases. Machine learning in the field of microbiology mainly adopts supervised learning and unsupervised learning, involving algorithms from classification and regression to clustering and dimensionality reduction. This Review explains crucial concepts in machine learning for unfamiliar readers, describes machine learning's current applications in clinical microbiology and infectious diseases, and summarizes important approaches clinicians must be aware of when evaluating research using machine learning.
Collapse
Affiliation(s)
- Cheng Xu
- Clinical Laboratory of Chun’an First People’s Hospital, Zhejiang Provincial People’s Hospital Chun’an Branch, Hangzhou Medical College Affiliated Chun’an Hospital, Hangzhou, Zhejiang, China
| | - Ling-Yun Zhao
- Department of Medicine & Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR, China
| | - Cun-Si Ye
- Department of Clinical Laboratory Medicine, Institution of Microbiology and Infectious Diseases, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Ke-Chen Xu
- School of Psychology, Zhejiang Normal University, Jinhua, China
- Key Laboratory of Intelligent Education Technology and Application of Zhejiang Province, Zhejiang Normal University, Jinhua, China
| | - Ke-Yang Xu
- Faculty of Chinese Medicine, and State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Macao SAR, China
| |
Collapse
|
7
|
Keramati F, Leijte GP, Bruse N, Grondman I, Habibi E, Ruiz-Moreno C, Megchelenbrink W, Peters van Ton AM, Heesakkers H, Bremmers ME, van Grinsven E, Tesselaar K, van Staveren S, van der Velden WJ, Preijers FW, Te Pas B, van de Loop R, Gerretsen J, Netea MG, Stunnenberg HG, Pickkers P, Kox M. Systemic inflammation impairs myelopoiesis and interferon type I responses in humans. Nat Immunol 2025; 26:737-747. [PMID: 40251340 PMCID: PMC12043512 DOI: 10.1038/s41590-025-02136-4] [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: 03/15/2023] [Accepted: 03/17/2025] [Indexed: 04/20/2025]
Abstract
Systemic inflammatory conditions are classically characterized by an acute hyperinflammatory phase, followed by a late immunosuppressive phase that elevates the susceptibility to secondary infections. Comprehensive mechanistic understanding of these phases is largely lacking. To address this gap, we leveraged a controlled, human in vivo model of lipopolysaccharide (LPS)-induced systemic inflammation encompassing both phases. Single-cell RNA sequencing during the acute hyperinflammatory phase identified an inflammatory CD163+SLC39A8+CALR+ monocyte-like subset (infMono) at 4 h post-LPS administration. The late immunosuppressive phase was characterized by diminished expression of type I interferon (IFN)-responsive genes in monocytes, impaired myelopoiesis and a pronounced attenuation of the immune response on a secondary LPS challenge 1 week after the first. The infMono gene program and impaired myelopoiesis were also detected in patient cohorts with bacterial sepsis and coronavirus disease. IFNβ treatment restored type-I IFN responses and proinflammatory cytokine production and induced monocyte maturation, suggesting a potential treatment option for immunosuppression.
Collapse
Affiliation(s)
- Farid Keramati
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Center for Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Guus P Leijte
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Niklas Bruse
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Inge Grondman
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ehsan Habibi
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Sprott Centre for Stem Cell Research, Regenerative Medicine Program, Ottawa Hospital Research Institute, Ottawa, Québec, Canada
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, Québec, Canada
| | - Cristian Ruiz-Moreno
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Wout Megchelenbrink
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
- Department of Precision Medicine, University of Campania Luigi Vanvitelli, Naples, Italy
| | | | - Hidde Heesakkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Manita E Bremmers
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Erinke van Grinsven
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Kiki Tesselaar
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Selma van Staveren
- Department of Respiratory Medicine and Center of Translational Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- TmonoCOAST, Amsterdam, The Netherlands
| | | | - Frank W Preijers
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Brigit Te Pas
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Raoul van de Loop
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jelle Gerretsen
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Mihai G Netea
- Department of Internal Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
- Human Genomics Laboratory, Craiova University of Medicine and Pharmacy, Craiova, Romania
| | - Hendrik G Stunnenberg
- Department of Molecular Biology, Faculty of Science, Radboud University, Nijmegen, The Netherlands.
- Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands.
| | - Peter Pickkers
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Matthijs Kox
- Department of Intensive Care Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
8
|
Kotagiri P, Rae WM, Bergamaschi L, Pombal D, Lee JY, Noor NM, Sojwal RS, Rubin SJS, Unger LW, Tolmeijer SH, Manferrari G, Bashford-Rogers RJM, Bingham DB, Stift A, Rogalla S, Gubatan J, Lee JC, Smith KGC, McKinney EF, Boyd SD, Lyons PA. Disease-specific B cell clones are shared between patients with Crohn's disease. Nat Commun 2025; 16:3689. [PMID: 40246842 PMCID: PMC12006383 DOI: 10.1038/s41467-025-58977-y] [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: 01/25/2023] [Accepted: 04/08/2025] [Indexed: 04/19/2025] Open
Abstract
B cells have important functions in gut homeostasis, and dysregulated B cell populations are frequently observed in patients with inflammatory bowel diseases, including both ulcerative colitis (UC) and Crohn's disease (CD). How these B cell perturbations contribute to disease remains largely unknown. Here, we perform deep sequencing of the B cell receptor (BCR) repertoire in four cohorts of patients with CD, together with healthy controls and patients with UC. We identify BCR clones that are shared between patients with CD but not found in healthy individuals nor in patients with UC, indicating CD-associated B cell immune responses. Shared clones are present in the inflamed gut mucosa, draining intestinal lymph nodes and blood, suggesting the presence of common CD-associated antigens that drive B cell responses in CD patients.
Collapse
Affiliation(s)
- Prasanti Kotagiri
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA.
| | - William M Rae
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Discovery Sciences, AstraZeneca, Cambridge Biomedical Campus, Cambridge, UK
| | - Laura Bergamaschi
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Diana Pombal
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Ji-Yeun Lee
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Nurulamin M Noor
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Raoul S Sojwal
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Samuel J S Rubin
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - Lukas W Unger
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Sofie H Tolmeijer
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Giulia Manferrari
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Rachael J M Bashford-Rogers
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | - David B Bingham
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Anton Stift
- Division of Visceral Surgery, Department of General Surgery, Medical University of Vienna, Vienna, Austria
| | - Stephan Rogalla
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, CA, 94305, USA
| | - James C Lee
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- The Francis Crick Institute and UCL Institute of Liver and Digestive Health, Division of Medicine, Royal Free Campus, London, UK
| | - Kenneth G C Smith
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Eoin F McKinney
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK
| | - Scott D Boyd
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Paul A Lyons
- Cambridge Institute of Therapeutic Immunology and Infectious Disease, Jeffrey Cheah Biomedical Centre, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge School of Clinical Medicine, Cambridge, UK.
| |
Collapse
|
9
|
Wu Y, Serna R, Gan W, Fan Z. Different patterns of leukocyte immune responses to infection of ancestral SARS-CoV-2 and its variants. Front Cell Infect Microbiol 2025; 15:1508120. [PMID: 40313462 PMCID: PMC12043629 DOI: 10.3389/fcimb.2025.1508120] [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: 10/08/2024] [Accepted: 03/27/2025] [Indexed: 05/03/2025] Open
Abstract
Background Contributions of leukocytes to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) defense have been reported extensively. However, it remains unclear whether there are different leukocyte responses to ancestral SARS-CoV-2 and its variants. Methods We analyzed peripheral blood leukocyte and subtype concentrations from 575 COVID-19 patients and 950 non-COVID-19 subjects registered at the University of Connecticut John Dempsey Hospital between 2020 and 2022, which covers the ancestral strain, Delta, and Omicron variants. Results We found that neutrophils, immature granulocytes, and monocytes were elevated, and lymphocytes were reduced after infection. These hyperactive neutrophils/immature granulocytes and suppressed lymphocytes/monocytes were associated with poorer prognosis in ancestral strain infection. Different from the ancestral strain, hyperactive immature granulocytes were not shown in the decedents of Delta infection, and immature granulocyte concentration was not observed to be associated with mortality. In Omicron infection, suppressed lymphocytes and monocytes were not shown in the decedents, and lymphocyte/monocyte concentrations were not associated with mortality. Conclusions Our findings provided insights into different leukocyte immune responses to ancestral SARS-CoV-2, Delta, and Omicron variants.
Collapse
Affiliation(s)
- Yuanyuan Wu
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Raphael Serna
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Wenqi Gan
- Department of Public Health Sciences, University of Connecticut School of Medicine, Farmington, CT, United States
| | - Zhichao Fan
- Department of Immunology, University of Connecticut School of Medicine, Farmington, CT, United States
| |
Collapse
|
10
|
De T, Coin L, Herberg J, Johnson MR, Järvelin MR. Plasma metabolomic signatures for copy number variants and COVID-19 risk loci in Northern Finland populations. Sci Rep 2025; 15:13172. [PMID: 40240424 PMCID: PMC12003712 DOI: 10.1038/s41598-025-94839-9] [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: 07/16/2024] [Accepted: 03/17/2025] [Indexed: 04/18/2025] Open
Abstract
Copy number variants (CNVs) are an important class of genomic variation known to be important for human physiology and diseases. Here we present genome-wide metabolomic signatures for CNVs in two Finnish cohorts-The Northern Finland Birth Cohort 1966 (NFBC 1966) and NFBC 1986. We have analysed and reported CNVs in over 9,300 individuals and characterised their dosage effect (CNV-metabolomic QTL) on 228 plasma lipoproteins and metabolites. We have reported reference (normal physiology) metabolomic signatures for up to ~ 2.6 million COVID-19 GWAS results from the National Institutes of Health (NIH) GRASP database, including for outcomes related to COVID-19 death, severity, and hospitalisation. Furthermore, by analysing two exemplar genes for COVID-19 severity namely LZTFL1 and OAS1, we have reported here two additional candidate genes for COVID-19 severity biology, (1) NFIX, a gene related to viral (adenovirus) replication and hematopoietic stem cells and (2) ACSL1, a known candidate gene for sepsis and bacterial inflammation. Based on our results and current literature we hypothesise that (1) charge imbalance across the cellular membrane between cations (Fe2+, Mg2+ etc.) and anions (e.g. ROS, hydroxide ion from cellular Fenton reactions, superoxide etc.), (2) iron trafficking within and between different cell types e.g., macrophages and (3) systemic oxidative stress response (e.g. lipid peroxidation mediated inflammation), together could be of relevance in severe COVID-19 cases. To conclude, our unique atlas of univariate and multivariate metabolomic signatures for CNVs (~ 7.2 million signatures) with deep annotations of various multi-omics data sets provide an important reference knowledge base for human metabolism and diseases.
Collapse
Affiliation(s)
- Tisham De
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK.
- Department of Genomics of Common Diseases, Imperial College London, London, UK.
- Department of Infectious Disease, Imperial College London, London, UK.
| | - Lachlan Coin
- Department of Infectious Disease, Imperial College London, London, UK
- Department of Microbiology and Immunology, Institute for Infection and Immunity, University of Melbourne at The Peter Doherty, Melbourne, Australia
| | - Jethro Herberg
- Department of Infectious Disease, Imperial College London, London, UK
| | | | - Marjo-Riitta Järvelin
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, UK
- Centre for Life Course Health Research, Faculty of Medicine, University of Oulu, Oulu, Finland
- Unit of Primary Health Care and Medical Research Center, Oulu University Hospital, Oulu, Finland
- Centre for Environment and Health, Imperial College London, London, UK
- Biocenter Oulu, University of Oulu, Oulu, Finland
| |
Collapse
|
11
|
Liu J, Guo L, Zhong J, Wu Y, Wang X, Tang X, Min K, Yang Y, Peng W, Wang Q, Ding T, Gu X, Zhang H, Liu Y, Huang C, Cao B, Wang J, Ren L, Yang J. Proteomic Analysis of 442 Clinical Plasma Samples From Individuals With Symptom Records Revealed Subtypes of Convalescent Patients Who Had COVID-19. J Med Virol 2025; 97:e70203. [PMID: 40207927 PMCID: PMC11984345 DOI: 10.1002/jmv.70203] [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: 09/30/2024] [Revised: 01/11/2025] [Accepted: 01/21/2025] [Indexed: 04/11/2025]
Abstract
After the coronavirus disease 2019 (COVID-19) pandemic, the postacute effects of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection have gradually attracted attention. To precisely evaluate the health status of convalescent patients with COVID-19, we analyzed symptom and proteome data of 442 plasma samples from healthy controls, hospitalized patients, and convalescent patients 6 or 12 months after SARS-CoV-2 infection. Symptoms analysis revealed distinct relationships in convalescent patients. Results of plasma protein expression levels showed that C1QA, C1QB, C2, CFH, CFHR1, and F10, which regulate the complement system and coagulation, remained highly expressed even at the 12-month follow-up compared with their levels in healthy individuals. By combining symptom and proteome data, 442 plasma samples were categorized into three subtypes: S1 (metabolism-healthy), S2 (COVID-19 retention), and S3 (long COVID). We speculated that convalescent patients reporting hair loss could have a better health status than those experiencing headaches and dyspnea. Compared to other convalescent patients, those reporting sleep disorders, appetite decrease, and muscle weakness may need more attention because they were classified into the S2 subtype, which had the most samples from hospitalized patients with COVID-19. Subtyping convalescent patients with COVID-19 may enable personalized treatments tailored to individual needs. This study provides valuable plasma proteomic datasets for further studies associated with long COVID.
Collapse
Grants
- This work was supported by grants from the National Key R&D Program of China (2023YFC2507102), the Chinese Academy of Medical Sciences Innovation Fund for Medical Sciences, China (CIFMS2022-I2M-1-011, CIFMS2022-I2M-2-001, CIFMS2021-I2M-1-057, CIFMS2021-I2M-1-049, CIFMS2021-I2M-1-044, CIFMS2021-I2M-1-016, CIFMS2021-I2M-1-001, 2022-I2M-CoV19-003, and CIFMS2022-I2M-JB-003), the National Natural Science Foundation of China (82341064), the Haihe Laboratory of Cell Ecosystem Innovation Fund (22HHXBSS00008 and 22HHKYZX0034), and State Key Laboratory Special Fund 2060204.
Collapse
Affiliation(s)
- Jiangfeng Liu
- Haihe Laboratory of Cell EcosystemTianjinChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Li Guo
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Jingchuan Zhong
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Yue Wu
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xinming Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
| | - Xiaoyue Tang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Kaiyuan Min
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Yehong Yang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Wanjun Peng
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Qiaochu Wang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Tao Ding
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
| | - Xiaoying Gu
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Hui Zhang
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Ying Liu
- Medical DepartmentJin Yin‐Tan HospitalWuhanHubeiChina
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical SciencesWuhanHubeiChina
| | - Chaolin Huang
- Medical DepartmentJin Yin‐Tan HospitalWuhanHubeiChina
- Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical SciencesWuhanHubeiChina
| | - Bin Cao
- Tsinghua University‐Peking University Joint Center for Life SciencesBeijingChina
- Department of Pulmonary and Critical Care MedicineNational Center for Respiratory Medicine, Center of Respiratory Medicine, National Clinical Research Center for Respiratory DiseasesBeijingChina
- Institute of Respiratory Medicine, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Department of Pulmonary and Critical Care MedicineCapital Medical UniversityBeijingChina
| | - Jianwei Wang
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Respiratory Disease PathogenomicsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Lili Ren
- NHC Key Laboratory of Systems Biology of Pathogens and Christophe Merieux LaboratoryInstitute of Pathogen Biology, Chinese Academy of Medical SciencesBeijingChina
- Key Laboratory of Respiratory Disease PathogenomicsChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Juntao Yang
- Haihe Laboratory of Cell EcosystemTianjinChina
- State Key Laboratory of Common Mechanism Research for Major Diseases, Department of Biochemistry and Molecular BiologySchool of Basic Medicine, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, Peking Union Medical CollegeBeijingChina
- Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
12
|
Lai C, Lu S, Yang Y, You X, Xu F, Deng X, Lan L, Guo Y, Kuang Z, Luo Y, Yuan L, Meng L, Wu X, Song Z, Jiang N. Myeloid-Driven Immune Suppression Subverts Neutralizing Antibodies and T Cell Immunity in Severe COVID-19. J Med Virol 2025; 97:e70335. [PMID: 40183283 PMCID: PMC11969634 DOI: 10.1002/jmv.70335] [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: 11/28/2024] [Revised: 02/21/2025] [Accepted: 02/27/2025] [Indexed: 04/05/2025]
Abstract
The objective of this study was to better understand immune failure mechanisms during severe acute respiratory syndrome coronavirus 2, SARS-CoV-2 infection, which are critical for developing targeted vaccines and effective treatments. We collected 34 cases representing different disease severities and performed high-quality single-cell TCR/BCR sequencing to analyze the peripheral immune cell profiles. Additionally, we assessed antibody-neutralizing activity through in vitro experiments. Our integrated multiomics analysis uncovers a profound immune paradox in severe COVID-19: hyperinflammation coexists with immunosuppression, driven by distinct yet interconnected dysregulatory mechanisms. Severe patients develop robust humoral immunity, evidenced by clonally expanded plasma cells producing neutralizing antibodies (e.g., IGHG1-dominated responses) and antigen-specific T cell activation. However, these protective responses are counteracted by myeloid-driven immunosuppression, particularly CD14+ HMGB2+ monocytes exhibiting metabolic reprogramming and HLA-DR downregulation, coupled with progressive T cell exhaustion characterized by IFN-γ/TNF-α hyperactivation and impaired antigen presentation. Importantly, prolonged viral persistence in severe cases arises from a failure to coordinate humoral and cellular immunity-antibody-mediated neutralization cannot compensate for defective cytotoxic T cell function and monocyte-mediated immune suppression. These findings highlight the necessity for therapeutic strategies that simultaneously enhance antibody effector functions (e.g., Fc optimization), restore exhausted T cells, and reverse myeloid suppression. They also highlight the importance of vaccines designed to elicit balanced B cell memory and durable T cell responses, which are critical to preventing severe disease progression. By addressing the dual challenges of hyperinflammation and immunosuppression, such approaches could restore immune coordination and improve outcomes in severe COVID-19.
Collapse
Grants
- This work was supported by the National Key Research and Development Program of China (2021YFC2501800, 2022YFA0806200, 2023YFC0872500, and 2024YFC3044600), the National Natural Science Foundation of China (82072214, 82272198, and 82202373), the Science and Technology of Shanghai Committee (21MC1930400, 22Y11900100, and 23Y31900100), and the Shanghai Municipal Health Commission (2023ZDFC0101).
Collapse
Affiliation(s)
- Cong Lai
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Su Lu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yilin Yang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xiaoyu You
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Feixiang Xu
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Xinran Deng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Lulu Lan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yuesheng Guo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Zhongshu Kuang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Yue Luo
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Li Yuan
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
| | - Lu Meng
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| | - Xueling Wu
- Department of Respiratory MedicineShanghai Jiaotong University School of Medicine, Renji HospitalShanghaiChina
| | - Zhenju Song
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Emergency Rescue and Critical CareFudan UniversityShanghaiChina
- Shanghai Institute of Infectious Disease and BiosecurityFudan UniversityShanghaiChina
| | - Ning Jiang
- Department of Emergency MedicineSchool of Life Sciences, Zhongshan HospitalFudan UniversityShanghaiChina
- Institute of Infection and HealthFudan UniversityShanghaiChina
| |
Collapse
|
13
|
Abstract
Mucosal-associated invariant T (MAIT) cells are evolutionarily conserved T cells that recognize microbial metabolites. They are abundant in humans and conserved during mammalian evolution, which suggests that they have important nonredundant functions. In this article, we discuss the evolutionary conservation of MAIT cells and describe their original developmental process. MAIT cells exert a wide variety of effector functions, from killing infected cells and promoting inflammation to repairing tissues. We provide insights into these functions and discuss how they result from the context of stimulation encountered by MAIT cells in different tissues and pathological settings. We describe how MAIT cell numbers and features are modified in disease states, focusing mainly on in vivo models. Lastly, we discuss emerging strategies to manipulate MAIT cells for therapeutic purposes.
Collapse
Affiliation(s)
- Marion Salou
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Rafael A Paiva
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
| | - Olivier Lantz
- Immunity and Cancer, INSERM U932, PSL University, Institut Curie, Paris, France; , ,
- Laboratoire d'Immunologie Clinique, Institut Curie, Paris, France
- Centre d'Investigation Clinique en Biothérapie, Gustave-Roussy and Institut Curie (CIC-BT1428), Paris, France
| |
Collapse
|
14
|
Li CX, Huang C, Chen DS. scPANDA: PAN-Blood Data Annotator with a 10-Million Single-Cell Atlas. CHINESE MEDICAL SCIENCES JOURNAL = CHUNG-KUO I HSUEH K'O HSUEH TSA CHIH 2025; 40:68-87. [PMID: 40164519 DOI: 10.24920/004472] [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] [Indexed: 04/02/2025]
Abstract
OBJECTIVES Recent advancements in single-cell RNA sequencing (scRNA-seq) have revolutionized the study of cellular heterogeneity, particularly within the hematological system. However, accurately annotating cell types remains challenging due to the complexity of immune cells. To address this challenge, we develop a PAN-blood single-cell Data Annotator (scPANDA), which leverages a comprehensive 10-million-cell atlas to provide precise cell type annotation. METHODS The atlas, constructed from data collected in 16 studies, incorporated rigorous quality control, preprocessing, and integration steps to ensure a high-quality reference for annotation. scPANDA utilizes a three-layer inference approach, progressively refining cell types from broad compartments to specific clusters. Iterative clustering and harmonization processes were employed to maintain cell type purity throughout the analysis. Furthermore, the performance of scPANDA was evaluated in three external datasets. RESULTS The atlas was structured hierarchically, consisting of 16 compartments, 54 classes, 4,460 low-level clusters (pd_cc_cl_tfs), and 611 high-level clusters (pmid_cts). Robust performance of the tool was demonstrated in annotating diverse immune scRNA-seq datasets, analyzing immune-tumor coexisting clusters in renal cell carcinoma, and identifying conserved cell clusters across species. CONCLUSIONS scPANDA exemplifies effective reference mapping with a large-scale atlas, enhancing the accuracy and reliability of blood cell type identification.
Collapse
Affiliation(s)
- Chang-Xiao Li
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China
| | - Can Huang
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China
| | - Dong-Sheng Chen
- State Key Laboratory of Common Mechanism Research for Major Diseases, Suzhou Institute of Systems Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Suzhou 215123, Jiangsu Province, China.
| |
Collapse
|
15
|
Gossez M, Vigneron C, Vandermoeten A, Lepage M, Courcol L, Coudereau R, Paidassai H, Jallades L, Lopez J, Kandara K, Ortillon M, Mommert M, Fabri A, Peronnet E, Grosjean C, Buisson M, Lukaszewicz AC, Rimmelé T, Argaud L, Cour M, Py BF, Thaunat O, Defrance T, Monneret G, Venet F. PD-L1 + plasma cells suppress T lymphocyte responses in patients with sepsis and mouse sepsis models. Nat Commun 2025; 16:3030. [PMID: 40155394 PMCID: PMC11953283 DOI: 10.1038/s41467-025-57706-9] [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: 03/28/2023] [Accepted: 02/28/2025] [Indexed: 04/01/2025] Open
Abstract
Sepsis, a leading cause of death in intensive care units, is associated with immune alterations that increase the patients' risk of secondary infections and mortality, so better understandings of the pathophysiology of sepsis-induced immunosuppression is essential for the development of therapeutic strategies. In a murine model of sepsis that recapitulates immune alterations observed in patients, here we demonstrate that PD-L1+CD44+B220LowCD138+IgM+ regulatory plasma cells are induced in spleen and regulate ex vivo proliferation and IFNɣ secretion induced by stimulation of T splenocytes. This effect is mediated both by cell-cell contact through increased PD-L1 expression on plasma cells and by production of a soluble factor. These observations are recapitulated in three cohorts of critically ill patients with bacterial and viral sepsis in association with increased mortality. Our findings thus reveal the function of regulatory plasma cells in the pathophysiology of sepsis-induced immune alterations, and present a potential therapeutic target for improving immune cell function impaired by sepsis.
Collapse
Affiliation(s)
- Morgane Gossez
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Clara Vigneron
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Alexandra Vandermoeten
- Service Commun des Animaleries de Rockefeller (SCAR) - Université Claude Bernard lyon1, Structure Fédérative de Recherche (SFR) Santé Lyon Est, Lyon, France
| | - Margot Lepage
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Louise Courcol
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Remy Coudereau
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Helena Paidassai
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Laurent Jallades
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
- Hospices Civils de Lyon, Lyon Sud University Hospital, Hematology Laboratory, Pierre-Bénite, France
| | - Jonathan Lopez
- Hospices Civils de Lyon, Biochemistry and Molecular Biology department, Lyon Est Faculty of Medicine, Université Claude Bernard Lyon 1, Université de Lyon, Lyon Sud University Hospital, Pierre-Bénite, France
| | - Khalil Kandara
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Marine Ortillon
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Marine Mommert
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Astrid Fabri
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
| | - Estelle Peronnet
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Clémence Grosjean
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Marielle Buisson
- Centre d'Investigation Clinique de Lyon (CIC 1407 Inserm), Hospices Civils de Lyon, Lyon, France
| | - Anne-Claire Lukaszewicz
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
- Hospices Civils de Lyon, Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Lyon, France
| | - Thomas Rimmelé
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
- Hospices Civils de Lyon, Anesthesia and Critical Care Medicine Department, Edouard Herriot Hospital, Lyon, France
| | - Laurent Argaud
- Hospices Civils de Lyon, Medical Intensive Care Department, Edouard Herriot Hospital, Lyon, France
| | - Martin Cour
- Hospices Civils de Lyon, Medical Intensive Care Department, Edouard Herriot Hospital, Lyon, France
| | - Bénédicte F Py
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Olivier Thaunat
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Thierry Defrance
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France
| | - Guillaume Monneret
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France
- EA 7426 "Pathophysiology of Injury-Induced Immunosuppression" (Université Claude Bernard Lyon 1, Hospices Civils de Lyon, bioMérieux), Lyon, France
| | - Fabienne Venet
- Hospices Civils de Lyon, Immunology Laboratory, Lyon-Sud & Edouard Herriot University Hospitals, Lyon, France.
- CIRI, Centre International de Recherche en Infectiologie, Univ Lyon, Inserm U1111, Université Claude Bernard-Lyon 1, CNRS, UMR5308, ENS de Lyon, Lyon, France.
| |
Collapse
|
16
|
Gavriilidis GI, Vasileiou V, Dimitsaki S, Karakatsoulis G, Giannakakis A, Pavlopoulos GA, Psomopoulos F. APNet, an explainable sparse deep learning model to discover differentially active drivers of severe COVID-19. Bioinformatics 2025; 41:btaf063. [PMID: 39921901 PMCID: PMC11897427 DOI: 10.1093/bioinformatics/btaf063] [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: 01/10/2024] [Revised: 01/18/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025] Open
Abstract
MOTIVATION Computational analyses of bulk and single-cell omics provide translational insights into complex diseases, such as COVID-19, by revealing molecules, cellular phenotypes, and signalling patterns that contribute to unfavourable clinical outcomes. Current in silico approaches dovetail differential abundance, biostatistics, and machine learning, but often overlook nonlinear proteomic dynamics, like post-translational modifications, and provide limited biological interpretability beyond feature ranking. RESULTS We introduce APNet, a novel computational pipeline that combines differential activity analysis based on SJARACNe co-expression networks with PASNet, a biologically informed sparse deep learning model, to perform explainable predictions for COVID-19 severity. The APNet driver-pathway network ingests SJARACNe co-regulation and classification weights to aid result interpretation and hypothesis generation. APNet outperforms alternative models in patient classification across three COVID-19 proteomic datasets, identifying predictive drivers and pathways, including some confirmed in single-cell omics and highlighting under-explored biomarker circuitries in COVID-19. AVAILABILITY AND IMPLEMENTATION APNet's R, Python scripts, and Cytoscape methodologies are available at https://github.com/BiodataAnalysisGroup/APNet.
Collapse
Affiliation(s)
- George I Gavriilidis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, GR57001, Greece
| | - Vasileios Vasileiou
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, GR57001, Greece
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, GR68100, Greece
| | - Stella Dimitsaki
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, GR57001, Greece
| | - Georgios Karakatsoulis
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, GR57001, Greece
| | - Antonis Giannakakis
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, GR68100, Greece
- University Research Institute of Maternal and Child Health and Precision Medicine, National and Kapodistrian University of Athens, Athens, GR11527, Greece
| | - Georgios A Pavlopoulos
- Institute for Fundamental Biomedical Research, BSRC “Alexander Fleming”, Vari, GR16672, Greece
- Center of New Biotechnologies & Precision Medicine, Department of Medicine, School of Health Sciences, National and Kapodistrian University of Athens, Athens, GR11528, Greece
| | - Fotis Psomopoulos
- Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, GR57001, Greece
| |
Collapse
|
17
|
Ying H, Wu X, Jia X, Yang Q, Liu H, Zhao H, Chen Z, Xu M, Wang T, Li M, Zhao Z, Zheng R, Wang S, Lin H, Xu Y, Lu J, Wang W, Ning G, Zheng J, Bi Y. Single-cell transcriptome-wide Mendelian randomization and colocalization reveals immune-mediated regulatory mechanisms and drug targets for COVID-19. EBioMedicine 2025; 113:105596. [PMID: 39933264 PMCID: PMC11867302 DOI: 10.1016/j.ebiom.2025.105596] [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: 08/20/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/13/2025] Open
Abstract
BACKGROUND COVID-19 continues to show long-term impacts on our health. Limited effective immune-mediated antiviral drugs have been launched. METHODS We conducted a Mendelian randomization (MR) and colocalization analysis using 26,597 single-cell expression quantitative trait loci (sc-eQTL) to proxy effects of expressions of 16,597 genes in 14 peripheral blood immune cells and tested them against four COVID-19 outcomes from COVID-19 Genetic Housing Initiative GWAS meta-analysis Round 7. We also carried out additional validations including colocalization, linkage disequilibrium check and host-pathogen interactome predictions. We integrated MR findings with clinical trial evidence from several drug gene related databases to identify drugs with repurposing potential. Finally, we developed a tier system and identified immune-cell-based prioritized drug targets for COVID-19. FINDINGS We identified 132 putative causal genes in 14 immune cells (343 MR associations) for COVID-19, with 58 genes that were not reported previously. 145 (73%) gene-COVID-19 pairs showed effects on COVID-19 in only one immune cell type, which implied widespread immune-cell specific effects. For pathway analyses, we found the putative causal genes were enriched in natural killer (NK) recruiting cells but de-enriched in NK cells. Using a deep learning model, we found 107 (81%) of the putative causal genes (41 novel genes) were predicted to interact with SARS-COV-2 proteins. Integrating the above evidence with drug trial information, we developed a tier system and prioritized 37 drug targets for COVID-19. INTERPRETATION Our study showcased the central role of immune-mediated regulatory mechanisms for COVID-19 and prioritized drug targets that might inform interventions for viral infectious diseases. FUNDING This work was supported by grants from the National Key Research and Development Program of China (2022YFC2505203).
Collapse
Affiliation(s)
- Hui Ying
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xueyan Wu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiaojing Jia
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qianqian Yang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haoyu Liu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huiling Zhao
- MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK
| | - Zhihe Chen
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Min Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Tiange Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mian Li
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhiyun Zhao
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ruizhi Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shuangyuan Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hong Lin
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Xu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jieli Lu
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Weiqing Wang
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guang Ning
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Zheng
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; MRC Integrative Epidemiology Unit (IEU), Bristol Medical School, University of Bristol, Oakfield House, Oakfield Grove, Bristol, BS8 2BN, UK.
| | - Yufang Bi
- Department of Endocrine and Metabolic Diseases, Shanghai Institute of Endocrine and Metabolic Diseases, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Shanghai National Clinical Research Center for Endocrine and Metabolic Diseases, Key Laboratory for Endocrine and Metabolic Diseases of the National Health Commission of the PR China, Shanghai National Center for Translational Medicine, Shanghai Digital Medicine Innovation Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
18
|
Jiang L, Dalgarno C, Papalexi E, Mascio I, Wessels HH, Yun H, Iremadze N, Lithwick-Yanai G, Lipson D, Satija R. Systematic reconstruction of molecular pathway signatures using scalable single-cell perturbation screens. Nat Cell Biol 2025; 27:505-517. [PMID: 40011560 PMCID: PMC12083445 DOI: 10.1038/s41556-025-01622-z] [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: 02/21/2024] [Accepted: 01/21/2025] [Indexed: 02/28/2025]
Abstract
Recent advancements in functional genomics have provided an unprecedented ability to measure diverse molecular modalities, but predicting causal regulatory relationships from observational data remains challenging. Here, we leverage pooled genetic screens and single-cell sequencing (Perturb-seq) to systematically identify the targets of signalling regulators in diverse biological contexts. We demonstrate how Perturb-seq is compatible with recent and commercially available advances in combinatorial indexing and next-generation sequencing, and perform more than 1,500 perturbations split across six cell lines and five biological signalling contexts. We introduce an improved computational framework (Mixscale) to address cellular variation in perturbation efficiency, alongside optimized statistical methods to learn differentially expressed gene lists and conserved molecular signatures. Finally, we demonstrate how our Perturb-seq derived gene lists can be used to precisely infer changes in signalling pathway activation for in vivo and in situ samples. Our work enhances our understanding of signalling regulators and their targets, and lays a computational framework towards the data-driven inference of an 'atlas' of perturbation signatures.
Collapse
Affiliation(s)
| | | | - Efthymia Papalexi
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | - Isabella Mascio
- New York Genome Center, New York, NY, USA
- Center for Genomics and Systems Biology, New York University, New York, NY, USA
| | | | | | | | | | | | - Rahul Satija
- New York Genome Center, New York, NY, USA.
- Center for Genomics and Systems Biology, New York University, New York, NY, USA.
| |
Collapse
|
19
|
Hoffman GE, Roussos P. Fast, flexible analysis of differences in cellular composition with crumblr. RESEARCH SQUARE 2025:rs.3.rs-5921338. [PMID: 40060050 PMCID: PMC11888541 DOI: 10.21203/rs.3.rs-5921338/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/21/2025]
Abstract
Changes in cell type composition play an important role in human health and disease. Recent advances in single-cell technology have enabled the measurement of cell type composition at increasing cell lineage resolution across large cohorts of individuals. Yet this raises new challenges for statistical analysis of these compositional data to identify changes in cell type frequency. We introduce crumblr (DiseaseNeurogenomics.github.io/crumblr), a scalable statistical method for analyzing count ratio data using precision-weighted linear mixed models incorporating random effects for complex study designs. Uniquely, crumblr performs statistical testing at multiple levels of the cell lineage hierarchy using a multivariate approach to increase power over tests of one cell type. In simulations, crumblr increases power compared to existing methods while controlling the false positive rate. We demonstrate the application of crumblr to published single-cell RNA-seq datasets for aging, tuberculosis infection in T cells, bone metastases from prostate cancer, and SARS-CoV-2 infection.
Collapse
Affiliation(s)
- Gabriel E Hoffman
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, New York
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| | - Panos Roussos
- Center for Disease Neurogenomics, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Center for Precision Medicine and Translational Therapeutics, James J. Peters VA Medical Center, Bronx, New York
- Mental Illness Research, Education and Clinical Centers, James J. Peters VA Medical Center, Bronx, New York
| |
Collapse
|
20
|
Sivakumar S, Jainarayanan A, Arbe-Barnes E, Sharma PK, Leathlobhair MN, Amin S, Reiss DJ, Heij L, Hegde S, Magen A, Tucci F, Sun B, Wu S, Anand NM, Slawinski H, Revale S, Nassiri I, Webber J, Hoeltzel GD, Frampton AE, Wiltberger G, Neumann U, Charlton P, Spiers L, Elliott T, Wang M, Couto S, Lila T, Sivakumar PV, Ratushny AV, Middleton MR, Peppa D, Fairfax B, Merad M, Dustin ML, Abu-Shah E, Bashford-Rogers R. Distinct immune cell infiltration patterns in pancreatic ductal adenocarcinoma (PDAC) exhibit divergent immune cell selection and immunosuppressive mechanisms. Nat Commun 2025; 16:1397. [PMID: 39915477 PMCID: PMC11802853 DOI: 10.1038/s41467-024-55424-2] [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: 09/28/2023] [Accepted: 12/11/2024] [Indexed: 02/09/2025] Open
Abstract
Pancreatic ductal adenocarcinoma has a dismal prognosis. A comprehensive analysis of single-cell multi-omic data from matched tumour-infiltrated CD45+ cells and peripheral blood in 12 patients, and two published datasets, reveals a complex immune infiltrate. Patients have either a myeloid-enriched or adaptive-enriched tumour microenvironment. Adaptive immune cell-enriched is intrinsically linked with highly distinct B and T cell clonal selection, diversification, and differentiation. Using TCR data, we see the largest clonal expansions in CD8 effector memory, senescent cells, and highly activated regulatory T cells which are induced within the tumour from naïve cells. We identify pathways that potentially lead to a suppressive microenvironment, including investigational targets TIGIT/PVR and SIRPA/CD47. Analysis of patients from the APACT clinical trial shows that myeloid enrichment had a shorter overall survival compared to those with adaptive cell enrichment. Strategies for rationale therapeutic development in this disease include boosting of B cell responses, targeting immunosuppressive macrophages, and specific Treg cell depletion approaches.
Collapse
Affiliation(s)
- Shivan Sivakumar
- Department of Oncology, University of Oxford, Oxford, OX3 7LF, UK.
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Dr, Headington, Oxford, OX3 7FY, UK.
- Department of Immunology and Immunotherapy, School of Infection, Inflammation and Immunology, College of Medicine and Health, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Ashwin Jainarayanan
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Dr, Headington, Oxford, OX3 7FY, UK
- Institute of Developmental and Regenerative Medicine (IDRM), Old Road Campus, Old Rd, Roosevelt Dr, Headington, University of Oxford, Oxford, OX3 7TY, UK
| | - Edward Arbe-Barnes
- Oxford University Clinical Academic Graduate School, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK
- UCL Institute of Immunity & Transplantation, The Pears Building, Pond Street, London, NW3 2PP, UK
| | | | - Maire Ni Leathlobhair
- Department of Microbiology, Trinity College, Dublin, Ireland
- Oxford Big Data Institute, Old Road Campus, University of Oxford, Oxford, OX3 7LF, UK
| | - Sakina Amin
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | | | - Lara Heij
- GROW School for Oncology and Developmental Biology, Department of Pathology, Maastricht University Medical Center, Maastricht, The Netherlands
- Department of Surgery and Transplantation, University Hospital RWTH Aachen, Aachen, Germany
| | - Samarth Hegde
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Assaf Magen
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Felicia Tucci
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
- Oxford Cancer Centre, Oxford, UK
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Bo Sun
- Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, OX3 7LD, UK
| | - Shihong Wu
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
- Oxford Cancer Centre, Oxford, UK
| | | | - Hubert Slawinski
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Santiago Revale
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Isar Nassiri
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Jonathon Webber
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Dr, Headington, Oxford, OX3 7FY, UK
| | - Gerard D Hoeltzel
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK
| | - Adam E Frampton
- Minimal Access Therapy Training Unit (MATTU), Leggett Building, University of Surrey, Daphne Jackson Road, Guildford, GU2 7WG, UK
- Department of Hepato-Pancreato-Biliary (HPB) Surgery, Royal Surrey County Hospital, Egerton Road, Guildford, GU2 7XX, UK
- Targeted Cancer Therapy Unit, Department of Clinical and Experimental Medicine, Faculty of Health and Medical Science, University of Surrey, Guildford, GU2 7WG, UK
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital Campus, London, W12 0NN, UK
| | - Georg Wiltberger
- Department of General, Visceral, and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
| | - Ulf Neumann
- Department of General, Visceral, and Transplantation Surgery, University Hospital of RWTH Aachen, Aachen, Germany
- Department of Surgery Maastricht University Medical Center (MUMC), Maastricht, The Netherlands
| | - Philip Charlton
- Department of Oncology, University of Oxford, Oxford, OX3 7LF, UK
| | - Laura Spiers
- Department of Oncology, University of Oxford, Oxford, OX3 7LF, UK
| | - Tim Elliott
- Centre for Immuno-oncology, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Maria Wang
- Bristol-Myers Squibb, Seattle, Seattle, WA, USA
| | - Suzana Couto
- Neomorph, Inc., 5590 Morehouse Dr, San Diego, CA, USA
| | - Thomas Lila
- Bristol-Myers Squibb, Seattle, Seattle, WA, USA
| | | | | | - Mark R Middleton
- Department of Oncology, University of Oxford, Oxford, OX3 7LF, UK
| | - Dimitra Peppa
- UCL Institute of Immunity & Transplantation, The Pears Building, Pond Street, London, NW3 2PP, UK
- Nuffield Department of Medicine, Old Road Campus, University of Oxford, Oxford, OX3 7BN, UK
| | - Benjamin Fairfax
- Department of Oncology, University of Oxford, Oxford, OX3 7LF, UK
| | - Miriam Merad
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, 1 Gustave L. Levy Pl, New York, NY, 10029, USA
| | - Michael L Dustin
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Dr, Headington, Oxford, OX3 7FY, UK
- Chinese Academy of Medical Sciences Oxford Institute, Nuffield Department of Medicine, University of Oxford, Oxford, OX3 7BN, UK
| | - Enas Abu-Shah
- Kennedy Institute of Rheumatology, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Roosevelt Dr, Headington, Oxford, OX3 7FY, UK.
- Sir William Dunn School of Pathology, South Parks Road, University of Oxford, Oxford, OX1 3RE, UK.
| | - Rachael Bashford-Rogers
- Department of Biochemistry, South Parks Road, University of Oxford, Oxford, OX1 3QU, UK.
- Oxford Cancer Centre, Oxford, UK.
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK.
| |
Collapse
|
21
|
Coates ML, Richoz N, Tuong ZK, Bowyer GS, Lee CYC, Ferdinand JR, Gillman E, McClure M, Dratva L, Teichmann SA, Jayne DR, Di Marco Barros R, Stewart BJ, Clatworthy MR. Temporal profiling of human lymphoid tissues reveals coordinated defense against viral challenge. Nat Immunol 2025; 26:215-229. [PMID: 39890933 PMCID: PMC11785532 DOI: 10.1038/s41590-024-02064-9] [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: 02/14/2023] [Accepted: 12/10/2024] [Indexed: 02/03/2025]
Abstract
Adaptive immunity is generated in lymphoid organs, but how these structures defend themselves during infection in humans is unknown. The nasal epithelium is a major site of viral entry, with adenoid nasal-associated lymphoid tissue (NALT) generating early adaptive responses. In the present study, using a nasopharyngeal biopsy technique, we investigated longitudinal immune responses in NALT after a viral challenge, using severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection as a natural experimental model. In acute infection, infiltrating monocytes formed a subepithelial and perifollicular shield, recruiting neutrophil extracellular trap-forming neutrophils, whereas tissue macrophages expressed pro-repair molecules during convalescence to promote the restoration of tissue integrity. Germinal center B cells expressed antiviral transcripts that inversely correlated with fate-defining transcription factors. Among T cells, tissue-resident memory CD8 T cells alone showed clonal expansion and maintained cytotoxic transcriptional programs into convalescence. Together, our study provides unique insights into how human nasal adaptive immune responses are generated and sustained in the face of viral challenge.
Collapse
Affiliation(s)
- Matthew L Coates
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | - Nathan Richoz
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | - Zewen K Tuong
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - Georgina S Bowyer
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | - Colin Y C Lee
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
| | - John R Ferdinand
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
| | - Eleanor Gillman
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | - Mark McClure
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | - Lisa Dratva
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
- Cambridge Stem Cell Institute, Cambridge, UK
| | - Sarah A Teichmann
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK
- Cambridge Stem Cell Institute, Cambridge, UK
| | - David R Jayne
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | | | - Benjamin J Stewart
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK
| | - Menna R Clatworthy
- Department of Medicine, Molecular Immunity Unit, University of Cambridge, Cambridge, UK.
- Cambridge Institute for Therapeutic Immunology and Infectious Diseases, Cambridge, UK.
- Cellular Genetics, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
22
|
Lagattuta KA, Kohlgruber AC, Abdelfattah NS, Nathan A, Rumker L, Birnbaum ME, Elledge SJ, Raychaudhuri S. The T cell receptor sequence influences the likelihood of T cell memory formation. Cell Rep 2025; 44:115098. [PMID: 39731734 PMCID: PMC11785489 DOI: 10.1016/j.celrep.2024.115098] [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: 07/31/2024] [Revised: 09/19/2024] [Accepted: 12/02/2024] [Indexed: 12/30/2024] Open
Abstract
The amino acid sequence of the T cell receptor (TCR) varies between T cells of an individual's immune system. Particular TCR residues nearly guarantee mucosal-associated invariant T (MAIT) and natural killer T (NKT) cell transcriptional fates. To define how the TCR sequence affects T cell fates, we analyze the paired αβTCR sequence and transcriptome of 961,531 single cells. We find that hydrophobic complementarity-determining region (CDR)3 residues promote regulatory T cell fates in both the CD8 and CD4 lineages. Most strikingly, we find a set of TCR sequence features that promote the T cell transition from naive to memory. We quantify the extent of these features through our TCR scoring function "TCR-mem." Using TCR transduction experiments, we demonstrate that increased TCR-mem promotes T cell activation, even among T cells that recognize the same antigen. Our results reveal a common set of TCR sequence features that enable T cell activation and immunological memory.
Collapse
MESH Headings
- Immunologic Memory/immunology
- Animals
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/chemistry
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Mice
- Memory T Cells/immunology
- Amino Acid Sequence
- Lymphocyte Activation/immunology
- Complementarity Determining Regions/immunology
- Mice, Inbred C57BL
- Receptors, Antigen, T-Cell, alpha-beta
- CD8-Positive T-Lymphocytes/immunology
Collapse
Affiliation(s)
- Kaitlyn A Lagattuta
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Ayano C Kohlgruber
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Nouran S Abdelfattah
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA
| | - Aparna Nathan
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Laurie Rumker
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michael E Birnbaum
- Koch Institute for Integrative Cancer Research, Cambridge, MA, USA; Department of Biomedical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA; Ragon Institute of MGH, MIT, and Harvard, Cambridge, MA, USA
| | - Stephen J Elledge
- Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Genetics, Harvard Medical School, Boston, MA, USA; Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Soumya Raychaudhuri
- Center for Data Sciences, Brigham and Women's Hospital, Boston, MA, USA; Division of Genetics, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA; Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA; Program in Medical and Population Genetics, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
23
|
Hao S, Tomic I, Lindsey BB, Jagne YJ, Hoschler K, Meijer A, Quiroz JMC, Meade P, Sano K, Peno C, Costa-Martins AG, Bogaert D, Kampmann B, Nakaya H, Krammer F, de Silva TI, Tomic A. Integrative Mapping of Pre-existing Immune Landscapes for Vaccine Response Prediction. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634302. [PMID: 39896552 PMCID: PMC11785181 DOI: 10.1101/2025.01.22.634302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Predicting individual vaccine responses remains a significant challenge due to the complexity and variability of immune processes. To address this gap, we developed immunaut, an open-source, data-driven framework implemented as an R package specifically designed for all systems vaccinologists seeking to analyze and predict immunological outcomes across diverse vaccination settings. Leveraging one of the most comprehensive live attenuated influenza vaccine (LAIV) datasets to date - 244 Gambian children enrolled in a phase 4 immunogenicity study - immunaut integrates humoral, mucosal, cellular, transcriptomic, and microbiological parameters collected before and after vaccination, providing an unprecedentedly holistic view of LAIV-induced immunity. Through advanced dimensionality reduction, clustering, and predictive modeling, immunaut identifies distinct immunophenotypic responder profiles and their underlying baseline determinants. In this study, immunaut delineated three immunophenotypes: (1) CD8 T-cell responders, marked by strong baseline mucosal immunity and extensive prior influenza virus exposure that boosts memory CD8 T-cell responses, without generating influenza virus-specific antibody responses; (2) Mucosal responders, characterized by pre-existing systemic influenza A virus immunity (specifically to H3N2) and stable epithelial integrity, leading to potent mucosal IgA expansions and subsequent seroconversion to influenza B virus; and (3) Systemic, broad influenza A virus responders, who start with relatively naive immunity and leverage greater initial viral replication to drive broad systemic antibody responses against multiple influenza A virus variants beyond those included in the LAIV vaccine. By integrating pathway-level analysis, model-derived contribution scores, and hierarchical decision rules, immunaut elucidates how distinct immunological landscapes shape each response trajectory and how key baseline features, including pre-existing immunity, mucosal preparedness, and cellular support, dictate vaccine outcomes. Collectively, these findings emphasize the power of integrative, predictive frameworks to advance precision vaccinology, and highlight immunaut as a versatile, community-available resource for optimizing immunization strategies across diverse populations and vaccine platforms.
Collapse
Affiliation(s)
- Stephanie Hao
- Atomic lab, The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University; Boston, MA, US
| | - Ivan Tomic
- Atomic lab, The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University; Boston, MA, US
| | - Benjamin B Lindsey
- The Florey Institute of Infection and NIHR Sheffield Biomedical Research Centre, University of Sheffield; Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road; Sheffield, UK
| | - Ya Jankey Jagne
- Vaccines and Immunity Theme, Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine; Fajara, The Gambia
| | - Katja Hoschler
- Respiratory Virus Unit, UK Health Security Agency; London, UK
| | - Adam Meijer
- National Institute for Public Health and the Environment; Bilthoven, The Netherlands
| | - Juan Manuel Carreño Quiroz
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Philip Meade
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Kaori Sano
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Chikondi Peno
- Centre for Inflammation Research, University of Edinburgh; Edinburgh, UK
| | - André G Costa-Martins
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo; São Paulo, Brazil
- Micromanufacturing Laboratory, Institute for Technological Research, São Paulo, Brazil
| | - Debby Bogaert
- Centre for Inflammation Research, University of Edinburgh; Edinburgh, UK
| | - Beate Kampmann
- Vaccines and Immunity Theme, London School of Hygiene & Tropical Medicine; London, UK
- Charité Centre for Global Health; Berlin, Germany
| | - Helder Nakaya
- Department of Clinical and Toxicological Analyses, School of Pharmaceutical Sciences, University of São Paulo; São Paulo, Brazil
- Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY, US
- Center for Vaccine Research and Pandemic Preparedness (C-VaRPP), Icahn School of Medicine at Mount Sinai, New York, NY, US
- Ignaz Semmelweis Institute, Interuniversity Institute for Infection Research, Medical University of Vienna, Vienna, Austria
- Department of Pathology, Molecular and Cell-Based Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, US
| | - Thushan I de Silva
- The Florey Institute of Infection and NIHR Sheffield Biomedical Research Centre, University of Sheffield; Sheffield, UK
- Division of Clinical Medicine, School of Medicine and Population Health, University of Sheffield, Beech Hill Road; Sheffield, UK
- Vaccines and Immunity Theme, London School of Hygiene & Tropical Medicine; London, UK
| | - Adriana Tomic
- Atomic lab, The National Emerging Infectious Diseases Laboratories (NEIDL), Boston University; Boston, MA, US
- Department of Virology, Immunology & Microbiology, Boston University Medical School; Boston, MA, US
- Biomedical Engineering, Boston University, College of Engineering; Boston, MA, US
| |
Collapse
|
24
|
Guerrera G, Sambucci M, Timperi E, Picozza M, Misiti A, Placido R, Corbisiero S, D’Orso S, Termine A, Fabrizio C, Gargano F, Eleuteri S, Marchioni L, Bordoni V, Coppola L, Iannetta M, Agrati C, Borsellino G, Battistini L. Identification of an immunological signature of long COVID syndrome. Front Immunol 2025; 15:1502937. [PMID: 39845978 PMCID: PMC11750999 DOI: 10.3389/fimmu.2024.1502937] [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: 09/27/2024] [Accepted: 12/16/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction Acute COVID-19 infection causes significant alterations in the innate and adaptive immune systems. While most individuals recover naturally, some develop long COVID (LC) syndrome, marked by persistent or new symptoms weeks to months after SARS-CoV-2 infection. Despite its prevalence, there are no clinical tests to distinguish LC patients from those fully recovered. Understanding the immunological basis of LC is essential for improving diagnostic and treatment approaches. Methods We performed deep immunophenotyping and functional assays to examine the immunological profiles of LC patients, individuals with active COVID-19, recovered patients, and healthy donors. This analysis assessed both innate and adaptive immune features, identifying potential biomarkers for LC syndrome. A Binomial Generalized Linear Model (BGLM) was used to pinpoint immune features characterizing LC. Results COVID-19 patients exhibited depletion of innate immune cell subsets, including plasmacytoid and conventional dendritic cells, classical, non-classical, and intermediate monocytes, and monocyte-derived inflammatory dendritic cells. Elevated basal inflammation was observed in COVID-19 patients compared to LC patients, whose immune profiles were closer to those of healthy donors and recovered individuals. However, LC patients displayed persistent immune alterations, including reduced T cell subsets (CD4, CD8, Tregs) and switched memory B cells, similar to COVID-19 patients. Through BGLM, a unique adaptive immune signature for LC was identified, featuring memory CD8 and gd T cells with low proliferative capacity and diminished expression of activation and homing receptors. Discussion The findings highlight a unique immunological signature associated with LC syndrome, characterized by persistent adaptive immune dysregulation. While LC patients displayed recovery in innate immune profiles comparable to healthy and Recovered individuals, deficits in T cell and memory B cell populations were evident, differentiating LC from full recovery. These findings provide insights into LC pathogenesis and may support the development of diagnostic tools and targeted therapies.
Collapse
Affiliation(s)
| | - Manolo Sambucci
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | - Mario Picozza
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Andrea Misiti
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
- Data Science Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Roberta Placido
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | - Silvia D’Orso
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Andrea Termine
- Data Science Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Carlo Fabrizio
- Data Science Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | | | - Sharon Eleuteri
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| | - Luisa Marchioni
- UOS Terapia Intensiva Postoperatoria e Assistenza Subintensiva, National Institute for Infectious Diseases IRCCS Lazzaro Spallanzani, Rome, Italy
| | - Veronica Bordoni
- Unit of Pathogen specific Immunity, Research Area of Hematology and Oncology, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Luigi Coppola
- Infectious disease Clinic, Policlinico Tor Vergata of Rome, Rome, Italy
| | - Marco Iannetta
- Department of Systems Medicine, Infectious Disease Clinic, Tor Vergata University, Rome, Italy
| | - Chiara Agrati
- Unit of Pathogen specific Immunity, Research Area of Hematology and Oncology, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | | | - Luca Battistini
- Neuroimmunology Unit, Santa Lucia Foundation IRCCS, Rome, Italy
| |
Collapse
|
25
|
Margiotta RG, Sozio E, Del Ben F, Beltrami AP, Cesselli D, Comar M, Devito A, Fabris M, Curcio F, Tascini C, Sanguinetti G. Investigating the relationship between the immune response and the severity of COVID-19: a large-cohort retrospective study. Front Immunol 2025; 15:1452638. [PMID: 39845955 PMCID: PMC11750771 DOI: 10.3389/fimmu.2024.1452638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 11/11/2024] [Indexed: 01/24/2025] Open
Abstract
The COVID-19 pandemic has left an indelible mark globally, presenting numerous challenges to public health. This crisis, while disruptive and impactful, has provided a unique opportunity to gather precious clinical data extensively. In this observational, case-control study, we utilized data collected at the Azienda Sanitaria Universitaria Friuli Centrale, Italy, to comprehensively characterize the immuno-inflammatory features in COVID-19 patients. Specifically, we employed multicolor flow cytometry, cytokine assays, and inflammatory biomarkers to elucidate the interplay between the infectious agent and the host's immune status. We characterized immuno-inflammatory profiles within the first 72 hours of hospital admission, stratified by age, disease severity, and time elapsed since symptom onset. Our findings indicate that patients admitted to the hospital shortly after symptom onset exhibit a distinct pattern compared to those who arrive later, characterized by a more active immune response and heightened cytokine activity, but lower markers of tissue damage. We used univariate and multivariate logistic regression models to identify informative markers for outcome severity. Predictors incorporating the immuno-inflammatory features significantly outperformed standard baselines, identifying up to 59% of patients with positive outcomes while maintaining a false omission rate as low as 4%. Overall, our study sheds light on the immuno-inflammatory aspects observed in COVID-19 patients prior to vaccination, providing insights for guiding the clinical management of first-time infections by a novel virus.
Collapse
Affiliation(s)
| | - Emanuela Sozio
- Infectious Disease Unit, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
| | - Fabio Del Ben
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Antonio Paolo Beltrami
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Daniela Cesselli
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Marco Comar
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | | | - Martina Fabris
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Francesco Curcio
- Department of Medicine (DMED), University of Udine, Udine, Italy
- Department of Laboratory Medicine, ASU FC, Udine, Italy
| | - Carlo Tascini
- Infectious Disease Unit, Azienda Sanitaria Universitaria Friuli Centrale (ASU FC), Udine, Italy
- Department of Medicine (DMED), University of Udine, Udine, Italy
| | - Guido Sanguinetti
- Physics Department, International School for Advanced Studies (SISSA), Trieste, Italy
| |
Collapse
|
26
|
Corvino D, Batstone M, Hughes BGM, Kempchen T, Ng SS, Salim N, Schneppenheim F, Rommel D, Kumar A, Pearson S, Madore J, Koufariotis LT, Steinheuer LM, Pathirana D, Thurley K, Hölzel M, Borcherding N, Braun M, Bald T. Type I Interferon Drives a Cellular State Inert to TCR-Stimulation and Could Impede Effective T-Cell Differentiation in Cancer. Eur J Immunol 2025; 55:e202451371. [PMID: 39529512 PMCID: PMC11739669 DOI: 10.1002/eji.202451371] [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: 07/09/2024] [Revised: 10/14/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Head and neck squamous cell carcinoma (HNSCC) is linked to human papillomavirus (HPV) infection. HPV-positive and HPV-negative HNSCC exhibit distinct molecular and clinical characteristics. Although checkpoint inhibitors have shown efficiency in recurrent/metastatic HNSCC, response variability persists regardless of HPV status. This study aimed to explore the CD8+ T-cell landscape in HPV-negative HNSCC. METHODS We performed simultaneous single-cell RNA and TCR sequencing of CD8+ tumor-infiltrating lymphocytes (TILs) from treatment-naïve HPV-negative HNSCC patients. Additionally, cells were stimulated ex vivo, which allowed for the tracking of clonal transcriptomic responses. RESULTS Our analysis identified a subset of CD8+ TILs highly enriched for interferon-stimulated genes (ISG). TCR analysis revealed ISG cells are clonally related to a population of granzyme K (GZMK)-expressing cells. However, unlike GZMK cells, which exhibited rapid effector-like phenotypes following stimulation, ISG cells were transcriptionally inert. Additionally, ISG cells showed specific enrichment within tumor and were found across multiple tumor entities. CONCLUSIONS ISG-enriched CD8+ TILs are a consistent feature of various tumor entities. These cells are poorly understood but possess characteristics that may impact antitumor immunity. Understanding the unique properties and functionality of ISG cells could offer innovative treatment approaches to improve patient outcomes in HPV-negative HNSCC and other cancer types.
Collapse
Affiliation(s)
- Dillon Corvino
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Martin Batstone
- Royal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Brett G. M Hughes
- Royal Brisbane and Women's HospitalBrisbaneQueenslandAustralia
- Faculty of MedicineUniversity of QueenslandBrisbaneQueenslandAustralia
| | - Tim Kempchen
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Susanna S Ng
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Nazhifah Salim
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | | | - Denise Rommel
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Ananthi Kumar
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Sally Pearson
- QIMR Berghofer Medical Research InstituteHerstonAustralia
| | - Jason Madore
- QIMR Berghofer Medical Research InstituteHerstonAustralia
| | | | - Lisa Maria Steinheuer
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Dilan Pathirana
- Faculty of Mathematics and Natural Sciencesand the Life and Medical Sciences Institute (LIMES)Rheinische Friedrich‐Wilhelms‐Universität BonnBonnGermany
| | - Kevin Thurley
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Michael Hölzel
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| | - Nicholas Borcherding
- Department of Pathology and ImmunologyWashington University School of MedicineSt. LouisMissouriUSA
| | - Matthias Braun
- Department of Pediatric Hematology, Oncology and ImmunodeficiencyUniversity Childrens Hospital of the Justus‐Liebig University GießenGießenGermany
| | - Tobias Bald
- Tumor‐ImmunobiologyInstitute for Experimental OncologyUniversity Hospital BonnBonnGermany
| |
Collapse
|
27
|
Hrovatin K, Sikkema L, Shitov VA, Heimberg G, Shulman M, Oliver AJ, Mueller MF, Ibarra IL, Wang H, Ramírez-Suástegui C, He P, Schaar AC, Teichmann SA, Theis FJ, Luecken MD. Considerations for building and using integrated single-cell atlases. Nat Methods 2025; 22:41-57. [PMID: 39672979 DOI: 10.1038/s41592-024-02532-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 10/22/2024] [Indexed: 12/15/2024]
Abstract
The rapid adoption of single-cell technologies has created an opportunity to build single-cell 'atlases' integrating diverse datasets across many laboratories. Such atlases can serve as a reference for analyzing and interpreting current and future data. However, it has become apparent that atlasing approaches differ, and the impact of these differences are often unclear. Here we review the current atlasing literature and present considerations for building and using atlases. Importantly, we find that no one-size-fits-all protocol for atlas building exists, but rather we discuss context-specific considerations and workflows, including atlas conceptualization, data collection, curation and integration, atlas evaluation and atlas sharing. We further highlight the benefits of integrated atlases for analyses of new datasets and deriving biological insights beyond what is possible from individual datasets. Our overview of current practices and associated recommendations will improve the quality of atlases to come, facilitating the shift to a unified, reference-based understanding of single-cell biology.
Collapse
Affiliation(s)
- Karin Hrovatin
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Lisa Sikkema
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Vladimir A Shitov
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany
| | - Graham Heimberg
- Department of OMNI Bioinformatics, Genentech, South San Francisco, CA, USA
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
| | - Maiia Shulman
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany
| | - Amanda J Oliver
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Michaela F Mueller
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Ignacio L Ibarra
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
| | - Hanchen Wang
- Department of Biological Research | AI Development, Genentech, South San Francisco, CA, USA
- Department of Computer Science, Stanford University, Palo Alto, CA, USA
| | - Ciro Ramírez-Suástegui
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Peng He
- Department of Pathology, University of California, San Francisco, San Francisco, CA, USA
| | - Anna C Schaar
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany
- TUM School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Theory of Condensed Matter Group, Department of Physics, Cavendish Laboratory, University of Cambridge, Cambridge, UK
- Cambridge Stem Cell Institute and Department of Medicine, University of Cambridge, Cambridge, UK
- CIFAR MacMillan Multiscale Human Programme, Toronto, Ontario, Canada
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Freising, Germany.
- Department of Mathematics, Technical University of Munich, Garching, Germany.
| | - Malte D Luecken
- Department of Computational Health, Institute of Computational Biology, Helmholtz Zentrum München, Munich, Germany.
- Comprehensive Pneumology Center (CPC) with the CPC-M bioArchive / Institute of Lung Health and Immunity (LHI), Helmholtz Zentrum München; Member of the German Center for Lung Research (DZL), Munich, Germany.
| |
Collapse
|
28
|
Yang Y, Miller H, Byazrova MG, Cndotti F, Benlagha K, Camara NOS, Shi J, Forsman H, Lee P, Yang L, Filatov A, Zhai Z, Liu C. The characterization of CD8 + T-cell responses in COVID-19. Emerg Microbes Infect 2024; 13:2287118. [PMID: 37990907 PMCID: PMC10786432 DOI: 10.1080/22221751.2023.2287118] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/19/2023] [Indexed: 11/23/2023]
Abstract
This review gives an overview of the protective role of CD8+ T cells in SARS-CoV-2 infection. The cross-reactive responses intermediated by CD8+ T cells in unexposed cohorts are described. Additionally, the relevance of resident CD8+ T cells in the upper and lower airway during infection and CD8+ T-cell responses following vaccination are discussed, including recent worrisome breakthrough infections and variants of concerns (VOCs). Lastly, we explain the correlation between CD8+ T cells and COVID-19 severity. This review aids in a deeper comprehension of the association between CD8+ T cells and SARS-CoV-2 and broadens a vision for future exploration.
Collapse
Affiliation(s)
- Yuanting Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
- Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, People’s Republic of China
| | - Heather Miller
- Cytek Biosciences, R&D Clinical Reagents, Fremont, CA, USA
| | - Maria G. Byazrova
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Fabio Cndotti
- Division of Immunology and Allergy, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Kamel Benlagha
- Institut de Recherche Saint-Louis, Université de Paris, Paris, France
| | - Niels Olsen Saraiva Camara
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo (USP), São Paulo, Brazil
| | - Junming Shi
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Huamei Forsman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pamela Lee
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Lu Yang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| | - Alexander Filatov
- Laboratory of Immunochemistry, National Research Center Institute of Immunology, Federal Medical Biological Agency of Russia, Moscow, Russia
| | - Zhimin Zhai
- Department of Hematology, The Second Hospital of Anhui Medical University, Hefei, People’s Republic of China
| | - Chaohong Liu
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, Hubei, People’s Republic of China
| |
Collapse
|
29
|
Rosenheim J, Gupta RK, Thakker C, Mann T, Bell LCK, Broderick CM, Madon K, Papargyris L, Dayananda P, Kwok AJ, Greenan-Barrett J, Wagstaffe HR, Conibear E, Fenn J, Hakki S, Lindeboom RGH, Dratva LM, Lemetais B, Weight CM, Venturini C, Kaforou M, Levin M, Kalinova M, Mann AJ, Catchpole A, Knight JC, Nikolić MZ, Teichmann SA, Killingley B, Barclay W, Chain BM, Lalvani A, Heyderman RS, Chiu C, Noursadeghi M. SARS-CoV-2 human challenge reveals biomarkers that discriminate early and late phases of respiratory viral infections. Nat Commun 2024; 15:10434. [PMID: 39616162 PMCID: PMC11608262 DOI: 10.1038/s41467-024-54764-3] [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: 07/21/2023] [Accepted: 11/19/2024] [Indexed: 02/27/2025] Open
Abstract
Blood transcriptional biomarkers of acute viral infections typically reflect type 1 interferon (IFN) signalling, but it is not known whether there are biological differences in their regulation that can be leveraged for distinct translational applications. We use high frequency sampling in the SARS-CoV-2 human challenge model to show induction of IFN-stimulated gene (ISG) expression with different temporal and cellular profiles. MX1 gene expression correlates with a rapid and transient wave of ISG expression across all cell types, which may precede PCR detection of replicative infection. Another ISG, IFI27, shows a delayed but sustained response restricted to myeloid cells, attributable to gene and cell-specific epigenetic regulation. These findings are reproducible in experimental and naturally acquired infections with influenza, respiratory syncytial virus and rhinovirus. Blood MX1 expression is superior to IFI27 expression for diagnosis of early infection, as a correlate of viral load and for discrimination of virus culture positivity. Therefore, MX1 expression offers potential to stratify patients for antiviral therapy or infection control interventions. Blood IFI27 expression is superior to MX1 expression for diagnostic accuracy across the time course of symptomatic infection and thereby, offers higher diagnostic yield for respiratory virus infections that incur a delay between transmission and testing.
Collapse
Affiliation(s)
- Joshua Rosenheim
- Division of Infection and Immunity, University College London, London, UK
| | - Rishi K Gupta
- Institute of Health Informatics, University College London, London, UK
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Clare Thakker
- Division of Infection and Immunity, University College London, London, UK
| | - Tiffeney Mann
- Division of Infection and Immunity, University College London, London, UK
| | - Lucy C K Bell
- Division of Infection and Immunity, University College London, London, UK
| | | | - Kieran Madon
- NIHR Health Protection Research Unit in Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, UK
| | - Loukas Papargyris
- Department of Infectious Disease, Imperial College London, London, UK
| | - Pete Dayananda
- Department of Infectious Disease, Imperial College London, London, UK
| | - Andrew J Kwok
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
- Department of Medicine and Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong
| | | | - Helen R Wagstaffe
- Department of Infectious Disease, Imperial College London, London, UK
| | - Emily Conibear
- NIHR Health Protection Research Unit in Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, UK
| | - Joe Fenn
- NIHR Health Protection Research Unit in Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, UK
| | - Seran Hakki
- NIHR Health Protection Research Unit in Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, UK
| | | | - Lisa M Dratva
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Briac Lemetais
- Division of Infection and Immunity, University College London, London, UK
| | - Caroline M Weight
- Division of Infection and Immunity, University College London, London, UK
| | - Cristina Venturini
- Infection, Immunity and Inflammation Department, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Myrsini Kaforou
- Department of Infectious Disease, Imperial College London, London, UK
| | - Michael Levin
- Department of Infectious Disease, Imperial College London, London, UK
| | | | | | | | - Julian C Knight
- Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Marko Z Nikolić
- UCL Respiratory, Division of Medicine, University College London, London, UK
- Department of Respiratory Medicine, University College London Hospitals NHS Foundation Trust, London, UK
| | - Sarah A Teichmann
- Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ben Killingley
- Department of Infectious Diseases, University College London Hospital NHS Foundation Trust, London, UK
| | - Wendy Barclay
- Department of Infectious Disease, Imperial College London, London, UK
| | - Benjamin M Chain
- Division of Infection and Immunity, University College London, London, UK
| | - Ajit Lalvani
- NIHR Health Protection Research Unit in Respiratory Infections, National Heart and Lung Institute, Imperial College London, London, UK
| | - Robert S Heyderman
- Division of Infection and Immunity, University College London, London, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Mahdad Noursadeghi
- Division of Infection and Immunity, University College London, London, UK.
| |
Collapse
|
30
|
Lu J, Xie Y, Li C, Yang J, Fu J. Tensor decomposition reveals trans-regulated gene modules in maize drought response. J Genet Genomics 2024:S1673-8527(24)00285-6. [PMID: 39522680 DOI: 10.1016/j.jgg.2024.10.011] [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: 05/29/2024] [Revised: 10/22/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024]
Abstract
When plants respond to drought stress, dynamic cellular changes occur, accompanied by alterations in gene expression, which often act through trans-regulation. However, the detection of trans-acting genetic variants and networks of genes is challenged by the large number of genes and markers. Using a tensor decomposition method, we identify trans-acting expression quantitative trait loci (trans-eQTLs) linked to gene modules, rather than individual genes, which were associated with maize drought response. Module-to-trait association analysis demonstrates that half of the modules are relevant to drought-related traits. Genome-wide association studies of the expression patterns of each module identify 286 trans-eQTLs linked to drought-responsive modules, the majority of which cannot be detected based on individual gene expression. Notably, the trans-eQTLs located in the regions selected during maize improvement tend towards relatively strong selection. We further prioritize the genes that affect the transcriptional regulation of multiple genes in trans, as exemplified by two transcription factor genes. Our analyses highlight that multidimensional reduction could facilitate the identification of trans-acting variations in gene expression in response to dynamic environments and serve as a promising technique for high-order data processing in future crop breeding.
Collapse
Affiliation(s)
- Jiawen Lu
- State Key Laboratory of Crop Gene Resources and Breeding, National Key Facility for Crop Gene Resources and Genetic Improvement, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Yuxin Xie
- State Key Laboratory of Crop Gene Resources and Breeding, National Key Facility for Crop Gene Resources and Genetic Improvement, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Chunhui Li
- State Key Laboratory of Crop Gene Resources and Breeding, National Key Facility for Crop Gene Resources and Genetic Improvement, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China
| | - Jinliang Yang
- Department of Agronomy and Horticulture, University of Nebraska-Lincoln, Lincoln, NE, 68588, USA
| | - Junjie Fu
- State Key Laboratory of Crop Gene Resources and Breeding, National Key Facility for Crop Gene Resources and Genetic Improvement, Institute of Crop Sciences, Chinese Academy of Agricultural Sciences, Beijing 100081, China.
| |
Collapse
|
31
|
Thomas T, Friedrich M, Rich-Griffin C, Pohin M, Agarwal D, Pakpoor J, Lee C, Tandon R, Rendek A, Aschenbrenner D, Jainarayanan A, Voda A, Siu JHY, Sanches-Peres R, Nee E, Sathananthan D, Kotliar D, Todd P, Kiourlappou M, Gartner L, Ilott N, Issa F, Hester J, Turner J, Nayar S, Mackerodt J, Zhang F, Jonsson A, Brenner M, Raychaudhuri S, Kulicke R, Ramsdell D, Stransky N, Pagliarini R, Bielecki P, Spies N, Marsden B, Taylor S, Wagner A, Klenerman P, Walsh A, Coles M, Jostins-Dean L, Powrie FM, Filer A, Travis S, Uhlig HH, Dendrou CA, Buckley CD. A longitudinal single-cell atlas of anti-tumour necrosis factor treatment in inflammatory bowel disease. Nat Immunol 2024; 25:2152-2165. [PMID: 39438660 PMCID: PMC11519010 DOI: 10.1038/s41590-024-01994-8] [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: 06/27/2023] [Accepted: 09/18/2024] [Indexed: 10/25/2024]
Abstract
Precision medicine in immune-mediated inflammatory diseases (IMIDs) requires a cellular understanding of treatment response. We describe a therapeutic atlas for Crohn's disease (CD) and ulcerative colitis (UC) following adalimumab, an anti-tumour necrosis factor (anti-TNF) treatment. We generated ~1 million single-cell transcriptomes, organised into 109 cell states, from 216 gut biopsies (41 subjects), revealing disease-specific differences. A systems biology-spatial analysis identified granuloma signatures in CD and interferon (IFN)-response signatures localising to T cell aggregates and epithelial damage in CD and UC. Pretreatment differences in epithelial and myeloid compartments were associated with remission outcomes in both diseases. Longitudinal comparisons demonstrated disease progression in nonremission: myeloid and T cell perturbations in CD and increased multi-cellular IFN signalling in UC. IFN signalling was also observed in rheumatoid arthritis (RA) synovium with a lymphoid pathotype. Our therapeutic atlas represents the largest cellular census of perturbation with the most common biologic treatment, anti-TNF, across multiple inflammatory diseases.
Collapse
Affiliation(s)
- Tom Thomas
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Matthias Friedrich
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | | | - Mathilde Pohin
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Devika Agarwal
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Julia Pakpoor
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Centre for Human Genetics, University of Oxford, Oxford, UK
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Carl Lee
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Ruchi Tandon
- University College London Hospitals NHS Foundation Trust, London, UK
| | - Aniko Rendek
- Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Dominik Aschenbrenner
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | | | - Alexandru Voda
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | | | - Eloise Nee
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Dharshan Sathananthan
- University of Adelaide, Adelaide, Australia
- Lyell McEwin Hospital, Adelaide, Australia
| | - Dylan Kotliar
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Peter Todd
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | | | - Lisa Gartner
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Nicholas Ilott
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fadi Issa
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Joanna Hester
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK
| | - Jason Turner
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Saba Nayar
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Jonas Mackerodt
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Fan Zhang
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
- Center for Health AI, University of Colorado Anschutz, Anschutz, CO, USA
| | - Anna Jonsson
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Michael Brenner
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Soumya Raychaudhuri
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | | | | | | | | | | | - Noah Spies
- Celsius Therapeutics, Cambridge, MA, USA
| | - Brian Marsden
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Stephen Taylor
- Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Allon Wagner
- Department of Electrical Engineering and Computer Science, University of California, Berkeley, Berkeley, CA, USA
- The Center for Computational Biology, University of California, Berkeley, Berkeley, CA, USA
| | - Paul Klenerman
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Alissa Walsh
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK
| | - Mark Coles
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | | | - Fiona M Powrie
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Andrew Filer
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
- National Institute for Health Research (NIHR) Birmingham Biomedical Research Centre and NIHR Clinical Research Facility, University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Birmingham Tissue Analytics, Institute of Translational Medicine, University of Birmingham, Birmingham, UK
| | - Simon Travis
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Holm H Uhlig
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
- Department of Paediatrics, University of Oxford, Oxford, UK.
| | - Calliope A Dendrou
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Centre for Human Genetics, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| | - Christopher D Buckley
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- Translational Gastroenterology & Liver Unit, John Radcliffe Hospital, Headington, Oxford, UK.
- Rheumatology Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|
32
|
Medina MA, Fuentes-Villalobos F, Quevedo C, Aguilera F, Riquelme R, Rioseco ML, Barria S, Pinos Y, Calvo M, Burbulis I, Kossack C, Alvarez RA, Garrido JL, Barria MI. Longitudinal transcriptional changes reveal genes from the natural killer cell-mediated cytotoxicity pathway as critical players underlying COVID-19 progression. eLife 2024; 13:RP94242. [PMID: 39470726 PMCID: PMC11521369 DOI: 10.7554/elife.94242] [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] [Indexed: 10/30/2024] Open
Abstract
Patients present a wide range of clinical severities in response severe acute respiratory syndrome coronavirus 2 infection, but the underlying molecular and cellular reasons why clinical outcomes vary so greatly within the population remains unknown. Here, we report that negative clinical outcomes in severely ill patients were associated with divergent RNA transcriptome profiles in peripheral immune cells compared with mild cases during the first weeks after disease onset. Protein-protein interaction analysis indicated that early-responding cytotoxic natural killer cells were associated with an effective clearance of the virus and a less severe outcome. This innate immune response was associated with the activation of select cytokine-cytokine receptor pathways and robust Th1/Th2 cell differentiation profiles. In contrast, severely ill patients exhibited a dysregulation between innate and adaptive responses affiliated with divergent Th1/Th2 profiles and negative outcomes. This knowledge forms the basis of clinical triage that may be used to preemptively detect high-risk patients before life-threatening outcomes ensue.
Collapse
Affiliation(s)
- Matias A Medina
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
| | | | - Claudio Quevedo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepciónChile
| | - Felipe Aguilera
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Biológicas, Universidad de ConcepciónConcepciónChile
| | - Raul Riquelme
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
- Hospital Dr. Eduardo Schütz SchroederPuerto MonttChile
| | - Maria Luisa Rioseco
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
- Hospital Dr. Eduardo Schütz SchroederPuerto MonttChile
| | - Sebastian Barria
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
- Hospital Dr. Eduardo Schütz SchroederPuerto MonttChile
| | | | - Mario Calvo
- Instituto de Medicina, Facultad de Medicina, Universidad AustralValdiviaChile
| | - Ian Burbulis
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
| | - Camila Kossack
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
| | - Raymond A Alvarez
- Division of Infectious Diseases, Department of Medicine, Immunology Institute, Icahn School of Medicine at Mount SinaiNew YorkUnited States
| | - Jose Luis Garrido
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
| | - Maria Ines Barria
- Facultad de Medicina y Ciencia, Universidad San SebastiánPuerto MonttChile
| |
Collapse
|
33
|
Paran FJ, Oyama R, Khasawneh A, Ai T, Ismanto HS, Sherif AA, Saputri DS, Ono C, Saita M, Takei S, Horiuchi Y, Yagi K, Matsuura Y, Okazaki Y, Takahashi K, Standley DM, Tabe Y, Naito T. BCR, not TCR, repertoire diversity is associated with favorable COVID-19 prognosis. Front Immunol 2024; 15:1405013. [PMID: 39530088 PMCID: PMC11550956 DOI: 10.3389/fimmu.2024.1405013] [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: 03/22/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction The SARS-CoV-2 pandemic has had a widespread and severe impact on society, yet there have also been instances of remarkable recovery, even in critically ill patients. Materials and methods In this study, we used single-cell RNA sequencing to analyze the immune responses in recovered and deceased COVID-19 patients during moderate and critical stages. Results Expanded T cell receptor (TCR) clones were predominantly SARS-CoV-2-specific, but represented only a small fraction of the total repertoire in all patients. In contrast, while deceased patients exhibited monoclonal B cell receptor (BCR) expansions without COVID-19 specificity, survivors demonstrated diverse and specific BCR clones. These findings suggest that neither TCR diversity nor BCR monoclonal expansions are sufficient for viral clearance and subsequent recovery. Differential gene expression analysis revealed that protein biosynthetic processes were enriched in survivors, but that potentially damaging mitochondrial ATP metabolism was activated in the deceased. Conclusion This study underscores that BCR repertoire diversity, but not TCR diversity, correlates with favorable outcomes in COVID-19.
Collapse
MESH Headings
- Humans
- COVID-19/immunology
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- SARS-CoV-2/immunology
- Prognosis
- Male
- Female
- Middle Aged
- Aged
- Single-Cell Analysis
- Adult
- B-Lymphocytes/immunology
Collapse
Affiliation(s)
- Faith Jessica Paran
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Rieko Oyama
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Abdullah Khasawneh
- Leading Center for the Development and Research of Cancer Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Tomohiko Ai
- Department of Clinical Laboratory Medicine, Juntendo University, Urayasu Hospital, Chiba, Japan
| | - Hendra Saputra Ismanto
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Aalaa Alrahman Sherif
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Dianita Susilo Saputri
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Chikako Ono
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Mizue Saita
- Department of General Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Satomi Takei
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Yuki Horiuchi
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Ken Yagi
- Laboratory for Genome Exploration Research Group, RIKEN Genomic Sciences Center, RIKEN, Yokohama, Japan
| | - Yoshiharu Matsuura
- Laboratory of Virus Control, Center for Infectious Disease Education and Research, Osaka University, Osaka, Japan
| | - Yasushi Okazaki
- Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Laboratory for Genome Exploration Research Group, RIKEN Genomic Sciences Center, RIKEN, Yokohama, Japan
| | - Kazuhisa Takahashi
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Respiratory Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Daron M. Standley
- Department of Genome Informatics, Research Institute for Microbial Diseases, Osaka University, Osaka, Japan
| | - Yoko Tabe
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of Clinical Laboratory Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Toshio Naito
- Department of Research Support Utilizing Bioresource Bank, Graduate School of Medicine, Juntendo University, Tokyo, Japan
- Department of General Medicine, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| |
Collapse
|
34
|
Chen W, Jiang S, Li C, Li S, Wang J, Xu R. Potential association between COVID-19 and neurological disorders: analysis of common genes and therapeutics. Front Neurol 2024; 15:1417183. [PMID: 39469068 PMCID: PMC11513677 DOI: 10.3389/fneur.2024.1417183] [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: 04/30/2024] [Accepted: 09/29/2024] [Indexed: 10/30/2024] Open
Abstract
As the COVID-19 pandemic persists, the increasing evidences suggest that the patients with COVID-19 may face the risks of the neurological complications and sequelae. To address this issue, we conducted a comprehensive study aimed at exploring the relationship between COVID-19 and various neurological disorders, with a particular focus on the shared dysregulated genes and the potential therapeutic targets. We selected six neurological disorders for investigation, including Alzheimer's disease, epilepsy, stroke, Parkinson's disease, and the sleep disorders. Through the bioinformatics analysis of the association between these disorders and COVID-19, we aimed to uncover the common molecular mechanisms and the potential treatment pathways. In this study, we utilized the publicly available RNA-Seq and microarray datasets, and employed tools such as Limma and DESeq2 for the differential gene analysis. Through the Gene Ontology and Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis, we explored the common biological features and pathways. Additionally, we focused on analyzing the regulatory roles of miRNA and transcription factors on the shared differentially expressed genes, and predicted the potential drugs interacting with these genes. These analyses contribute to a better understanding of the relationship between COVID-19 and the neurological disorders, and provide a theoretical basis for the future treatment strategies. Through this research, we aim to offer the deeper insights to the scientific community and present the new perspectives for the clinical practice in addressing the challenges of the neurological complications and sequelae faced by the COVID-19 patients.
Collapse
Affiliation(s)
- Wenzhi Chen
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Shishi Jiang
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Cheng Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Shu Li
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| | - Junling Wang
- Department of Neurology, Xiangya Hospital, Central South University, Changsha, China
| | - Renshi Xu
- Department of Neurology, Jiangxi Provincial People’s Hospital, The Clinical College of Nanchang Medical College, The First Affiliated Hospital of Nanchang Medical College, National Regional Center for Neurological Diseases, Xiangya Hospital of Central South University Jiangxi Hospital, Nanchang, China
| |
Collapse
|
35
|
Zhang X, Chen Z, Zheng J, Feng C, Zhao B, Lan L, Liu D. Dynamic Characteristics of Lymphocyte Subsets and Their Predictive Value for Disease Progression and Prognosis in Primary Infection and Unvaccinated COVID-19 Patients. Int J Gen Med 2024; 17:4559-4577. [PMID: 39403609 PMCID: PMC11472736 DOI: 10.2147/ijgm.s478912] [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: 07/20/2024] [Accepted: 10/06/2024] [Indexed: 03/17/2025] Open
Abstract
AIM Our cohort study aimed to investigate the dynamic changes of lymphocyte subsets and their abilities to predict disease severity and prognosis in primary infection and unvaccinated COVID-19 patients. METHODS A total of 773 cases, including 718 primary infection and unvaccinated COVID-19 patients and 55 controls. COVID-19 patients were assigned to severe and nonsevere groups according to disease severity, as well as survival and death groups according to prognosis. Serum samples were collected to measure the numbers of total lymphocytes and lymphocyte subsets. The differences among different severity groups were analyzed. Spearman correlation was performed to assess associations between lymphocyte subsets and disease severity and prognosis. Meanwhile, receiver operating characteristic (ROC) curves were also analyzed to find optimal cutoff points. RESULTS At admission, the severe group demonstrated significantly lower total lymphocyte counts and percentages, CD3+ and CD3+CD4+ T cell counts and percentages, CD3+CD8+ T cell counts, CD19+ B cell counts and CD56+ NK cell counts and percentages than the nonsevere group. Meanwhile, compared with the survival group, the death group also had lower total lymphocyte counts and percentages, CD3+, CD3+CD4+ and CD3+CD8+ T cell counts. Additionally, differences in these parameters were also noticed within four weeks after admission. Furthermore, Spearman analysis reported that disease severity was negatively correlated with lymphocyte counts and percentages, CD3+, CD3+CD4+ and CD3+CD8+ T cell counts, CD3+ and CD3+CD4+ T cell percentages (r=-0.166, -0.179, -0.173, -0.186, -0.127, -0.117, -0.149, respectively)(all P<0.05). The prognosis of death was also negatively correlated with total lymphocyte counts and percentages, CD3+, CD3+CD4+ and CD3+CD8+ T cell counts (r=-0.125, -0.121, -0.123, -0.123, -0.091, respectively)(all P<0.05). CONCLUSION In primary infection and unvaccinated COVID-19 patients total lymphocytes and T cell, B cell and NK cell subsets at COVID-19 onset play valuable roles in predicting disease severity and prognosis. CLINICAL TRIAL REGISTRY Chinese Clinical Trial Register ChiCTR2000034563.
Collapse
Affiliation(s)
- Xinyi Zhang
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
- Department of Endocrinology & Metabolism, Sichuan University West China Hospital, Chengdu, People’s Republic of China
| | - Zhu Chen
- Department of Drug Clinical Trial Center, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
| | - Jun Zheng
- Medical Department, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
| | - Chen Feng
- Legal Services Division, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
| | - Bennan Zhao
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
| | - Lijuan Lan
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
| | - Dafeng Liu
- The First Ward of Internal Medicine, Public Health Clinical Centre of Chengdu, Chengdu, People’s Republic of China
| |
Collapse
|
36
|
Yuan J, Zhang W, Qie B, Xie Y, Zhu B, Chen C, Qiu W, Sun H, Zhao B, Long Y. Utilizing press needle acupuncture to treat mild-to-moderate COVID-19: A single-blind, randomized controlled trial. Medicine (Baltimore) 2024; 103:e39810. [PMID: 39465704 PMCID: PMC11460845 DOI: 10.1097/md.0000000000039810] [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: 04/25/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND In China, acupuncture has been employed as an adjunctive therapy for coronavirus disease 2019 (COVID-19). Press needle acupuncture is a special type of acupuncture that provides prolonged stimulation to acupuncture points and simultaneously reduces the pain associated with traditional acupuncture. This study assessed the effectiveness of integrating press needles alongside pharmacologic treatment in patients with mild-to-moderate COVID-19. METHODS Patients hospitalized with mild-to-moderate COVID-19 symptoms between December 2022 and January 2023 were included in the study. The enrolled patients were randomly assigned to receive pharmacologic treatment alone (control group) or both pharmacologic treatment and press needle acupuncture (intervention group). Patients were evaluated for clinical outcomes, including symptom scores, deterioration rates, fever durations, and nucleic acid test results. The patients' complete blood count and C-reactive protein levels were also analyzed using venous blood samples both before and after treatment. RESULTS Both groups exhibited a reduction in clinical symptom scores, but symptoms regressed faster in the intervention group. Nucleic acid test negativity was achieved faster in the intervention group than in the control group. The intervention group also had a lower deterioration rate. Furthermore, the increase in the lymphocyte count and decrease in C-reactive protein levels following treatment were more pronounced in the intervention group than in the control group. CONCLUSION This study suggests that utilizing press needle acupuncture as an adjunct to pharmacologic treatment can be effective in patients with mild-to-moderate COVID-19 symptoms.
Collapse
Affiliation(s)
- Jiawei Yuan
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Weizhen Zhang
- NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Beibei Qie
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yuhua Xie
- Taihe Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Binbin Zhu
- Guangdong Work Injury Rehabilitation Hospital, Guangzhou, Guangdong, China
| | - Cheng Chen
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wenwei Qiu
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Huanwen Sun
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Bin Zhao
- Southern Medical University, Guangzhou, Guangdong, China
| | - Yaqiu Long
- Baiyun Branch, NanFang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
37
|
Ma Y, Ji J, Liu X, Zheng X, Xu L, Zhou Q, Li Z, Yang L. Integrative Analysis by Mendelian Randomization and Large-Scale Single-Cell Transcriptomics Reveals Causal Links between B Cell Subtypes and Diabetic Kidney Disease. KIDNEY DISEASES (BASEL, SWITZERLAND) 2024; 10:327-345. [PMID: 39430286 PMCID: PMC11488840 DOI: 10.1159/000539689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 06/03/2024] [Indexed: 10/22/2024]
Abstract
Introduction The increasing incidence of diabetic kidney disease (DKD) and the challenges in its management highlight the necessity for a deeper understanding of its pathogenesis. While recent studies have underscored the substantial impact of circulating immunity on the development of diabetic microvascular complications such as retinopathy and neuropathy, research on circulating immunity in DKD remains limited. Methods This study utilized Mendelian randomization analysis to explore the potential independent causal relationships between circulating immune cells and DKD pathogenesis. Additionally, a combination of single-cell disease relevance score (scDRS) and immune cell infiltration analysis was employed to map the circulating immunity landscape in DKD patients. Results Ten immune traits, including 5 of B cells, 2 of T cells, 2 of granulocytes, and one of monocytes, were defined to be associated with the pathogenesis of DKD. Notably, IgD - CD27 - B cell Absolute Count (IVW: OR, 1.102 [1.023-1.189], p = 0.011) and IgD - CD24 - B cell Absolute Count (IVW: OR, 1.106 [1.030-1.188], p = 0.005) were associated with promoting DKD pathogenesis, while CD24 + CD27 + B cell %B cell (IVW: OR, 0.943 [0.898-0.989], p = 0.016) demonstrated a protective effect against DKD onset. The presence of B cell-activating factor receptor (BAFF-R) on CD20 - CD38 - B cell (IVW: OR, 0.946 [0.904-0.989], p = 0.015) and BAFF-R on IgD - CD38 + B cell (IVW: OR, 0.902 [0.834-0.975], p = 0.009) also indicated a potential role in preventing DKD. scDRS analysis revealed that two main subsets of B cells, naïve B and memory B cells, had a higher proportion of DKD-related cells or a higher scDRS score of DKD phenotype, indicating their strong association with DKD. Furthermore, immune infiltrate deconvolution analysis showed a notable decrease in the circulating memory B cells and class-switched memory B cells in DKD patients compared to those of DM patients without DKD. Conclusion Our study revealed the causal relations between circulating immunity and DKD susceptibility, particularly highlighted the potential roles of B cell subtypes in DKD development. Further studies addressing the related mechanisms would broaden the current understanding of DKD pathogenesis.
Collapse
Affiliation(s)
- Yuan Ma
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Ji
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
- Department of Nephrology, The Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Xintong Liu
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xizi Zheng
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Lingyi Xu
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Qingqing Zhou
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zehua Li
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Li Yang
- Key Laboratory of Renal Disease-Ministry of Health of China, Key Laboratory of CKD Prevention and Treatment (Peking University)-Ministry of Education of China, Renal Division, Peking University First Hospital, Peking University Institute of Nephrology, Beijing, China
- Research Units of Diagnosis and Treatment of Immune-Mediated Kidney Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
38
|
Babadei O, Strobl B, Müller M, Decker T. Transcriptional control of interferon-stimulated genes. J Biol Chem 2024; 300:107771. [PMID: 39276937 PMCID: PMC11489399 DOI: 10.1016/j.jbc.2024.107771] [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: 06/01/2024] [Revised: 08/27/2024] [Accepted: 08/29/2024] [Indexed: 09/17/2024] Open
Abstract
Interferon-induced genes are among the best-studied groups of coregulated genes. Nevertheless, intense research into their regulation, supported by new technologies, is continuing to provide insights into their many layers of transcriptional regulation and to reveal how cellular transcriptomes change with pathogen-induced innate and adaptive immunity. This article gives an overview of recent findings on interferon-induced gene regulation, paying attention to contributions beyond the canonical JAK-STAT pathways.
Collapse
Affiliation(s)
- Olga Babadei
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Vienna, Austria
| | - Birgit Strobl
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mathias Müller
- Institute of Animal Breeding and Genetics, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Thomas Decker
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Vienna, Austria; University of Vienna, Center for Molecular Biology, Department of Microbiology, Immunobiology and Genetics, Vienna, Austria.
| |
Collapse
|
39
|
Hédou J, Marić I, Bellan G, Einhaus J, Gaudillière DK, Ladant FX, Verdonk F, Stelzer IA, Feyaerts D, Tsai AS, Ganio EA, Sabayev M, Gillard J, Amar J, Cambriel A, Oskotsky TT, Roldan A, Golob JL, Sirota M, Bonham TA, Sato M, Diop M, Durand X, Angst MS, Stevenson DK, Aghaeepour N, Montanari A, Gaudillière B. Discovery of sparse, reliable omic biomarkers with Stabl. Nat Biotechnol 2024; 42:1581-1593. [PMID: 38168992 PMCID: PMC11217152 DOI: 10.1038/s41587-023-02033-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 10/16/2023] [Indexed: 01/05/2024]
Abstract
Adoption of high-content omic technologies in clinical studies, coupled with computational methods, has yielded an abundance of candidate biomarkers. However, translating such findings into bona fide clinical biomarkers remains challenging. To facilitate this process, we introduce Stabl, a general machine learning method that identifies a sparse, reliable set of biomarkers by integrating noise injection and a data-driven signal-to-noise threshold into multivariable predictive modeling. Evaluation of Stabl on synthetic datasets and five independent clinical studies demonstrates improved biomarker sparsity and reliability compared to commonly used sparsity-promoting regularization methods while maintaining predictive performance; it distills datasets containing 1,400-35,000 features down to 4-34 candidate biomarkers. Stabl extends to multi-omic integration tasks, enabling biological interpretation of complex predictive models, as it hones in on a shortlist of proteomic, metabolomic and cytometric events predicting labor onset, microbial biomarkers of pre-term birth and a pre-operative immune signature of post-surgical infections. Stabl is available at https://github.com/gregbellan/Stabl .
Collapse
Affiliation(s)
- Julien Hédou
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Ivana Marić
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Grégoire Bellan
- Télécom Paris, Institut Polytechnique de Paris, Paris, France
| | - Jakob Einhaus
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology and Neuropathology, University Hospital and Comprehensive Cancer Center Tübingen, Tübingen, Germany
| | - Dyani K Gaudillière
- Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University, Stanford, CA, USA
| | | | - Franck Verdonk
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Sorbonne University, GRC 29, AP-HP, DMU DREAM, Department of Anesthesiology and Intensive Care, Hôpital Saint-Antoine, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Ina A Stelzer
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | - Dorien Feyaerts
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Amy S Tsai
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Edward A Ganio
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Maximilian Sabayev
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Joshua Gillard
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jonas Amar
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Amelie Cambriel
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Tomiko T Oskotsky
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Alennie Roldan
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Jonathan L Golob
- Department of Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Marina Sirota
- Bakar Computational Health Sciences Institute, University of California, San Francisco, San Francisco, CA, USA
| | - Thomas A Bonham
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Masaki Sato
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Maïgane Diop
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | - Xavier Durand
- École Polytechnique, Institut Polytechnique de Paris, Paris, France
| | - Martin S Angst
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
| | | | - Nima Aghaeepour
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University, Stanford, CA, USA
- Department of Biomedical Data Science, Stanford University, Stanford, CA, USA
| | - Andrea Montanari
- Department of Statistics, Stanford University, Stanford, CA, USA
- Department of Electrical Engineering, Stanford University, Stanford, CA, USA
| | - Brice Gaudillière
- Department of Anesthesiology, Perioperative & Pain Medicine, Stanford University, Stanford, CA, USA.
- Department of Pediatrics, Stanford University, Stanford, CA, USA.
| |
Collapse
|
40
|
Da Silva Filho J, Herder V, Gibbins MP, Dos Reis MF, Melo GC, Haley MJ, Judice CC, Val FFA, Borba M, Tavella TA, de Sousa Sampaio V, Attipa C, McMonagle F, Wright D, de Lacerda MVG, Costa FTM, Couper KN, Marcelo Monteiro W, de Lima Ferreira LC, Moxon CA, Palmarini M, Marti M. A spatially resolved single-cell lung atlas integrated with clinical and blood signatures distinguishes COVID-19 disease trajectories. Sci Transl Med 2024; 16:eadk9149. [PMID: 39259811 DOI: 10.1126/scitranslmed.adk9149] [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: 09/26/2023] [Revised: 02/15/2024] [Accepted: 08/05/2024] [Indexed: 09/13/2024]
Abstract
COVID-19 is characterized by a broad range of symptoms and disease trajectories. Understanding the correlation between clinical biomarkers and lung pathology during acute COVID-19 is necessary to understand its diverse pathogenesis and inform more effective treatments. Here, we present an integrated analysis of longitudinal clinical parameters, peripheral blood markers, and lung pathology in 142 Brazilian patients hospitalized with COVID-19. We identified core clinical and peripheral blood signatures differentiating disease progression between patients who recovered from severe disease compared with those who succumbed to the disease. Signatures were heterogeneous among fatal cases yet clustered into two patient groups: "early death" (<15 days until death) and "late death" (>15 days). Progression to early death was characterized systemically and in lung histopathological samples by rapid endothelial and myeloid activation and the presence of thrombi associated with SARS-CoV-2+ macrophages. In contrast, progression to late death was associated with fibrosis, apoptosis, and SARS-CoV-2+ epithelial cells in postmortem lung tissue. In late death cases, cytotoxicity, interferon, and T helper 17 (TH17) signatures were only detectable in the peripheral blood after 2 weeks of hospitalization. Progression to recovery was associated with higher lymphocyte counts, TH2 responses, and anti-inflammatory-mediated responses. By integrating antemortem longitudinal blood signatures and spatial single-cell lung signatures from postmortem lung samples, we defined clinical parameters that could be used to help predict COVID-19 outcomes.
Collapse
Affiliation(s)
- João Da Silva Filho
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Vanessa Herder
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Matthew P Gibbins
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
| | - Monique Freire Dos Reis
- Department of Education and Research, Oncology Control Centre of Amazonas State (FCECON), Manaus, Brazil
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Federal University of Amazonas, Manaus, Brazil
- Amazonas Oncology Control Center Foundation, Manaus, Brazil
| | | | - Michael J Haley
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Carla Cristina Judice
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
| | - Fernando Fonseca Almeida Val
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Mayla Borba
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Delphina Rinaldi Abdel Aziz Emergency Hospital (HPSDRA), Manaus, Brazil
| | - Tatyana Almeida Tavella
- Department of Genetics, Evolution, Microbiology and Immunology, University of Campinas, Campinas, Brazil
- INSERM U1016, CNRS UMR8104, University of Paris Cité, Institut Cochin, Paris, France
| | | | - Charalampos Attipa
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Department of Tropical Disease Biology, Liverpool School of Tropical Medicine, Liverpool, UK
- Royal (Dick) School of Veterinary Studies and Roslin Institute, University of Edinburgh, Edinburgh, UK
| | - Fiona McMonagle
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Glasgow Imaging Facility/School of Infection and Immunity, University of Glasgow, Glasgow, UK
| | - Derek Wright
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
| | - Marcus Vinicius Guimaraes de Lacerda
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
- Instituto Leônidas e Maria Deane, Fiocruz, Manaus, Brazil
- University of Texas Medical Branch, Galveston, TX, USA
| | | | - Kevin N Couper
- Department of Immunology, Immunity to Infection and Respiratory Medicine, University of Manchester, Manchester, UK
| | - Wuelton Marcelo Monteiro
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Luiz Carlos de Lima Ferreira
- Postgraduate Program in Tropical Medicine, University of Amazonas State, Manaus, Brazil
- Tropical Medicine Foundation Dr. Heitor Vieira Dourado, Manaus, Brazil
| | - Christopher Alan Moxon
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- (C.A.M.)
| | - Massimo Palmarini
- MRC-University of Glasgow Centre for Virus Research, Glasgow, UK
- (M.P.)
| | - Matthias Marti
- Wellcome Centre for Integrative Parasitology, School of Infection and Immunity, University of Glasgow, Glasgow, UK
- Institute of Parasitology Zurich (IPZ), VetSuisse Faculty, University of Zurich, Zurich, Switzerland
- (M.M.)
| |
Collapse
|
41
|
Samaran J, Peyré G, Cantini L. scConfluence: single-cell diagonal integration with regularized Inverse Optimal Transport on weakly connected features. Nat Commun 2024; 15:7762. [PMID: 39237488 PMCID: PMC11377776 DOI: 10.1038/s41467-024-51382-x] [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: 03/11/2024] [Accepted: 08/06/2024] [Indexed: 09/07/2024] Open
Abstract
The abundance of unpaired multimodal single-cell data has motivated a growing body of research into the development of diagonal integration methods. However, the state-of-the-art suffers from the loss of biological information due to feature conversion and struggles with modality-specific populations. To overcome these crucial limitations, we here introduce scConfluence, a method for single-cell diagonal integration. scConfluence combines uncoupled autoencoders on the complete set of features with regularized Inverse Optimal Transport on weakly connected features. We extensively benchmark scConfluence in several single-cell integration scenarios proving that it outperforms the state-of-the-art. We then demonstrate the biological relevance of scConfluence in three applications. We predict spatial patterns for Scgn, Synpr and Olah in scRNA-smFISH integration. We improve the classification of B cells and Monocytes in highly heterogeneous scRNA-scATAC-CyTOF integration. Finally, we reveal the joint contribution of Fezf2 and apical dendrite morphology in Intra Telencephalic neurons, based on morphological images and scRNA.
Collapse
Affiliation(s)
- Jules Samaran
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, Paris, France
| | - Gabriel Peyré
- CNRS and DMA de l'Ecole Normale Supérieure, CNRS, Ecole Normale Supérieure, Université PSL, Paris, France
| | - Laura Cantini
- Institut Pasteur, Université Paris Cité, CNRS UMR 3738, Machine Learning for Integrative Genomics Group, Paris, France.
| |
Collapse
|
42
|
Lakshmikanth T, Consiglio C, Sardh F, Forlin R, Wang J, Tan Z, Barcenilla H, Rodriguez L, Sugrue J, Noori P, Ivanchenko M, Piñero Páez L, Gonzalez L, Habimana Mugabo C, Johnsson A, Ryberg H, Hallgren Å, Pou C, Chen Y, Mikeš J, James A, Dahlqvist P, Wahlberg J, Hagelin A, Holmberg M, Degerblad M, Isaksson M, Duffy D, Kämpe O, Landegren N, Brodin P. Immune system adaptation during gender-affirming testosterone treatment. Nature 2024; 633:155-164. [PMID: 39232147 PMCID: PMC11374716 DOI: 10.1038/s41586-024-07789-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/04/2024] [Indexed: 09/06/2024]
Abstract
Infectious, inflammatory and autoimmune conditions present differently in males and females. SARS-CoV-2 infection in naive males is associated with increased risk of death, whereas females are at increased risk of long COVID1, similar to observations in other infections2. Females respond more strongly to vaccines, and adverse reactions are more frequent3, like most autoimmune diseases4. Immunological sex differences stem from genetic, hormonal and behavioural factors5 but their relative importance is only partially understood6-8. In individuals assigned female sex at birth and undergoing gender-affirming testosterone therapy (trans men), hormone concentrations change markedly but the immunological consequences are poorly understood. Here we performed longitudinal systems-level analyses in 23 trans men and found that testosterone modulates a cross-regulated axis between type-I interferon and tumour necrosis factor. This is mediated by functional attenuation of type-I interferon responses in both plasmacytoid dendritic cells and monocytes. Conversely, testosterone potentiates monocyte responses leading to increased tumour necrosis factor, interleukin-6 and interleukin-15 production and downstream activation of nuclear factor kappa B-regulated genes and potentiation of interferon-γ responses, primarily in natural killer cells. These findings in trans men are corroborated by sex-divergent responses in public datasets and illustrate the dynamic regulation of human immunity by sex hormones, with implications for the health of individuals undergoing hormone therapy and our understanding of sex-divergent immune responses in cisgender individuals.
Collapse
Affiliation(s)
| | - Camila Consiglio
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
- Department of Laboratory Medicine, Lund University, Lund, Sweden
| | - Fabian Sardh
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden
| | - Rikard Forlin
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jun Wang
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Ziyang Tan
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Hugo Barcenilla
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Lucie Rodriguez
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jamie Sugrue
- Translational Immunology Unit, Institut Pasteur, Paris, France
| | - Peri Noori
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Margarita Ivanchenko
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Laura Piñero Páez
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Laura Gonzalez
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | | | - Anette Johnsson
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Henrik Ryberg
- Department of Clinical Chemistry, Sahlgrenska University Hospital, Gothenburg, Sweden
- Department of Internal Medicine and Clinical Nutrition, University of Gothenburg, Gothenburg, Sweden
| | - Åsa Hallgren
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Christian Pou
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Yang Chen
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Jaromír Mikeš
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Anna James
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden
| | - Per Dahlqvist
- Department of Public Health and Clinical Medicine, Umeå University, Umeå, Sweden
| | | | - Anders Hagelin
- ANOVA, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Mats Holmberg
- ANOVA, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marie Degerblad
- ANOVA, Karolinska University Hospital, Stockholm, Sweden
- Department of Molecular Medicine and Surgery, Karolinska Institutet, Solna, Sweden
| | - Magnus Isaksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Darragh Duffy
- Translational Immunology Unit, Institut Pasteur, Paris, France
| | - Olle Kämpe
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden
- Department of Endocrinology, Metabolism and Diabetes, Karolinska University Hospital, Stockholm, Sweden
| | - Nils Landegren
- Center for Molecular Medicine, Department of Medicine, Karolinska Institutet, Solna, Sweden.
- Science for Life Laboratory, Department of Medical Biochemistry and Microbiology, Uppsala University, Uppsala, Sweden.
| | - Petter Brodin
- Department of Women's and Children's Health, Karolinska Institutet, Solna, Sweden.
- Medical Research Council, Laboratory of Medical Sciences, London, UK.
- Department of Immunology and Inflammation, Imperial College London, London, UK.
| |
Collapse
|
43
|
Ferrena A, Zheng XY, Jackson K, Hoang B, Morrow BE, Zheng D. scDAPP: a comprehensive single-cell transcriptomics analysis pipeline optimized for cross-group comparison. NAR Genom Bioinform 2024; 6:lqae134. [PMID: 39345754 PMCID: PMC11437360 DOI: 10.1093/nargab/lqae134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Revised: 07/07/2024] [Accepted: 09/18/2024] [Indexed: 10/01/2024] Open
Abstract
Single-cell transcriptomics profiling has increasingly been used to evaluate cross-group (or condition) differences in cell population and cell-type gene expression. This often leads to large datasets with complex experimental designs that need advanced comparative analysis. Concurrently, bioinformatics software and analytic approaches also become more diverse and constantly undergo improvement. Thus, there is an increased need for automated and standardized data processing and analysis pipelines, which should be efficient and flexible too. To address these, we develop the single-cell Differential Analysis and Processing Pipeline (scDAPP), a R-based workflow for comparative analysis of single cell (or nucleus) transcriptomic data between two or more groups and at the levels of single cells or 'pseudobulking' samples. The pipeline automates many steps of pre-processing using data-learnt parameters, uses previously benchmarked software, and generates comprehensive intermediate data and final results that are valuable for both beginners and experts of scRNA-seq analysis. Moreover, the analytic reports, augmented by extensive data visualization, increase the transparency of computational analysis and parameter choices, while facilitate users to go seamlessly from raw data to biological interpretation. scDAPP is freely available under the MIT license, with source code, documentation and sample data at the GitHub (https://github.com/bioinfoDZ/scDAPP).
Collapse
Affiliation(s)
- Alexander Ferrena
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Institute for Clinical and Translational Research, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Xiang Yu Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Kevyn Jackson
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bang Hoang
- Department of Orthopedic Surgery, Montefiore Medical Center, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Bernice E Morrow
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Departments of Obstetrics and Gynecology, and Pediatrics, Albert Einstein College of Medicine, Bronx, NY, USA
| | - Deyou Zheng
- Department of Genetics, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neurology, Albert Einstein College of Medicine, Bronx, NY, USA
- Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
44
|
Wang M, Zhang D, Lei T, Zhou Y, Qin H, Wu Y, Liu S, Zhang L, Jia K, Dong Y, Wang S, Li Y, Fan Y, Gui L, Dong Y, Zhang W, Li Z, Hou J. Interferon-responsive neutrophils and macrophages extricate SARS-CoV-2 Omicron critical patients from the nasty fate of sepsis. J Med Virol 2024; 96:e29889. [PMID: 39206862 DOI: 10.1002/jmv.29889] [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: 04/22/2024] [Revised: 07/24/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
The SARS-CoV-2 Omicron variant is characterized by its high transmissibility, which has caused a worldwide epidemiological event. Yet, it turns ominous once the disease progression degenerates into severe pneumonia and sepsis, presenting a horrendous lethality. To elucidate the alveolar immune or inflammatory landscapes of Omicron critical-ill patients, we performed single-cell RNA-sequencing (scRNA-seq) of bronchoalveolar lavage fluid (BALF) from the patients with critical pneumonia caused by Omicron infection, and analyzed the correlation between the clinical severity scores and different immune cell subpopulations. In the BALF of Omicron critical patients, the alveolar violent myeloid inflammatory environment was determined. ISG15+ neutrophils and CXCL10+ macrophages, both expressed the interferon-stimulated genes (ISGs), were negatively correlated with clinical pulmonary infection score, while septic CST7+ neutrophils and inflammatory VCAN+ macrophages were positively correlated with sequential organ failure assessment. The percentages of ISG15+ neutrophils were associated with more protective alveolar epithelial cells, and may reshape CD4+ T cells to the exhaustive phenotype, thus preventing immune injuries. The CXCL10+ macrophages may promote plasmablast/plasma cell survival and activation as well as the production of specific antibodies. As compared to the previous BALF scRNA-seq data from SARS-CoV-2 wild-type/Alpha critical patients, the subsets of neutrophils and macrophages with pro-inflammatory and immunoregulatory features presented obvious distinctions, suggesting an immune disparity in Omicron variants. Overall, this study provides a BALF single-cell atlas of Omicron critical patients, and suggests that alveolar interferon-responsive neutrophils and macrophages may extricate SARS-CoV-2 Omicron critical patients from the nasty fate of sepsis.
Collapse
Affiliation(s)
- Mu Wang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Dingji Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Ting Lei
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Ye Zhou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Hao Qin
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Yanfeng Wu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Shuxun Liu
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Liyuan Zhang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Kaiwei Jia
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yue Dong
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Suyuan Wang
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yunhui Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yiwen Fan
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Liangchen Gui
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Yuchao Dong
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Wei Zhang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Second Military Medical University, Shanghai, China
| | - Zhixuan Li
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| | - Jin Hou
- National Key Laboratory of Immunity and Inflammation, Institute of Immunology, Second Military Medical University, Shanghai, China
| |
Collapse
|
45
|
Vu Manh TP, Gouin C, De Wolf J, Jouneau L, Pascale F, Bevilacqua C, Ar Gouilh M, Da Costa B, Chevalier C, Glorion M, Hannouche L, Urien C, Estephan J, Magnan A, Le Guen M, Marquant Q, Descamps D, Dalod M, Schwartz-Cornil I, Sage E. SARS-CoV2 infection in whole lung primarily targets macrophages that display subset-specific responses. Cell Mol Life Sci 2024; 81:351. [PMID: 39147987 PMCID: PMC11335275 DOI: 10.1007/s00018-024-05322-z] [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: 11/09/2023] [Revised: 05/22/2024] [Accepted: 06/05/2024] [Indexed: 08/17/2024]
Abstract
Deciphering the initial steps of SARS-CoV-2 infection, that influence COVID-19 outcomes, is challenging because animal models do not always reproduce human biological processes and in vitro systems do not recapitulate the histoarchitecture and cellular composition of respiratory tissues. To address this, we developed an innovative ex vivo model of whole human lung infection with SARS-CoV-2, leveraging a lung transplantation technique. Through single-cell RNA-seq, we identified that alveolar and monocyte-derived macrophages (AMs and MoMacs) were initial targets of the virus. Exposure of isolated lung AMs, MoMacs, classical monocytes and non-classical monocytes (ncMos) to SARS-CoV-2 variants revealed that while all subsets responded, MoMacs produced higher levels of inflammatory cytokines than AMs, and ncMos contributed the least. A Wuhan lineage appeared to be more potent than a D614G virus, in a dose-dependent manner. Amidst the ambiguity in the literature regarding the initial SARS-CoV-2 cell target, our study reveals that AMs and MoMacs are dominant primary entry points for the virus, and suggests that their responses may conduct subsequent injury, depending on their abundance, the viral strain and dose. Interfering on virus interaction with lung macrophages should be considered in prophylactic strategies.
Collapse
Affiliation(s)
- Thien-Phong Vu Manh
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France.
| | - Carla Gouin
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Julien De Wolf
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Luc Jouneau
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Université Paris-Saclay, INRAE, UVSQ, BREED, 78350, Jouy-en-Josas, France
| | - Florentina Pascale
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Claudia Bevilacqua
- Université Paris-Saclay, INRAE, AgroParisTech, GABI, 78350, Jouy-en-Josas, France
| | - Meriadeg Ar Gouilh
- Department of Virology, Univ Caen Normandie, Dynamicure INSERM UMR 1311, CHU Caen, 14000, Caen, France
| | - Bruno Da Costa
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | | | - Matthieu Glorion
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| | - Laurent Hannouche
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Céline Urien
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Jérôme Estephan
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Antoine Magnan
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Pulmonology, Foch Hospital, 92150, Suresnes, France
| | - Morgan Le Guen
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Anesthesiology, Foch Hospital, 92150, Suresnes, France
| | - Quentin Marquant
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Pulmonology, Foch Hospital, 92150, Suresnes, France
- Delegation to Clinical Research and Innovation, Foch Hospital, 92150, Suresnes, France
| | - Delphyne Descamps
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
| | - Marc Dalod
- Aix-Marseille University, CNRS, INSERM, CIML, Centre d'Immunologie de Marseille-Luminy, Turing Center for Living Systems, 13009, Marseille, France
| | | | - Edouard Sage
- Université Paris-Saclay, INRAE, UVSQ, VIM, 78350, Jouy-en-Josas, France
- Department of Thoracic Surgery and Lung Transplantation, Foch Hospital, 92150, Suresnes, France
| |
Collapse
|
46
|
Tisch C, Xourgia E, Exadaktylos A, Ziaka M. Potential use of sodium glucose co-transporter 2 inhibitors during acute illness: a systematic review based on COVID-19. Endocrine 2024; 85:660-675. [PMID: 38448675 PMCID: PMC11291544 DOI: 10.1007/s12020-024-03758-8] [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: 12/15/2023] [Accepted: 02/19/2024] [Indexed: 03/08/2024]
Abstract
OBJECTIVE SGLT-2i are increasingly recognized for their benefits in patients with cardiometabolic risk factors. Additionally, emerging evidence suggests potential applications in acute illnesses, including COVID-19. This systematic review aims to evaluate the effects of SGLT-2i in patients facing acute illness, particularly focusing on SARS-CoV-2 infection. METHODS Following PRISMA guidelines, a systematic search of PubMed, Scopus, medRxiv, Research Square, and Google Scholar identified 22 studies meeting inclusion criteria, including randomized controlled trials and observational studies. Data extraction and quality assessment were conducted independently. RESULTS Out of the 22 studies included in the review, six reported reduced mortality in DM-2 patients taking SGLT-2i, while two found a decreased risk of hospitalization. Moreover, one study demonstrated a lower in-hospital mortality rate in DM-2 patients under combined therapy of metformin plus SGLT-2i. However, three studies showed a neutral effect on the risk of hospitalization. No increased risk of developing COVID-19 was associated with SGLT-2i use in DM-2 patients. Prior use of SGLT-2i was not associated with ICU admission and need for MV. The risk of acute kidney injury showed variability, with inconsistent evidence regarding diabetic ketoacidosis. CONCLUSION Our systematic review reveals mixed findings on the efficacy of SGLT-2i use in COVID-19 patients with cardiometabolic risk factors. While some studies suggest potential benefits in reducing mortality and hospitalizations, others report inconclusive results. Further research is needed to clarify optimal usage and mitigate associated risks, emphasizing caution in clinical interpretation.
Collapse
Affiliation(s)
- Carmen Tisch
- Department of Internal Medicine, Thun General Hospital, Thun, Switzerland
| | - Eleni Xourgia
- Department of Cardiology, Inselspital, University Hospital, University of Bern, 3008, Bern, Switzerland
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Aristomenis Exadaktylos
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland
| | - Mairi Ziaka
- Department of Emergency Medicine, Inselspital, University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
47
|
Monticone G, Huang Z, Hewins P, Cook T, Mirzalieva O, King B, Larter K, Miller-Ensminger T, Sanchez-Pino MD, Foster TP, Nichols OV, Ramsay AJ, Majumder S, Wyczechowska D, Tauzier D, Gravois E, Crabtree JS, Garai J, Li L, Zabaleta J, Barbier MT, Del Valle L, Jurado KA, Miele L. Novel immunomodulatory properties of adenosine analogs promote their antiviral activity against SARS-CoV-2. EMBO Rep 2024; 25:3547-3573. [PMID: 39009832 PMCID: PMC11315900 DOI: 10.1038/s44319-024-00189-4] [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: 07/26/2023] [Revised: 04/30/2024] [Accepted: 06/14/2024] [Indexed: 07/17/2024] Open
Abstract
The COVID-19 pandemic reminded us of the urgent need for new antivirals to control emerging infectious diseases and potential future pandemics. Immunotherapy has revolutionized oncology and could complement the use of antivirals, but its application to infectious diseases remains largely unexplored. Nucleoside analogs are a class of agents widely used as antiviral and anti-neoplastic drugs. Their antiviral activity is generally based on interference with viral nucleic acid replication or transcription. Based on our previous work and computer modeling, we hypothesize that antiviral adenosine analogs, like remdesivir, have previously unrecognized immunomodulatory properties which contribute to their therapeutic activity. In the case of remdesivir, we here show that these properties are due to its metabolite, GS-441524, acting as an Adenosine A2A Receptor antagonist. Our findings support a new rationale for the design of next-generation antiviral agents with dual - immunomodulatory and intrinsic - antiviral properties. These compounds could represent game-changing therapies to control emerging viral diseases and future pandemics.
Collapse
Affiliation(s)
- Giulia Monticone
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | - Zhi Huang
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Peter Hewins
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Thomasina Cook
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Oygul Mirzalieva
- Department of Biochemistry and Molecular Biology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Brionna King
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Kristina Larter
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Taylor Miller-Ensminger
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Maria D Sanchez-Pino
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Timothy P Foster
- Department of Microbiology, Immunology & Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Olga V Nichols
- Department of Microbiology, Immunology & Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Alistair J Ramsay
- Department of Microbiology, Immunology & Parasitology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Samarpan Majumder
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Dorota Wyczechowska
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Darlene Tauzier
- Precision Medicine Program, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Department of Pathology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Elizabeth Gravois
- Precision Medicine Program, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Department of Pathology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Judy S Crabtree
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Precision Medicine Program, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Jone Garai
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Li Li
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Jovanny Zabaleta
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Mallory T Barbier
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Luis Del Valle
- Department of Interdisciplinary Oncology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
- Department of Pathology, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| | - Kellie A Jurado
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Lucio Miele
- Department of Genetics, School of Medicine, Louisiana State University Health Sciences Center, New Orleans, LA, USA
| |
Collapse
|
48
|
Lisowska KA, Ciesielska-Figlon K, Komorniczak M, Bułło-Piontecka B, Dębska-Ślizień A, Wardowska A. Peripheral Blood Mononuclear Cells and Serum Cytokines in Patients with Lupus Nephritis after COVID-19. Int J Mol Sci 2024; 25:8278. [PMID: 39125849 PMCID: PMC11311954 DOI: 10.3390/ijms25158278] [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: 07/01/2024] [Revised: 07/18/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Systemic lupus erythematosus (SLE) patients have an increased risk of infections and infection-related mortality. Therefore, during the global SARS-CoV-2 pandemic, SLE patients were particularly vulnerable to SARS-CoV-2 infections. Also, compared to other patients, SLE patients seem to develop more severe manifestations of coronavirus disease 2019 (COVID-19), with higher rates of hospitalization, invasive ventilation requirements, or death. This study evaluated the immune parameters after SARS-CoV-2 infection in SLE patients. We analyzed subpopulations of peripheral blood cells collected from patients with renal manifestation of SLE (lupus nephritis, LN). LN patients were divided into two subgroups: those unexposed to SARS-CoV-2 (LN CoV-2(-)) and those who had confirmed COVID-19 (LN-CoV-2(+)) six months earlier. We analyzed basic subpopulations of T cells, B cells, monocytes, dendritic cells (DCs), and serum cytokines using flow cytometry. All collected data were compared to a healthy control group without SARS-CoV-2 infection in medical history. LN patients were characterized by a decreased percentage of helper T (Th) cells and an increased percentage of cytotoxic T (Tc) cells regardless of SARS-CoV-2 infection. LN CoV-2(+) patients had a higher percentage of regulatory T cells (Tregs) and plasmablasts (PBs) and a lower percentage of non-switched memory (NSM) B cells compared to LN CoV-2(-) patients or healthy controls (HC CoV-2(-)). LN patients had a higher percentage of total monocytes compared with HC CoV-2(-). LN CoV-2(+) patients had a higher percentage of classical and intermediate monocytes than LN CoV-2(-) patients and HC CoV-2(-). LN CoV-2(+) patients had higher serum IL-6 levels than HC CoV-2(-), while LN CoV-2(-) patients had higher levels of serum IL-10. LN patients are characterized by disturbances in the blood's basic immunological parameters. However, SARS-CoV-2 infection influences B-cell and monocyte compartments.
Collapse
Affiliation(s)
- Katarzyna A. Lisowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.A.L.)
| | - Klaudia Ciesielska-Figlon
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.A.L.)
| | - Michał Komorniczak
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Barbara Bułło-Piontecka
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Alicja Dębska-Ślizień
- Department of Nephrology, Transplantology and Internal Medicine, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland
| | - Anna Wardowska
- Department of Pathophysiology, Faculty of Medicine, Medical University of Gdańsk, 80-211 Gdańsk, Poland; (K.A.L.)
| |
Collapse
|
49
|
Rabøl Andersen L, Hindsberger B, Bastrup Israelsen S, Pedersen L, Bela Szecsi P, Benfield T. Higher levels of IL-1ra, IL-6, IL-8, MCP-1, MIP-3α, MIP-3β, and fractalkine are associated with 90-day mortality in 132 non-immunomodulated hospitalized patients with COVID-19. PLoS One 2024; 19:e0306854. [PMID: 38985797 PMCID: PMC11236197 DOI: 10.1371/journal.pone.0306854] [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: 03/15/2024] [Accepted: 06/25/2024] [Indexed: 07/12/2024] Open
Abstract
INTRODUCTION Immune dysregulation with an excessive release of cytokines has been identified as a key driver in the development of severe COVID-19. The aim of this study was to evaluate the initial cytokine profile associated with 90-day mortality and respiratory failure in a cohort of patients hospitalized with COVID 19 that did not receive immunomodulatory therapy. METHODS Levels of 45 cytokines were measured in blood samples obtained at admission from patients with confirmed COVID-19. Logistic regression analysis was utilized to determine the association between cytokine levels and outcomes. The primary outcome was death within 90 days from admission and the secondary outcome was need for mechanical ventilation. RESULTS A total of 132 patients were included during the spring of 2020. We found that one anti-inflammatory cytokine, one pro-inflammatory cytokine, and five chemokines were associated with the odds of 90-day mortality, specifically: interleukin-1 receptor antagonist, interleukin-6, interleukin-8, monocyte chemoattractant protein-1, macrophage inflammatory protein-3α, macrophage inflammatory protein-3β, and fractalkine. All but fractalkine were also associated with the odds of respiratory failure during admission. Monocyte chemoattractant protein-1 showed the strongest estimate of association with both outcomes. CONCLUSION We showed that one anti-inflammatory cytokine, one pro-inflammatory cytokine, and five chemokines were associated with 90-day mortality in patients hospitalized with COVID-19 that did not receive immunomodulatory therapy.
Collapse
Affiliation(s)
- Liv Rabøl Andersen
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
| | - Bettina Hindsberger
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
| | - Simone Bastrup Israelsen
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
| | - Lise Pedersen
- Department of Clinical Biochemistry, Holbaek Hospital, Holbaek, Denmark
| | - Pal Bela Szecsi
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Biochemistry, Holbaek Hospital, Holbaek, Denmark
| | - Thomas Benfield
- Center of Clinical Research and Disruption of Infectious Diseases (CREDID), Department of Infectious Diseases, Copenhagen University Hospital—Amager and Hvidovre, Hvidovre, Denmark
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
50
|
Curion F, Rich-Griffin C, Agarwal D, Ouologuem S, Rue-Albrecht K, May L, Garcia GEL, Heumos L, Thomas T, Lason W, Sims D, Theis FJ, Dendrou CA. Panpipes: a pipeline for multiomic single-cell and spatial transcriptomic data analysis. Genome Biol 2024; 25:181. [PMID: 38978088 PMCID: PMC11229213 DOI: 10.1186/s13059-024-03322-7] [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: 03/14/2023] [Accepted: 06/25/2024] [Indexed: 07/10/2024] Open
Abstract
Single-cell multiomic analysis of the epigenome, transcriptome, and proteome allows for comprehensive characterization of the molecular circuitry that underpins cell identity and state. However, the holistic interpretation of such datasets presents a challenge given a paucity of approaches for systematic, joint evaluation of different modalities. Here, we present Panpipes, a set of computational workflows designed to automate multimodal single-cell and spatial transcriptomic analyses by incorporating widely-used Python-based tools to perform quality control, preprocessing, integration, clustering, and reference mapping at scale. Panpipes allows reliable and customizable analysis and evaluation of individual and integrated modalities, thereby empowering decision-making before downstream investigations.
Collapse
Affiliation(s)
- Fabiola Curion
- Department of Computational Health, Institute of Computational Biology, Helmholtz Munich, Germany
- Department of Mathematics, School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | - Charlotte Rich-Griffin
- Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Devika Agarwal
- Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
| | - Sarah Ouologuem
- Department of Computational Health, Institute of Computational Biology, Helmholtz Munich, Germany
| | - Kevin Rue-Albrecht
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Lilly May
- Department of Computational Health, Institute of Computational Biology, Helmholtz Munich, Germany
| | - Giulia E L Garcia
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Doctoral Training Centre, University of Oxford, Oxford, UK
| | - Lukas Heumos
- Department of Computational Health, Institute of Computational Biology, Helmholtz Munich, Germany
- Comprehensive Pneumology Center With the CPC-M bioArchive, Helmholtz Zentrum Munich, Member of the German Center for Lung Research (DZL), Munich, Germany
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany
| | - Tom Thomas
- Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Oxford, UK
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK
- Nuffield Department of Medicine, Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Wojciech Lason
- Nuffield Department of Medicine, Respiratory Medicine Unit, Experimental Medicine Division, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - David Sims
- MRC WIMM Centre for Computational Biology, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, UK
| | - Fabian J Theis
- Department of Computational Health, Institute of Computational Biology, Helmholtz Munich, Germany.
- Department of Mathematics, School of Computation, Information and Technology, Technical University of Munich, Munich, Germany.
- TUM School of Life Sciences Weihenstephan, Technical University of Munich, Munich, Germany.
| | - Calliope A Dendrou
- Nuffield Department of Medicine, Centre for Human Genetics, University of Oxford, Oxford, UK.
- Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, Kennedy Institute of Rheumatology, University of Oxford, Oxford, UK.
- NIHR Oxford Biomedical Research Centre, Oxford, UK.
| |
Collapse
|