1
|
Patel PA, Ripp AT, Nguyen SA, Duffy AN, Soler ZM, Eskandari R, White DR, Schlosser RJ. Increased incidence of intracranial complications following pediatric sinogenic and otogenic infections in the post-COVID-19 Era: A systematic review and meta-analysis. Int J Pediatr Otorhinolaryngol 2025; 193:112364. [PMID: 40279858 DOI: 10.1016/j.ijporl.2025.112364] [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: 02/17/2025] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 04/29/2025]
Abstract
BACKGROUND This systematic-review and meta-analysis aims to evaluate and summarize the prevalence of pediatric intracranial complications following sinogenic or otogenic infections before and after the COVID-19 pandemic. METHODS A literature search was performed using the PubMed, Scopus, and CINAHL databases to answer the question: In pediatric patients, was there an increase in the prevalence or severity of intracranial complications due to sinogenic or otogenic infections during and after the COVID-19 pandemic? Publications which included primary data on patients under the age of 18 years old, focusing on intracranial complications following otogenic and sinogenic infections were included. RESULTS Of 1025 abstracts screened, 18 studies were included. There were no significant differences in age or sex between the two cohorts. Compared to the pre-COVID era, post-COVID infections were more likely to have neurologic complications upon presentation [11.4 % (1.6-53.0) vs 50.1 % (13.9-86.2), p < 0.01], cerebral venous sinus thrombosis (CVST) [14.1 % (10.6-18.2) vs 40.5 % (25.2-56.9), p < 0.01], intraparenchymal abscess [40.3 % (43.9-72.2) vs 54.9 % (25.2-87.1), p < 0.01], and meningitis [10.6 % (0.0-39.4) vs 40.2 % (13.4-70.8), p < 0.01]. Metronidazole use [38.7 % (31.8-46.0) vs 71.9 % (51.3-88.6), p < 0.01], craniectomy [16.1 % (1.3-42.8) vs 37.4 % (2.9-83.0), p = 0.02], and burr holes [16.8 % (11.5-23.3) vs 26.6 % (12.7-43.3), p = 0.02] were increased in the post-COVID cohort. CONCLUSION There are considerable differences in neurologic deficits, CVST, intraparenchymal abscesses, meningitis, and treatment modalities in pre- and post-COVID cohorts of children with intracranial complications of otorhinogenic origin. Further research is required to determine the underlying mechanism for these differences.
Collapse
Affiliation(s)
- Pranav A Patel
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA
| | - Asher T Ripp
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA; SUNY Downstate School of Medicine, Brooklyn, NY, USA
| | - Shaun A Nguyen
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA.
| | - Alexander N Duffy
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA
| | - Zachary M Soler
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA
| | - Ramin Eskandari
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA
| | - David R White
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA
| | - Rodney J Schlosser
- Department of Otolaryngology-Head and Neck Surgery, Medical University of South Carolina, 135 Rutledge Avenue, Charleston, SC, 29425-5500, USA
| |
Collapse
|
2
|
Hu Z, Przytycki PF, Pollard KS. CellWalker2: Multi-omic discovery using hierarchical cell type relationships. CELL GENOMICS 2025:100886. [PMID: 40409272 DOI: 10.1016/j.xgen.2025.100886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 11/04/2024] [Accepted: 04/28/2025] [Indexed: 05/25/2025]
Abstract
Tissues are composed of cells with a wide range of similarities to each other, yet existing methods for single-cell genomics treat cell types as discrete labels. To address this gap, we developed CellWalker2, a graph diffusion-based model for the annotation and mapping of multi-modal data. With our open-source software package, hierarchically related cell types can be probabilistically matched across contexts and used to annotate cells, genomic regions, or gene sets. Additional features include estimating statistical significance and enabling gene expression and chromatin accessibility to be jointly modeled. Through simulation studies, we show that CellWalker2 performs better than existing methods in cell-type annotation and mapping. We then use multi-omics data from the brain and immune system to demonstrate CellWalker2's ability to assign high-resolution cell-type labels to regulatory elements and TFs and to quantify both conserved and divergent cell-type relationships between species.
Collapse
Affiliation(s)
- Zhirui Hu
- Gladstone Institute of Data Science & Biotechnology, 1650 Owens Street, San Francisco, CA 94158, USA
| | - Pawel F Przytycki
- Gladstone Institute of Data Science & Biotechnology, 1650 Owens Street, San Francisco, CA 94158, USA; Faculty of Computing & Data Sciences, Boston University, 665 Commonwealth Avenue, Boston, MA 02215, USA
| | - Katherine S Pollard
- Gladstone Institute of Data Science & Biotechnology, 1650 Owens Street, San Francisco, CA 94158, USA; Department of Epidemiology & Biostatistics, University of California, San Francisco, 1650 Owens Street, San Francisco, CA 94158, USA; Chan Zuckerberg Biohub SF, 499 Illinois Street, San Francisco, CA 94158, USA.
| |
Collapse
|
3
|
Nehar-Belaid D, Mejías A, Xu Z, Marches R, Yerrabelli R, Chen G, Mertz S, Ye F, Sánchez PJ, Tsang JS, Aydillo T, Miorin L, Cupic A, García-Sastre A, Ucar D, Banchereau JF, Pascual V, Ramilo O. SARS-CoV-2 induced immune perturbations in infants vary with disease severity and differ from adults' responses. Nat Commun 2025; 16:4562. [PMID: 40379618 PMCID: PMC12084365 DOI: 10.1038/s41467-025-59411-z] [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: 10/14/2024] [Accepted: 04/22/2025] [Indexed: 05/19/2025] Open
Abstract
Differences in immune profiles of children and adults with COVID-19 have been previously described. However, no systematic studies have been reported from infants hospitalized with severe disease. We applied a multidimensional approach to decipher the immune responses of SARS-CoV-2 infected infants (n = 26; 10 subacute, 11 moderate and 5 severe disease; median age = 1.6 months) and matched controls (n = 14; median age = 2 months). Single cell (scRNA-seq) profiling of PBMCs revealed substantial alterations in cell composition in SARS-CoV-2 infected infants; with most cell-types switching to an interferon-stimulated gene (ISGhi) state including: (i) CD14+ monocytes co-expressing ISGs and inflammasome-related molecules, (ii) ISGhi naive CD4+ T cells, (iii) ISGhi proliferating cytotoxic CD8+ T cells, and (iv) ISGhi naive and transitional B cells. We observe increased serum concentrations of both interferons and inflammatory cytokines in infected infants. Antibody responses to SARS-CoV-2 are also consistently detected in the absence of anti-IFN autoantibodies. Compared with infected adults, infants display a similar ISG signature in monocytes but a markedly enhanced ISG signature in T and B cells. These findings provide insights into the distinct immune responses to SARS-CoV-2 in the first year of life and underscore the importance of further defining the unique features of early life immunity.
Collapse
Affiliation(s)
| | - Asunción Mejías
- Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zhaohui Xu
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Radu Marches
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Rushil Yerrabelli
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Guo Chen
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Sara Mertz
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Fang Ye
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA
| | - Pablo J Sánchez
- Department of Pediatrics, Division of Neonatology and Center for Perinatal Research, Ohio Perinatal Research Network, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, USA
| | - John S Tsang
- Center for Systems and Engineering Immunology, Departments of Immunobiology and Biomedical Engineering, Yale University, New Haven, CT, 06520, USA
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA
- Chan Zuckerberg Biohub NY, New Haven, CT, USA
| | - Teresa Aydillo
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Lisa Miorin
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Anastasija Cupic
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Adolfo García-Sastre
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Global Health and Emerging Pathogens Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Graduate School of Biomedical Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
- Department of Medicine, Division of Infectious Diseases, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
- Department of Pathology, Molecular and Cell-Based Medicine, One Gustave L. Levy Place, New York, NY, 10029, USA
- Icahn Genomics Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY, 10029, USA
| | - Duygu Ucar
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA
| | - Jacques F Banchereau
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, 06032, USA.
- Immunoledge LLC, Montclair, NJ, USA.
| | - Virginia Pascual
- Drukier Institute for Children's Health and Department of Pediatrics, Weill Cornell Medicine, New York, NY, USA.
| | - Octavio Ramilo
- Department of Pediatrics, Nationwide Children's Hospital and The Ohio State University College of Medicine, Columbus, OH, USA.
- Center for Vaccines and Immunity, Abigail Wexner Research Institute at Nationwide Children's Hospital, Columbus, OH, 43205, USA.
- Department of Infectious Diseases, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
4
|
Li S, Lv H, Zhang R, Li J, Chen Z, Yang N, Dai S. Aging-related alternative splicing drive neoantigen emergence revealed by transcriptome analysis of 1,255 human blood samples. FRONTIERS IN AGING 2025; 6:1575862. [PMID: 40417629 PMCID: PMC12098113 DOI: 10.3389/fragi.2025.1575862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 04/30/2025] [Indexed: 05/27/2025]
Abstract
This study aimed to identify age-related genes and alternative splicing (AS) events by comprehensive transcriptome analysis of 1,255 healthy blood samples from individuals aged 8-87 years. We identified 1,029 up-regulated and 1,186 down-regulated genes in older individuals, including 17 genes overlapped with known aging-associated genes, such as TFAP2A and Klotho. Gene set enrichment analysis revealed significant alterations in immunoregulatory and metabolic pathways during aging. However, many senescence-associated secretory phenotypes (SASP) involved genes did not exhibit changes in gene expression, suggesting that AS events may reveal additional age-related mechanisms. Aging also altered 6,320 AS events in 4,566 genes, impacting immune-related protein domains. The RNA-binding protein RBMS3 emerged as a key regulator of aging-specific AS events. In addition, neoantigen prediction analyses further identified potential neoantigens generated by aging-related AS events, with the HLA-C14:02 allele presenting the most neoantigenic peptides. Notably, 60 neoantigenic peptides were confirmed using proteomic data from elderly individuals, suggesting their potential as novel targets for anti-aging immunotherapy. Our study provides new insights into the role of alternative splicing in aging, highlights promising avenues for anti-aging immunotherapy.
Collapse
Affiliation(s)
- Shuhan Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Haohao Lv
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Renxin Zhang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Jinjun Li
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Zhiyuan Chen
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Naixue Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| | - Shaoxing Dai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, China
- Yunnan Key Laboratory of Primate Biomedical Research, Kunming, Yunnan, China
| |
Collapse
|
5
|
Mangiola S, Brown R, Zhan C, Berthelet J, Guleria S, Liyanage C, Ostrouska S, Wilcox J, Merdas M, Fuge-Larsen P, Bell C, Schröder J, Mielke LA, Mariadason JM, Tsao SCH, Chen Y, Yadav VK, Vodala S, Anderson RL, Merino D, Behren A, Yeo B, Papenfuss AT, Pal B. Circulating immune cells exhibit distinct traits linked to metastatic burden in breast cancer. Breast Cancer Res 2025; 27:73. [PMID: 40340807 PMCID: PMC12063295 DOI: 10.1186/s13058-025-01982-2] [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: 06/23/2024] [Accepted: 02/14/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND Circulating immune cells play a crucial role in the anti-tumour immune response, yet the systemic immune system in metastatic breast cancers is not fully characterised. Investigating the cellular and molecular changes in peripheral blood mononuclear cells (PBMCs) from breast cancer patients could elucidate the role of circulating immune cells in metastasis and aid in identifying biomarkers for disease burden and progression. METHODS In this study, we characterised the systemic immune landscape associated with varying levels of metastatic burden by analysing the single-cell transcriptomes of PBMCs from breast cancer patients and healthy controls. Our research focused on identifying changes in immune cell composition, transcriptional programs, and immune-cell communication networks linked to metastatic burden. Additionally, we compared these PBMC features onto a single-cell atlas of primary breast tumours to study corresponding traits in tumour-infiltrating immune cells. RESULTS In metastatic breast cancer, PBMCs exhibit a significant downregulation of the adaptive immune system and a decreased number and activity of unconventional T cells, such as γδ T cells. Additionally, metastatic burden is associated with impaired cell communication pathways involved in immunomodulatory functions. We also identified a gene signature derived from myeloid cells shared between tumour immune infiltrates and circulating immune cells in breast cancer patients. CONCLUSIONS Our study provides a comprehensive single-cell molecular profile of the peripheral immune system in breast cancer, offering a valuable resource for understanding metastatic disease in terms of tumour burden. By identifying immune traits linked to metastasis, we have unveiled potential new biomarkers of metastatic disease.
Collapse
Affiliation(s)
- S Mangiola
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
- South Australian immunoGENomics Cancer Institute, Adelaide, SA, 5005, Australia.
| | - R Brown
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Zhan
- South Australian immunoGENomics Cancer Institute, Adelaide, SA, 5005, Australia
| | - J Berthelet
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Guleria
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Liyanage
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Ostrouska
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - J Wilcox
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - M Merdas
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - P Fuge-Larsen
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - C Bell
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - J Schröder
- Peter Doherty Institute for Infection and Immunity, Parkville, VIC, 3052, Australia
- The University of Melbourne, Parkville, VIC, 3052, Australia
| | - L A Mielke
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC, 3086, Australia
| | - J M Mariadason
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - S Chang-Hao Tsao
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - Y Chen
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia
| | - V K Yadav
- Rutgers New Jersey Medical School, Newark, NJ, USA
| | - S Vodala
- Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, USA
| | - R L Anderson
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - D Merino
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - A Behren
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
| | - B Yeo
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia
- Austin Health, Heidelberg, VIC, 3084, Australia
| | - A T Papenfuss
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, 3052, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, VIC, 3052, Australia.
| | - B Pal
- School of Cancer Medicine, La Trobe University, Bundoora, VIC, 3086, Australia.
- Olivia Newton-John Cancer Research Institute, Heidelberg, VIC, 3084, Australia.
| |
Collapse
|
6
|
Joud A, Bteich F, Stella I, Klein O. Empyemas secondary to ENT infections in children before, during and after the COVID-19 pandemic. Neurochirurgie 2025; 71:101644. [PMID: 39890059 DOI: 10.1016/j.neuchi.2025.101644] [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: 10/07/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVES This study aims to analyze a series of cases admitted with intracranial empyema, either subdural or epidural, secondary to otorhinolaryngological (ENT) infections over a period of 10 years, including the COVID-19 pandemic. The incidence, characteristics, severity, and management of these conditions, as well as the influence of COVID-19, are described below. METHODS The authors conducted a retrospective review of all of the children admitted to the Pediatric Neurosurgery Department of Nancy with intracranial empyemas secondary to a confirmed sinus (sinogenic) or mastoid (otogenic) infection between 2014 and 2024. They recorded their age, clinical presentation, initial Glasgow Coma Scale (GCS) score, bacteriological results, as well as the number and type of neurosurgical procedures they were subjected to, and their clinical outcome. These results were compared across two periods: before, and after the start of the COVID-19 pandemic. RESULTS Nineteen children in total were surgically treated, with twelve having a subdural location for their empyema, and seven located exclusively in the epidural compartment. The clinical presentation and evolution were positive in all except for one epidural empyema (6/7), and in the majority of the subdural locations (10/12). Twelve patients (63,16%) were treated after the start of the COVID pandemic, including 11 between 2022 and 2023. No differences were observed in patient characteristics, bacterial population and prognosis between these two periods. No patient was proven positive for COVID-19 at the time of their treatment. CONCLUSIONS Empyemas secondary to ENT infections are potentially serious pathologies, whose prognosis has clearly improved over the years. The incidence has significantly increased during the COVID-19 pandemic, without changing the characteristics or prognosis of the pathology. This increase mainly took place in the last 2 years of the pandemic in our department. The incidence remains even higher than before 2020.
Collapse
Affiliation(s)
- Anthony Joud
- Pediatric Neurosurgery Department, CHRU Nancy, France.
| | - Fred Bteich
- Pediatric Neurosurgery Department, CHRU Nancy, France.
| | - Irène Stella
- Pediatric Neurosurgery Department, CHRU Nancy, France.
| | - Olivier Klein
- Pediatric Neurosurgery Department, CHRU Nancy, France.
| |
Collapse
|
7
|
Purdy R, John M, Bray A, Clare AJ, Copland DA, Chan YK, Henderson RH, Nerinckx F, Leroy BP, Yang P, Pennesi ME, MacLaren RE, Fischer MD, Dick AD, Xue K. Gene Therapy-Associated Uveitis (GTAU): Understanding and mitigating the adverse immune response in retinal gene therapy. Prog Retin Eye Res 2025; 106:101354. [PMID: 40090458 DOI: 10.1016/j.preteyeres.2025.101354] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 03/18/2025]
Abstract
Retinal gene therapy using adeno-associated viral (AAV) vectors has been a groundbreaking step-change in the treatment of inherited retinal diseases (IRDs) and could also be used to treat more common retinal diseases such as age-related macular degeneration and diabetic retinopathy. The delivery and expression of therapeutic transgenes in the eye is limited by innate and adaptive immune responses against components of the vector product, which has been termed gene therapy-associated uveitis (GTAU). This is clinically important as intraocular inflammation could lead to irreversible loss of retinal cells, deterioration of visual function and reduced durability of treatment effect associated with a costly one-off treatment. For retinal gene therapy to achieve an improved efficacy and safety profile for treating additional IRDs and more common diseases, the risk of GTAU must be minimised. We have collated insights from pre-clinical research, clinical trials, and the real-world implementation of AAV-mediated retinal gene therapy to help understand the risk factors for GTAU. We draw attention to an emerging framework, which includes patient demographics, vector construct, vector dose, route of administration, and choice of immunosuppression regime. Importantly, we consider efforts to date and potential future strategies to mitigate the adverse immune response across each of these domains. We advocate for more targeted immunomodulatory approaches to the prevention and treatment of GTAU based on better understanding of the underlying immune response.
Collapse
Affiliation(s)
- Ryan Purdy
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Molly John
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Alison J Clare
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; University College London (UCL) Institute of Ophthalmology, London, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - David A Copland
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; University College London (UCL) Institute of Ophthalmology, London, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - Ying Kai Chan
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, USA; Cirrus Therapeutics, Cambridge, MA, USA
| | - Robert H Henderson
- University College London (UCL) Great Ormond Street Institute of Child Health, London, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK
| | - Fanny Nerinckx
- Chirec Delta Hospital, Brussels, Belgium; Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium
| | - Bart P Leroy
- Department of Ophthalmology, Ghent University Hospital, Ghent, Belgium; Center for Medical Genetics Ghent (CMGG), Ghent University Hospital, Ghent, Belgium; Department of Head & Skin, Ghent University, Ghent, Belgium
| | - Paul Yang
- Casey Eye Institute, Oregon Health & Science University, Portland, USA
| | - Mark E Pennesi
- Casey Eye Institute, Oregon Health & Science University, Portland, USA; Retina Foundation of the Southwest, Dallas, TX, USA
| | - Robert E MacLaren
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - M Dominik Fischer
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Andrew D Dick
- Academic Unit of Ophthalmology, Translational Health Sciences, University of Bristol, Bristol, UK; University College London (UCL) Institute of Ophthalmology, London, UK; NIHR Biomedical Research Centre of Ophthalmology, Moorfields Eye Hospital, London, UK
| | - Kanmin Xue
- Nuffield Laboratory of Ophthalmology, Nuffield Department of Clinical Neurosciences, University of Oxford, Oxford, UK; Oxford University Hospitals NHS Foundation Trust, Oxford, UK; Great Ormond Street Hospital for Children NHS Foundation Trust, London, UK.
| |
Collapse
|
8
|
Shang Z, Huang L, Qin S. The underlying mechanism behind the different outcomes of COVID-19 in children and adults. Front Immunol 2025; 16:1440169. [PMID: 40370452 PMCID: PMC12075420 DOI: 10.3389/fimmu.2025.1440169] [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: 05/29/2024] [Accepted: 04/10/2025] [Indexed: 05/16/2025] Open
Abstract
Coronavirus disease 2019 (COVID-19), caused by SARS-CoV-2, has affected hundreds of millions of people globally, resulting in millions of deaths. During this pandemic, children have demonstrated greater resistance than adults, exhibiting lower infection rates, reduced mortality, and milder symptoms. Summarizing the differences in resistance between children and adults during COVID-19 can provide insights into protective mechanisms and potential implications for future treatments. In this review, we focused on summarizing and discussing the mechanisms for better protection of children in COVID-19. These protective mechanisms encompass several factors: the baseline expression of cell surface receptor ACE2 and hydrolase TMPRSS2, the impact of complications on COVID-19, and age-related cytokine profiles. Additionally, differences in local and systemic immune responses between children and adults also contribute significantly, particularly interferon responses, heterologous protection from non-COVID-19 vaccinations, and immune status variations influenced by micronutrient levels. The advantageous protection mechanisms of these children may provide insights into the prevention and treatment of COVID-19. Importantly, while age-related metabolic profiles and differential COVID-19 vaccine responses may contribute to protection in children, current comparative research remains limited and requires further investigation.
Collapse
Affiliation(s)
- Zifang Shang
- Research Experiment Center, Meizhou People’s Hospital, Meizhou Academy of Medical Sciences, Meizhou, Guangdong, China
- Guangdong Engineering Technological Research Center of Clinical Molecular Diagnosis and Antibody Drugs, Meizhou People's Hospital, Meizhou, Guangdong, China
| | - Ling Huang
- Department of Critical Medicine, Shenzhen Clinical Research Centre for Geriatrics, Shenzhen People’s Hospital, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong, China
| | - Shijie Qin
- Innovative Vaccine and Immunotherapy Research Center, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
- Paediatric Research Institute, Shenzhen Children’s Hospital, Shenzhen, China
| |
Collapse
|
9
|
Akhtar E, Kuddusi RU, Talukder MT, Jakarea M, Haq MA, Hossain MS, Vandenent M, Islam MZ, Zaman RU, Razzaque A, Sarker P, Raqib R. Functional T cell response to COVID-19 vaccination with or without natural infection with SARS-CoV-2 in adults and children. Sci Rep 2025; 15:13341. [PMID: 40247005 PMCID: PMC12006499 DOI: 10.1038/s41598-025-95870-6] [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: 09/01/2024] [Accepted: 03/24/2025] [Indexed: 04/19/2025] Open
Abstract
Severe COVID-19 is rare in children suggesting differences in immune response between children and adults. Limited information is available on how cellular immunity is modulated by COVID-19 vaccination and prior infection, and whether it is differentially modulated in children compared to adults. Here, we aimed to compare COVID-19 vaccine-induced functional T cell response between adults and children with and without previous SARS-CoV-2 infection. Adults (18-45 years; n = 45) and children (5-10 years; n = 51;), who received Pfizer-BioNTech COVID-19 vaccine or remained unvaccinated, and previously infected or not with SARS-CoV-2 were selected from two cross-sectional SARS-CoV-2 serosurveillance studies conducted in Bangladesh. Plasma nucleocapsid (N)-specific antibodies were measured by electrochemiluminescence immunoassay; IFN-γ, perforin and granzyme B secreting T cells were assessed using ELISpot assay. Vaccination in adults without previous infection, induced higher frequencies of IFN-γ and granzyme B secreting T lymphocytes compared to unvaccinated adults, while it increased only IFN-γ expression in vaccinated children. Previous infection increased IFN-γ response in unvaccinated adults only. Unvaccinated children showed higher granzyme B expression compared to adults irrespective of infection status. In vaccinated individuals, prior infection induced perforin expression in both adults and children. Children showed slightly different functional T cell response than adults in response to COVID-19 vaccination and infection. mRNA vaccination provided higher IFN-γ response in both adults and children, but induced cytotoxic T lymphocyte (CTL) response in adults only. Future studies may evaluate the impact of other types of COVID-19 vaccines on functional T cell immunity in children to confirm the findings.
Collapse
Affiliation(s)
- Evana Akhtar
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Rakib Ullah Kuddusi
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Tanvir Talukder
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Jakarea
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Ahsanul Haq
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Md Shamim Hossain
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | | | | | - Rashid U Zaman
- Foreign, Commonwealth and Development Office, British High Commission, Dhaka, 1212, Bangladesh
| | - Abdur Razzaque
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Protim Sarker
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh
| | - Rubhana Raqib
- icddr,b, 68 Shaheed Tajuddin Ahmed Sarani, Mohakhali, Dhaka, 1212, Bangladesh.
| |
Collapse
|
10
|
Lin J, Tang M, Yuan S, Chen J, Yang G, Zhang H, Li W, Zhang J, Zhang L, Yin Y. Local Versus Systemic Cytokine Dynamics in Pediatric Pneumonia. Pediatr Pulmonol 2025; 60:e71093. [PMID: 40243399 DOI: 10.1002/ppul.71093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/27/2025] [Accepted: 04/06/2025] [Indexed: 04/18/2025]
Abstract
No significant correlation was observed between most local and systemic cytokines in children with pneumonia. The observed elevation of specific cytokines such as IL-2, IL-2R, and IL-4 in the BALF of pneumonia patients, alongside increases in IL-2, IL-6, and IL-12p70 in peripheral blood, underscores the robust local and systemic inflammatory response in pneumonia.
Collapse
Affiliation(s)
- Jilei Lin
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Mingyu Tang
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Shuhua Yuan
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Jiande Chen
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Guijun Yang
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Huishan Zhang
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Wanlin Li
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Jing Zhang
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Lei Zhang
- International Medical Department, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
| | - Yong Yin
- Department of Respiratory Medicine, Shanghai Jiaotong University School of Medicine, Shanghai Children's Medical Center, Shanghai, China
- Department of Respiratory Medicine, Hainan Branch of Shanghai Children's Medical Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical College, Sanya City, China
- Department of Respiratory Medicine, Linyi Maternal and Child Healthcare Hospital, Linyi City, China
- Shanghai Children's Medical Center Pediatric Medical Complex (Pudong), Shanghai, China
- Pediatric AI Clinical Application and Research Center, Shanghai Children's Medical Center, Shanghai, China
| |
Collapse
|
11
|
Case JB, Jain S, Suthar MS, Diamond MS. SARS-CoV-2: The Interplay Between Evolution and Host Immunity. Annu Rev Immunol 2025; 43:29-55. [PMID: 39705164 DOI: 10.1146/annurev-immunol-083122-043054] [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] [Indexed: 12/22/2024]
Abstract
The persistence of SARS-CoV-2 infections at a global level reflects the repeated emergence of variant strains encoding unique constellations of mutations. These variants have been generated principally because of a dynamic host immune landscape, the countermeasures deployed to combat disease, and selection for enhanced infection of the upper airway and respiratory transmission. The resulting viral diversity creates a challenge for vaccination efforts to maintain efficacy, especially regarding humoral aspects of protection. Here, we review our understanding of how SARS-CoV-2 has evolved during the pandemic, the immune mechanisms that confer protection, and the impact viral evolution has had on transmissibility and adaptive immunity elicited by natural infection and/or vaccination. Evidence suggests that SARS-CoV-2 evolution initially selected variants with increased transmissibility but currently is driven by immune escape. The virus likely will continue to drift to maintain fitness until countermeasures capable of disrupting transmission cycles become widely available.
Collapse
Affiliation(s)
- James Brett Case
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| | - Shilpi Jain
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mehul S Suthar
- Emory Vaccine Center, Emory National Primate Research Center, Atlanta, Georgia, USA
- Center for Childhood Infections and Vaccines of Children's Healthcare of Atlanta, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Michael S Diamond
- Department of Pathology & Immunology; Department of Molecular Microbiology; and Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Medicine, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
12
|
Kökrek E, Pir P. Distinct deregulation trends of transcriptional protein complexes in aging naive T cells. J Leukoc Biol 2025; 117:qiae231. [PMID: 39437255 DOI: 10.1093/jleuko/qiae231] [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: 08/02/2024] [Revised: 09/19/2024] [Accepted: 10/21/2024] [Indexed: 10/25/2024] Open
Abstract
The impact of aging on T cell subsets, specifically CD4+ and CD8+ T cells, leading to immune system dysfunction has been the focus of scientific investigation due to its potential to reverse age-associated deterioration. Transcriptomic and epigenomic studies have identified the primary regulators in T cell aging. However, comprehending the underlying dynamic mechanisms requires studying these proteins with their interactors. Here, we integrated single-cell RNA sequencing data of naive CD4+ and CD8+ T cells obtained from 3 different age groups with protein-protein and domain-domain interaction networks to predict and compare the transcriptional protein complexes and identify their capacity to explain age-associated variances. Our novel approach revealed significant effects of aging on the repertoire of complexes, which remains unchanged in naive CD4+ T cells, while in naive CD8+ T cells, it diminishes. In both cell types, there was major deregulation of complexes with the same composition, involving a range of transcription factors. This aging-associated deregulation is characterized by a specific set of protein complexes in naive CD4+ T cells, but this pattern is not observed in naive CD8+ T cells. SMAD3 and BCL11A complexes emerge as key markers in defining a trajectory in aging naive CD4+ T cells. These complexes can accurately distinguish between 3 different age groups, indicating their potential as targets. The direct link between SMAD3 and FOS complexes whose regulatory role has been previously implicated in aging and MBD3 as the novel key link between SMAD3 and BCL11A complexes implicates a coordinated mechanism in age-associated deregulation.
Collapse
Affiliation(s)
- Emel Kökrek
- Department of Molecular Biology and Genetics, Kadir Has University, Cibali, Kadir Has Cd., 34083 Fatih/Istanbul, Turkey
- Department of Bioengineering, Gebze Technical University, Cumhuriyet, 2254. Sk. No:2, 41400 Gebze/Kocaeli, Turkey
| | - Pınar Pir
- Department of Bioengineering, Gebze Technical University, Cumhuriyet, 2254. Sk. No:2, 41400 Gebze/Kocaeli, Turkey
| |
Collapse
|
13
|
Hurst JH, Mohan AA, Dalapati T, George IA, Aquino JN, Lugo DJ, Pfeiffer TS, Rodriguez J, Rotta AT, Turner NA, Burke TW, McClain MT, Henao R, DeMarco CT, Louzao R, Denny TN, Walsh KM, Xu Z, Mejias A, Ramilo O, Woods CW, Kelly MS. Age-associated differences in mucosal and systemic host responses to SARS-CoV-2 infection. Nat Commun 2025; 16:2383. [PMID: 40064870 PMCID: PMC11894178 DOI: 10.1038/s41467-025-57655-3] [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: 10/19/2023] [Accepted: 02/26/2025] [Indexed: 03/14/2025] Open
Abstract
Age is among the strongest risk factors for severe outcomes from SARS-CoV-2 infection. Here we describe upper respiratory tract (URT) and peripheral blood transcriptomes of 202 participants (age range of 1 week to 83 years), including 137 non-hospitalized individuals with mild SARS-CoV-2 infection and 65 healthy individuals. Among healthy children and adolescents, younger age is associated with higher URT expression of innate and adaptive immune pathways. SARS-CoV-2 infection induces broad upregulation of URT innate and adaptive immune responses among children and adolescents. Peripheral blood responses among SARS-CoV-2-infected children and adolescents are dominated by interferon pathways, while upregulation of myeloid activation, inflammatory, and coagulation pathways is observed only in adults. Among SARS-CoV-2-infected individuals, fever is associated with blunted URT immune responses and more pronounced systemic immune activation. These findings demonstrate that immune responses to SARS-CoV-2 differ across the lifespan, from distinct signatures in childhood and adolescence to age-associated alterations in adults.
Collapse
Affiliation(s)
- Jillian H Hurst
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Aditya A Mohan
- Department of Biomedical Engineering, Duke University School of Medicine, Durham, NC, USA
| | - Trisha Dalapati
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | - Ian A George
- Duke University School of Medicine, Durham, NC, USA
| | - Jhoanna N Aquino
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Debra J Lugo
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Trevor S Pfeiffer
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Javier Rodriguez
- Children's Clinical Research Unit, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Alexandre T Rotta
- Department of Pediatrics, Division of Pediatric Critical Care Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Nicholas A Turner
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
| | - Thomas W Burke
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Center for Infectious Disease Diagnostics and Innovation, Duke University School of Medicine, Durham, NC, USA
| | - Micah T McClain
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Center for Infectious Disease Diagnostics and Innovation, Duke University School of Medicine, Durham, NC, USA
- Durham Veterans Affairs Medical Center, Durham, NC, USA
| | - Ricardo Henao
- Department of Biostatistics and Informatics, Duke University, Durham, NC, USA
- Duke Clinical Research Institute, Duke University School of Medicine, Durham, NC, USA
| | - C Todd DeMarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Raul Louzao
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kyle M Walsh
- Children's Health and Discovery Institute, Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC, USA
| | - Zhaohui Xu
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Asuncion Mejias
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Octavio Ramilo
- Department of Infectious Diseases, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Christopher W Woods
- Department of Medicine, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA
- Center for Infectious Disease Diagnostics and Innovation, Duke University School of Medicine, Durham, NC, USA
- Durham Veterans Affairs Medical Center, Durham, NC, USA
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Matthew S Kelly
- Department of Pediatrics, Division of Infectious Diseases, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
14
|
van Baarle D, Nawijn MC. Variant-specific local tissue response to SARS-CoV-2 in the nasal mucosa. Nat Immunol 2025; 26:152-154. [PMID: 39880999 DOI: 10.1038/s41590-024-02067-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Affiliation(s)
- Debbie van Baarle
- Department of Medical Microbiology and Infection Prevention, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- Center for Infectious Diseases Prevention, National Institute for Public Health and the Environment, Bilthoven, the Netherlands
| | - Martijn C Nawijn
- Department of Pathology & Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands.
| |
Collapse
|
15
|
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
|
16
|
Walsh JML, Miao VN, Owings AH, Tang Y, Bromley JD, Kazer SW, Kimler K, Asare C, Ziegler CGK, Ibrahim S, Jivanjee T, George M, Navia AW, Drake RS, Parker A, Billingsley BC, Dotherow P, Tarugu S, Kota SK, Laird H, Wichman TG, Davis YT, Dhaliwal NS, Pride Y, Guo Y, Senitko M, Harvey J, Bates JT, Diamond G, Garrett MR, Robinson DA, Frame IJ, Lyons JJ, Robinson TO, Shalek AK, Horwitz BH, Glover SC, Ordovas-Montanes J. Variants and vaccines impact nasal immunity over three waves of SARS-CoV-2. Nat Immunol 2025; 26:294-307. [PMID: 39833605 DOI: 10.1038/s41590-024-02052-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: 04/16/2024] [Accepted: 12/05/2024] [Indexed: 01/22/2025]
Abstract
Viral variant and host vaccination status impact infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), yet how these factors shift cellular responses in the human nasal mucosa remains uncharacterized. We performed single-cell RNA sequencing (scRNA-seq) on nasopharyngeal swabs from vaccinated and unvaccinated adults with acute Delta and Omicron SARS-CoV-2 infections and integrated with data from acute infections with ancestral SARS-CoV-2. Patients with Delta and Omicron exhibited greater similarity in nasal cell composition driven by myeloid, T cell and SARS-CoV-2hi cell subsets, which was distinct from that of ancestral cases. Delta-infected samples had a marked increase in viral RNA, and a subset of PER2+EGR1+GDF15+ epithelial cells was enriched in SARS-CoV-2 RNA+ cells in all variants. Prior vaccination was associated with increased frequency and activation of nasal macrophages. Expression of interferon-stimulated genes negatively correlated with coronavirus disease 2019 (COVID-19) severity in patients with ancestral and Delta but not Omicron variants. Our study defines nasal cell responses and signatures of disease severity across SARS-CoV-2 variants and vaccination.
Collapse
Affiliation(s)
- Jaclyn M L Walsh
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Vincent N Miao
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
| | - Anna H Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Ying Tang
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
| | - Joshua D Bromley
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Graduate Program in Microbiology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Samuel W Kazer
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Department of Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
| | - Kyle Kimler
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Pediatric Hematology/Oncology, Boston Children's Hospital, Boston, MA, USA
| | - Chelsea Asare
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Carly G K Ziegler
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
| | - Samira Ibrahim
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Tasneem Jivanjee
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Micayla George
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Andrew W Navia
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Riley S Drake
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Adam Parker
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | | | - Paul Dotherow
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Spurthi Tarugu
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Sai K Kota
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Hannah Laird
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - T Grant Wichman
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yesenia T Davis
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Neha S Dhaliwal
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yilianys Pride
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Yanglin Guo
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Michal Senitko
- Division of Pulmonary, Critical Care and Sleep Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jessie Harvey
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
| | - John T Bates
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Gill Diamond
- Department of Oral Immunology and Infectious Diseases, University of Louisville School of Dentistry, Louisville, KY, USA
| | - Michael R Garrett
- Department of Medicine, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - D Ashley Robinson
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
| | - I J Frame
- Department of Pathology, University of Mississippi Medical Center, Jackson, MS, USA
| | - Jonathan J Lyons
- Division of Allergy and Immunology, Department of Medicine, University of California San Diego, La Jolla, CA, USA
- Veterans Affairs San Diego Healthcare System, La Jolla, CA, USA
| | - Tanya O Robinson
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
| | - Alex K Shalek
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Program in Health Sciences and Technology, Harvard Medical School and MIT, Boston, MA, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
- Harvard Graduate Program in Biophysics, Cambridge, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Bruce H Horwitz
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA
- Program in Immunology, Harvard Medical School, Boston, MA, USA
- Division of Emergency Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Sarah C Glover
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Cell and Molecular Biology, University of Mississippi Medical Center, Jackson, MS, USA
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University, New Orleans, LA, USA
| | - Jose Ordovas-Montanes
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA.
- Program in Immunology, Harvard Medical School, Boston, MA, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, USA.
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, USA.
- Harvard Stem Cell Institute, Cambridge, MA, USA.
| |
Collapse
|
17
|
Zhou X, Wu Y, Zhu Z, Lu C, Zhang C, Zeng L, Xie F, Zhang L, Zhou F. Mucosal immune response in biology, disease prevention and treatment. Signal Transduct Target Ther 2025; 10:7. [PMID: 39774607 PMCID: PMC11707400 DOI: 10.1038/s41392-024-02043-4] [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: 06/01/2024] [Revised: 09/05/2024] [Accepted: 10/27/2024] [Indexed: 01/11/2025] Open
Abstract
The mucosal immune system, as the most extensive peripheral immune network, serves as the frontline defense against a myriad of microbial and dietary antigens. It is crucial in preventing pathogen invasion and establishing immune tolerance. A comprehensive understanding of mucosal immunity is essential for developing treatments that can effectively target diseases at their entry points, thereby minimizing the overall impact on the body. Despite its importance, our knowledge of mucosal immunity remains incomplete, necessitating further research. The outbreak of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has underscored the critical role of mucosal immunity in disease prevention and treatment. This systematic review focuses on the dynamic interactions between mucosa-associated lymphoid structures and related diseases. We delve into the basic structures and functions of these lymphoid tissues during disease processes and explore the intricate regulatory networks and mechanisms involved. Additionally, we summarize novel therapies and clinical research advances in the prevention of mucosal immunity-related diseases. The review also addresses the challenges in developing mucosal vaccines, which aim to induce specific immune responses while maintaining tolerance to non-pathogenic microbes. Innovative therapies, such as nanoparticle vaccines and inhalable antibodies, show promise in enhancing mucosal immunity and offer potential for improved disease prevention and treatment.
Collapse
Affiliation(s)
- Xiaoxue Zhou
- School of Medicine, Hangzhou City University, Hangzhou, China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Yuchen Wu
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhipeng Zhu
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China
| | - Chu Lu
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China
| | - Chunwu Zhang
- The First School of Medicine, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Linghui Zeng
- School of Medicine, Hangzhou City University, Hangzhou, China
| | - Feng Xie
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| | - Long Zhang
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling Network, Life Sciences Institute, Zhejiang University, Hangzhou, China.
- The MOE Basic Research and Innovation Center for the Targeted Therapeutics of Solid Tumors, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Fangfang Zhou
- The First Affiliated Hospital, the Institutes of Biology and Medical Sciences, Suzhou Medical College, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
18
|
Huang L, Song Z, Lu C, Wang S, Guo C, Lai XH, Zhao Z. A narrative review focusing on randomized clinical trials of vitamin D supplementation for COVID-19 disease. Front Nutr 2025; 11:1461485. [PMID: 39839285 PMCID: PMC11745885 DOI: 10.3389/fnut.2024.1461485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/01/2024] [Indexed: 01/23/2025] Open
Abstract
Current evidence is inconsistent on whether vitamin D supplementation can prevent COVID-19 infection or improve its clinical outcomes. To better understand and look into the issue, we went through the background knowledge of COVID-19 and vitamin D, searched in Pubmed [by using key words in the title containing "randomized clinical trial", "COVID-19", and "vitamin D (25-hydroxyvitamin D, or cholecalciferol, or calcidiol, or calcifediol) supplementation"] for publications of studies on vitamin D/supplementation in COVID-19 patients, especially those about the randomized clinical trials (RCTs). After reviewing these papers, we did a short background review of vitamin D and the pathophysiology of COVID-19, summarized the key features of the 25 RCTs in text and tabulated in a table of some of the features, commented, compared and discussed the differences between RCTs (for example, change the serum 25-hydroxyvitamin D concentration from nmol/L to ng/mL, making the comparison easier). The take-home question of the review is that serum 25-hydroxyvitamin D concentration is an important indicator of the supplementation effect of vitamin D correction but may not be reliable in predicting the supplementation effect on the clinical outcomes of COVID-19.
Collapse
Affiliation(s)
- Limi Huang
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhiwei Song
- Department of Infection Diseases, Xianju County People's Hospital, Taizhou, Zhejiang, China
| | - Chaosheng Lu
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Shenwen Wang
- School of Information Engineering, Hebei GEO University, Shijiazhuang, Hebei, China
| | - Changsheng Guo
- Shaoxing BWK Biotechnology Co., Ltd., Zhuji City High-Tech Entrepreneurship Center, Shaoxing, Zhejiang, China
| | - Xin-He Lai
- Department of Pediatrics, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Shenzhen Boya Gene Technology Co., Ltd., Shenzhen, China
| | - Zhenfeng Zhao
- Hebei Huiji Technology Co., Ltd., Shijiazhuang, Hebei, China
| |
Collapse
|
19
|
Valencia-Gutiérrez MM, Gómez-López M, Pérez-Vielma NM, Lázaro-Aguilar P, Aguilera-Sosa VR. [Clinical characteristics and cytokine gene expression in patients with post-COVID-19 syndrome]. REVISTA MEDICA DEL INSTITUTO MEXICANO DEL SEGURO SOCIAL 2025; 63:e6275. [PMID: 40267303 PMCID: PMC12080597 DOI: 10.5281/zenodo.14200098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/31/2024] [Indexed: 04/25/2025]
Abstract
Background Post-COVID-19 syndrome occurs 3 months after COVID-19 infection and lasts at least 2 months. There is insufficient information on the genes associated with immune system dysregulation. Objective To evaluate gene expression and its relationship with post-COVID-19 syndrome. Material and methods Cross-sectional, retrolective and analytical study which included 56 patients with post-COVID-19 syndrome. Clinical characteristics were recorded and serotonin IL-4, IL-1β, SOCS3, ILF13, and IFNL4 genes were analyzed with TRIzol® Reagent and PCR-RT techniques. Results The prevalence of post-COVID-19 syndrome was 82.1%, with no differences in relation to the severity of symptoms or comorbidities. The clinical characteristics related to the presence of the syndrome were female sex with an odds ratio (OR) of 4.25 (95% confidence interval [95% CI] 1.02-17.69), and drug consumption with an OR of 8.25 (95% CI 0.97-70.50). Protective factors for fatigue were serotonin expression with an OR of 0.238 (95% CI 0.060-0.949); for concentration problems, SOCS3 with an OR of 0.188 (95% CI 0.037-0.946), and for memory impairment the IFNL4 with an OR of 0.094 (95% CI 0.015-0.586). Conclusions The associated factors with post-COVID-19 syndrome were female sex, drug use, and gene dysregulation of serotonin, INFL4 and SOCS3.
Collapse
Affiliation(s)
- María Magdalena Valencia-Gutiérrez
- Instituto Mexicano del Seguro Social, Órgano de Operación Administrativa Desconcentrada Michoacán, Hospital General de Zona No. 83, Servicio de Urgencias. Morelia, Michoacán, MéxicoInstituto Mexicano del Seguro SocialMéxico
| | - Modesto Gómez-López
- Instituto Politécnico Nacional, Escuela Superior de Medicina, Área de Posgrado Biología Molecular del Proceso Inflamatorio. Ciudad de México, MéxicoInstituto Politécnico NacionalMéxico
| | - Nadia Mabel Pérez-Vielma
- Instituto Politécnico Nacional, Centro Interdisciplinario de Ciencias de la Salud-Unidad Santo Tomás, Área de Posgrado. Ciudad de México, México Instituto Politécnico NacionalMéxico
| | - Paulina Lázaro-Aguilar
- Instituto Politécnico Nacional, Centro Interdisciplinario de Ciencias de la Salud-Unidad Santo Tomás, Área de Posgrado. Ciudad de México, México Instituto Politécnico NacionalMéxico
| | - Víctor Ricardo Aguilera-Sosa
- Instituto Politécnico Nacional, Centro Interdisciplinario de Ciencias de la Salud-Unidad Santo Tomás, Área de Posgrado. Ciudad de México, México Instituto Politécnico NacionalMéxico
| |
Collapse
|
20
|
Tan CL, Lindner K, Boschert T, Meng Z, Rodriguez Ehrenfried A, De Roia A, Haltenhof G, Faenza A, Imperatore F, Bunse L, Lindner JM, Harbottle RP, Ratliff M, Offringa R, Poschke I, Platten M, Green EW. Prediction of tumor-reactive T cell receptors from scRNA-seq data for personalized T cell therapy. Nat Biotechnol 2025; 43:134-142. [PMID: 38454173 PMCID: PMC11738991 DOI: 10.1038/s41587-024-02161-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2023] [Accepted: 02/01/2024] [Indexed: 03/09/2024]
Abstract
The identification of patient-derived, tumor-reactive T cell receptors (TCRs) as a basis for personalized transgenic T cell therapies remains a time- and cost-intensive endeavor. Current approaches to identify tumor-reactive TCRs analyze tumor mutations to predict T cell activating (neo)antigens and use these to either enrich tumor infiltrating lymphocyte (TIL) cultures or validate individual TCRs for transgenic autologous therapies. Here we combined high-throughput TCR cloning and reactivity validation to train predicTCR, a machine learning classifier that identifies individual tumor-reactive TILs in an antigen-agnostic manner based on single-TIL RNA sequencing. PredicTCR identifies tumor-reactive TCRs in TILs from diverse cancers better than previous gene set enrichment-based approaches, increasing specificity and sensitivity (geometric mean) from 0.38 to 0.74. By predicting tumor-reactive TCRs in a matter of days, TCR clonotypes can be prioritized to accelerate the manufacture of personalized T cell therapies.
Collapse
Affiliation(s)
- C L Tan
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - K Lindner
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases, Heidelberg, Germany
| | - T Boschert
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz, Germany
| | - Z Meng
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - A Rodriguez Ehrenfried
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- Helmholtz Institute for Translational Oncology, Mainz, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
| | - A De Roia
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
- DNA Vector Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - G Haltenhof
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
| | | | | | - L Bunse
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany
| | | | - R P Harbottle
- DNA Vector Laboratory, German Cancer Research Center, Heidelberg, Germany
| | - M Ratliff
- Department of Neurosurgery, University Hospital Mannheim, Mannheim, Germany
| | - R Offringa
- Department of General, Visceral and Transplantation Surgery, University Hospital Heidelberg, Heidelberg, Germany
- Division of Molecular Oncology of Gastrointestinal Tumors, German Cancer Research Center, Heidelberg, Germany
- Sino-German Laboratory of Personalized Medicine for Pancreatic Cancer, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - I Poschke
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany
- Immune Monitoring Unit, National Center for Tumor Diseases, Heidelberg, Germany
| | - M Platten
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany.
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany.
- Immune Monitoring Unit, National Center for Tumor Diseases, Heidelberg, Germany.
- Helmholtz Institute for Translational Oncology, Mainz, Germany.
- German Cancer Research Center-Hector Cancer Institute at the Medical Faculty Mannheim, University of Heidelberg, Mannheim, Germany.
| | - E W Green
- CCU Neuroimmunology and Brain Tumor Immunology, German Cancer Research Center, Heidelberg, Germany.
- German Cancer Consortium, Core Center Heidelberg, Heidelberg, Germany.
- Department of Neurology, Medical Faculty Mannheim, Mannheim Center for Translational Neuroscience, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
21
|
Bagala I, Namuganga JF, Nayebare P, Cuu G, Katairo T, Nabende I, Gonahasa S, Nassali M, Tukwasibwe S, Dorsey G, Nankabirwa JI, Bakeera-Kitaka S, Kiguli S, Greenhouse B, Ssewanyana I, Kamya MR, Briggs J. Seroprevalence of SARS-CoV-2 and risk factors for infection among children in Uganda: A serial cross-sectional study. PLoS One 2024; 19:e0312554. [PMID: 39715211 PMCID: PMC11665985 DOI: 10.1371/journal.pone.0312554] [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: 06/21/2024] [Accepted: 10/07/2024] [Indexed: 12/25/2024] Open
Abstract
BACKGROUND Understanding COVID-19's impact on children is vital for public health policy, yet age-specific data is scarce, especially in Uganda. This study examines SARS-CoV-2 seroprevalence and risk factors among Ugandan children at two timepoints, along with COVID-19-related knowledge and practices in households, including adult vaccination status. METHODS Baseline surveys were conducted in 12 communities from April to May 2021 (post-Alpha wave) and follow-up surveys in 32 communities from November 2021 to March 2022 (Omicron wave). Household questionnaires and blood samples were collected to test for malaria by microscopy and for SARS-CoV-2 using a Luminex assay. Seroprevalence was estimated at both the survey and community level. Mixed-effects logistic regression models assessed the association between individual and household factors and SARS-CoV-2 seropositivity in children, adjusting for household clustering. RESULTS More households reported disruptions in daily life at baseline compared to follow-up, though economic impacts lingered. By the follow-up survey, 52.7% of adults had received at least one COVID-19 vaccine dose. Overall seroprevalence in children was higher at follow-up compared to baseline (71.6% versus 19.2%, p < 0.001). Seroprevalence in children ranged across communities from 6-37% at baseline and 50-90% at follow-up. At baseline, children from the poorest households were more likely to be infected. Increasing age remained the only consistent risk factor for SARS-CoV-2 seroconversion at both timepoints. CONCLUSIONS Results indicate that a larger number of children were infected during the Delta and Omicron waves of COVID-19 compared to the Alpha wave. This study is the largest seroprevalence survey in children in Uganda, providing evidence that most children were infected with SARS-CoV-2 before the vaccine was widely available to pediatric populations. Pediatric infections were vastly underreported by case counts, highlighting the importance of seroprevalence surveys in assessing disease burden when testing and reporting rates are limited and many cases are mild or asymptomatic.
Collapse
Affiliation(s)
- Irene Bagala
- Makerere University College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Gloria Cuu
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Isaiah Nabende
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Martha Nassali
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Grant Dorsey
- University of California, San Francisco, CA, United States of America
| | - Joaniter I. Nankabirwa
- Makerere University College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Sarah Kiguli
- Makerere University College of Health Sciences, Kampala, Uganda
| | - Bryan Greenhouse
- University of California, San Francisco, CA, United States of America
| | - Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Central Public Health Laboratory, Butabika, Uganda
| | - Moses R. Kamya
- Makerere University College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Jessica Briggs
- University of California, San Francisco, CA, United States of America
| |
Collapse
|
22
|
Dassaye R, Chetty T, Daniels B, Gaffoor Z, Spooner E, Ramraj T, Mthethwa N, Nsibande DF, Pillay S, Bhana A, Magasana V, Reddy T, Mohlabi K, Moore PL, Burgers WA, de Oliveira T, Msomi N, Goga A. SARS-CoV-2 Infections in a Triad of Primary School Learners (Grades 1-7), Their Parents, and Teachers in KwaZulu-Natal, South Africa: Protocol for a Cross-Sectional and Nested Case-Cohort Study. JMIR Res Protoc 2024; 13:e52713. [PMID: 39700491 PMCID: PMC11695960 DOI: 10.2196/52713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 07/29/2024] [Accepted: 09/16/2024] [Indexed: 12/21/2024] Open
Abstract
BACKGROUND In low- and middle-income countries (LMICs) such as South Africa, there is paucity of data on SARS-CoV-2 infections among children attending school, including seroprevalence and transmission dynamics. OBJECTIVE This pilot study aims to assess (1) the prevalence of self-reported or confirmed SARS-CoV-2 prior infections, COVID-19 symptoms (including long COVID), seroprevalence of SARS-CoV-2 antibodies, and general/mental health, (2) longitudinal changes in SARS-CoV-2 seroprevalence, and (3) SARS-CoV-2 acute infections, immune responses, transmission dynamics, and symptomatic versus asymptomatic contacts in a unique cohort of unvaccinated primary school learners, their parents, teachers, and close contacts in semirural primary school settings. METHODS Learners (grades 1-7) from primary schools in KwaZulu-Natal, South Africa, their parents, and teachers will be invited to enroll into the COVID kids school study (CoKiDSS). CoKiDSS comprises 3 parts: a cross-sectional survey (N=640), a follow-up survey (n=300), and a nested case-cohort substudy. Finger-prick blood and saliva samples will be collected for serological and future testing, respectively, in the cross-sectional (451 learners:147 parents:42 teachers) and follow-up (210 learners:70 parents:20 teachers) surveys. The nested case-cohort substudy will include cases from the cross-sectional survey with confirmed current SARS-CoV-2 infection (n=30) and their close contacts (n=up to 10 per infected participant). Finger-prick blood (from all substudy participants), venous blood (from cases), and nasal swabs (from cases and contacts) will be collected for serological testing, immunological testing, and viral genome sequencing, respectively. Questionnaires covering sociodemographic and general and mental health information, prior and current SARS-CoV-2 symptoms and testing information, vaccination status, preventative behavior, and lifestyle will be administered. Statistical methods will include generalized linear mixed models, intracluster correlation, descriptive analysis, and graphical techniques. RESULTS A total of 645 participants were enrolled into the cross-sectional survey between May and August 2023. A subset of 300 participants were followed up in the follow-up survey in October 2023. Screening of the participants into the nested case-cohort substudy is planned between November 2023 and September 2024. Data cleanup and analysis for the cross-sectional survey is complete, while those for the follow-up survey and nested case substudy will be completed by the third quarter of 2024. The dissemination and publication of results is anticipated for the fourth quarter of 2024. CONCLUSIONS This study provides data from an LMIC setting on the impact of SARS-CoV-2 on school-attending learners, their parents, and teachers 3 years after the SARS-CoV-2 pandemic was declared and 21-24 months after resumption of normal school attendance. In particular, this study will provide data on the prevalence of self-reported or confirmed SARS-CoV-2 prior infection, prior and current symptoms, seroprevalence, changes in seroprevalence, SARS-CoV-2 transmission, SARS-CoV-2 adaptive immune responses, and symptoms of long COVID and mental health among a triad of learners, their parents, and teachers. INTERNATIONAL REGISTERED REPORT IDENTIFIER (IRRID) DERR1-10.2196/52713.
Collapse
Affiliation(s)
- Reshmi Dassaye
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Terusha Chetty
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Discipline of Public Health Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - Brodie Daniels
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Zakir Gaffoor
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Elizabeth Spooner
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Trisha Ramraj
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Ncengani Mthethwa
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Duduzile Faith Nsibande
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Saresha Pillay
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Arvin Bhana
- Center for Rural Health, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, South Africa
- Health Systems Research Unit, South African Medical Research Council, Durban, South Africa
| | - Vuyolwethu Magasana
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Tarylee Reddy
- Biostatistics Research Unit, South African Medical Research Council, Durban, South Africa
| | - Khanya Mohlabi
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
| | - Penelope Linda Moore
- Centre for the AIDS Programme of Research in South Africa, Durban, South Africa
- National Institute for Communicable Diseases of the National Health Laboratory Service, Johannesburg, South Africa
- South African Medical Research Council Antibody Immunity Research Unit, School of Pathology, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Wendy A Burgers
- Wellcome Centre for Infectious Diseases Research in Africa, University of Cape Town, Cape Town, South Africa
- Division of Medical Virology, Department of Pathology, University of Cape Town, Cape Town, South Africa
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Tulio de Oliveira
- Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
- Centre for Epidemic Response and Innovation, School of Data Science and Computational Thinking, Stellenbosch University, Stellenbosch, South Africa
- KwaZulu-Natal Research Innovation and Sequencing Platform, Nelson R Mandela School of Medicine, University of KwaZulu-Natal, Durban, South Africa
- Department of Global Health, University of Washington, Seattle, WA, United States
| | - Nokukhanya Msomi
- Discipline of Virology, School of Laboratory Medicine and Medical Sciences, University of KwaZulu Natal, Durban, South Africa
- National Health Laboratory Service, Durban, South Africa
| | - Ameena Goga
- HIV and Other Infectious Diseases Research Unit, South African Medical Research Council, Cape Town, South Africa
- Department of Paediatrics and Child Health, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
23
|
Oh DS, Kim E, Normand R, Lu G, Shook LL, Lyall A, Jasset O, Demidkin S, Gilbert E, Kim J, Akinwunmi B, Tantivit J, Tirard A, Arnold BY, Slowikowski K, Goldberg MB, Filbin MR, Hacohen N, Nguyen LH, Chan AT, Yu XG, Li JZ, Yonker L, Fasano A, Perlis RH, Pasternak O, Gray KJ, Choi GB, Drew DA, Sen P, Villani AC, Edlow AG, Huh JR. SARS-CoV-2 infection elucidates features of pregnancy-specific immunity. Cell Rep 2024; 43:114933. [PMID: 39504241 PMCID: PMC11724703 DOI: 10.1016/j.celrep.2024.114933] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 07/30/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
Pregnancy is a risk factor for increased severity of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and other respiratory infections, but the mechanisms underlying this risk are poorly understood. To gain insight into the role of pregnancy in modulating immune responses at baseline and upon SARS-CoV-2 infection, we collected peripheral blood mononuclear cells and plasma from 226 women, including 152 pregnant individuals and 74 non-pregnant women. We find that SARS-CoV-2 infection is associated with altered T cell responses in pregnant women, including a clonal expansion of CD4-expressing CD8+ T cells, diminished interferon responses, and profound suppression of monocyte function. We also identify shifts in cytokine and chemokine levels in the sera of pregnant individuals, including a robust increase of interleukin-27, known to drive T cell exhaustion. Our findings reveal nuanced pregnancy-associated immune responses, which may contribute to the increased susceptibility of pregnant individuals to viral respiratory infection.
Collapse
Affiliation(s)
- Dong Sun Oh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Eunha Kim
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; BK21 Graduate Program, Department of Biomedical Sciences and Department of Neuroscience, Korea University College of Medicine, Seoul 02841, Republic of Korea
| | - Rachelly Normand
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Guangqing Lu
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lydia L Shook
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Amanda Lyall
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Olyvia Jasset
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Stepan Demidkin
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Emily Gilbert
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Joon Kim
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Babatunde Akinwunmi
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jessica Tantivit
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Alice Tirard
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Benjamin Y Arnold
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA
| | - Kamil Slowikowski
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA
| | - Marcia B Goldberg
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Infectious Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Microbiology, Harvard Medical School, Boston, MA 02115, USA; Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Michael R Filbin
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Emergency Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Nir Hacohen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA
| | - Long H Nguyen
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Andrew T Chan
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA
| | - Xu G Yu
- Infectious Disease Division, Brigham and Women's Hospital, Boston, MA 02115, USA; Ragon Institute of MGH, MIT and Harvard, Cambridge, MA 02139, USA
| | - Jonathan Z Li
- Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Lael Yonker
- Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alessio Fasano
- Division of Pediatric Gastroenterology and Nutrition, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Roy H Perlis
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Ofer Pasternak
- Department of Psychiatry, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA; Department of Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Kathryn J Gray
- Department of Obstetrics & Gynecology, University of Washington, Seattle, WA 98195, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - David A Drew
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| | - Pritha Sen
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA; Transplant, Oncology, and Immunocompromised Host Group, Division of Infectious Diseases, Brigham and Women's Hospital, Boston, MA 02115, USA; Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Alexandra-Chloé Villani
- Center for Immunology and Inflammatory Diseases, Department of Medicine, Massachusetts General Hospital, Boston, MA 02129, USA; Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, MA 02114, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Harvard Medical School, Boston, MA 02115, USA.
| | - Andrea G Edlow
- Department of Obstetrics, Gynecology and Reproductive Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA; Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA 02114, USA.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
24
|
Vela LC, Carrere L, Naasz C, Kalavacherla S, Tan TS, de Armas L, Gao C, Yu XG, Pahwa SG, Luzuriaga K, Lichterfeld M. Profound reduction of HIV-1 reservoir cells over 3 decades of antiretroviral therapy started in early infancy. JCI Insight 2024; 10:e186550. [PMID: 39541163 PMCID: PMC11721289 DOI: 10.1172/jci.insight.186550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 11/08/2024] [Indexed: 11/16/2024] Open
Abstract
HIV-1 reservoir cells persist indefinitely during suppressive antiretroviral therapy (ART) in individuals who acquire infection in adulthood, but little is known about the longitudinal evolution of viral reservoir cells during long-term ART started during early infancy. We studied 2 fraternal twins who acquired HIV-1 perinatally, started ART at week 10 after birth and remained on ART for 28 years. We observed that the frequency of genome-intact proviruses, determined by single-genome near-full-length proviral sequencing, declined by approximately 4,000- to 13,000-fold during this period, indicating enhanced decay rates of intact proviruses even after adjusting for dilution effects from somatic growth. Despite analyzing more than one billion PBMC after 28 years of ART in each participant, no intact proviruses were detected in 1 participant, and 1 intact provirus was isolated in the other. The longitudinal decline of defective proviruses in the 2 participants was more similar to proviral decay kinetics reported in individuals who started ART during adulthood; moreover, clonal sequence clusters were readily detectable for defective proviruses but not for intact proviruses after 28 years of ART in the 2 twins. Together, these data suggest decreased long-term stability and increased immunological vulnerability of intact proviruses during long-term ART started in early infancy.
Collapse
Affiliation(s)
- Liliana C. Vela
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Leah Carrere
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Chloe Naasz
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Sruthi Kalavacherla
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Toong Seng Tan
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Lesley de Armas
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Ce Gao
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
| | - Xu G. Yu
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| | - Savita G. Pahwa
- Department of Microbiology and Immunology, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Katherine Luzuriaga
- Program in Molecular Medicine, University of Massachusetts Chan School of Medicine, Worcester, Massachusetts, USA
| | - Mathias Lichterfeld
- Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts, USA
- Infectious Disease Division, Brigham and Women’s Hospital, Boston, Massachusetts, USA
| |
Collapse
|
25
|
Zhao C, Bai Y, Wang W, Amonkar GM, Mou H, Olejnik J, Hume AJ, Mühlberger E, Lukacs NW, Fearns R, Lerou PH, Ai X. Activation of STAT3-mediated ciliated cell survival protects against severe infection by respiratory syncytial virus. J Clin Invest 2024; 134:e183978. [PMID: 39484716 PMCID: PMC11527452 DOI: 10.1172/jci183978] [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: 06/17/2024] [Accepted: 08/15/2024] [Indexed: 11/03/2024] Open
Abstract
Respiratory syncytial virus (RSV) selectively targets ciliated cells in human bronchial epithelium and can cause bronchiolitis and pneumonia, mostly in infants. To identify molecular targets of intervention during RSV infection in infants, we investigated how age regulates RSV interaction with the bronchial epithelium barrier. Employing precision-cut lung slices and air-liquid interface cultures generated from infant and adult human donors, we found robust RSV virus spread and extensive apoptotic cell death only in infant bronchial epithelium. In contrast, adult bronchial epithelium showed no barrier damage and limited RSV infection. Single nuclear RNA-Seq revealed age-related insufficiency of an antiapoptotic STAT3 activation response to RSV infection in infant ciliated cells, which was exploited to facilitate virus spread via the extruded apoptotic ciliated cells carrying RSV. Activation of STAT3 and blockade of apoptosis rendered protection against severe RSV infection in infant bronchial epithelium. Lastly, apoptotic inhibitor treatment of a neonatal mouse model of RSV infection mitigated infection and inflammation in the lung. Taken together, our findings identify a STAT3-mediated antiapoptosis pathway as a target to battle severe RSV disease in infants.
Collapse
Affiliation(s)
- Caiqi Zhao
- Division of Newborn Medicine, Department of Pediatrics and
| | - Yan Bai
- Division of Newborn Medicine, Department of Pediatrics and
| | - Wei Wang
- Division of Newborn Medicine, Department of Pediatrics and
| | | | - Hongmei Mou
- The Mucosal Immunology and Biology Research Center, Massachusetts General Hospital for Children, Boston, Massachusetts, USA
| | - Judith Olejnik
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Adam J. Hume
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Elke Mühlberger
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
- National Emerging Infectious Diseases Laboratories, Boston University, Boston, Massachusetts, USA
| | - Nicholas W. Lukacs
- Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, Michigan, USA
| | - Rachel Fearns
- Department of Virology, Immunology & Microbiology, Boston University Chobanian & Avedisian School of Medicine, Boston, Massachusetts, USA
| | - Paul H. Lerou
- Division of Newborn Medicine, Department of Pediatrics and
| | - Xingbin Ai
- Division of Newborn Medicine, Department of Pediatrics and
| |
Collapse
|
26
|
Hoytema van Konijnenburg DP, Nigrovic PA, Zanoni I. Regional specialization within the mammalian respiratory immune system. Trends Immunol 2024; 45:871-891. [PMID: 39438172 PMCID: PMC11560516 DOI: 10.1016/j.it.2024.09.011] [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: 07/17/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The respiratory tract is exposed to infection from inhaled pathogens, including viruses, bacteria, and fungi. So far, a comprehensive assessment that integrates common and distinct aspects of the immune response along different areas of the respiratory tract has been lacking. Here, we discuss key recent findings regarding anatomical, functional, and microbial factors driving regional immune adaptation in the mammalian respiratory system, how they differ between mice and humans, and the similarities and differences with the gastrointestinal tract. We demonstrate that, under evolutionary pressure, mammals evolved spatially organized immune defenses that vary between the upper and lower respiratory tract. Overall, we propose that the functional specialization of the immune response along the respiratory tract has fundamental implications for the management of infectious or inflammatory diseases.
Collapse
Affiliation(s)
| | - Peter A Nigrovic
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Rheumatology, Inflammation, and Immunity, Brigham and Women's Hospital, Boston, MA, USA
| | - Ivan Zanoni
- Division of Immunology, Boston Children's Hospital, and Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
27
|
Ocón B, Xiang M, Bi Y, Tan S, Brulois K, Ayesha A, Kunte M, Zhou C, LaJevic M, Lazarus N, Mengoni F, Sharma T, Montgomery S, Hooper JE, Huang M, Handel T, Dawson JRD, Kufareva I, Zabel BA, Pan J, Butcher EC. A lymphocyte chemoaffinity axis for lung, non-intestinal mucosae and CNS. Nature 2024; 635:736-745. [PMID: 39293486 PMCID: PMC11887596 DOI: 10.1038/s41586-024-08043-2] [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: 03/12/2024] [Accepted: 09/12/2024] [Indexed: 09/20/2024]
Abstract
Tissue-selective chemoattractants direct lymphocytes to epithelial surfaces to establish local immune environments, regulate immune responses to food antigens and commensal organisms, and protect from pathogens. Homeostatic chemoattractants for small intestines, colon and skin are known1,2, but chemotropic mechanisms selective for respiratory tract and other non-intestinal mucosal tissues remain poorly understood. Here we leveraged diverse omics datasets to identify GPR25 as a lymphocyte receptor for CXCL17, a chemoattractant cytokine whose expression by epithelial cells of airways, upper gastrointestinal and squamous mucosae unifies the non-intestinal mucosal tissues and distinguishes them from intestinal mucosae. Single-cell transcriptomic analyses show that GPR25 is induced on innate lymphocytes before emigration to the periphery, and is imprinted in secondary lymphoid tissues on activated B and T cells responding to immune challenge. GPR25 characterizes B and T tissue resident memory cells and regulatory T lymphocytes in non-intestinal mucosal tissues and lungs in humans and mediates lymphocyte homing to barrier epithelia of the airways, oral cavity, stomach, and biliary and genitourinary tracts in mouse models. GPR25 is also expressed by T cells in cerebrospinal fluid and CXCL17 by neurons, suggesting a role in central nervous system (CNS) immune regulation. We reveal widespread imprinting of GPR25 on regulatory T cells, suggesting a mechanistic link to population genetics evidence that GPR25 is protective in autoimmunity3,4. Our results define a GPR25-CXCL17 chemoaffinity axis with the potential to integrate immunity and tolerance at non-intestinal mucosae and the CNS.
Collapse
Affiliation(s)
- Borja Ocón
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Menglan Xiang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Yuhan Bi
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA.
| | - Serena Tan
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kevin Brulois
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Aiman Ayesha
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Manali Kunte
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Catherine Zhou
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Melissa LaJevic
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Nicole Lazarus
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Francesca Mengoni
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, Verona, Italy
- Department of Engineering for Innovation Medicine, University of Verona, Verona, Italy
| | - Tanya Sharma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Stephen Montgomery
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jody E Hooper
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Mian Huang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Tracy Handel
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - John R D Dawson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Irina Kufareva
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Brian A Zabel
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Junliang Pan
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Eugene C Butcher
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
28
|
Zakar-Polyák E, Csordas A, Pálovics R, Kerepesi C. Profiling the transcriptomic age of single-cells in humans. Commun Biol 2024; 7:1397. [PMID: 39462118 PMCID: PMC11513945 DOI: 10.1038/s42003-024-07094-5] [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: 01/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Although aging clocks predicting the age of individual organisms have been extensively studied, the age of individual cells remained largely unexplored. Most recently single-cell omics clocks were developed for the mouse, however, extensive profiling the age of human cells is still lacking. To fill this gap, here we use available scRNA-seq data of 1,058,909 blood cells of 508 healthy, human donors (between 19 and 75 years), for developing single-cell transcriptomic clocks and predicting the age of human blood cells. By the application of the proposed cell-type-specific single-cell clocks, our main observations are that (i) transcriptomic age is associated with cellular senescence; (ii) the transcriptomic age of classical monocytes as well as naive B and T cells is decreased in moderate COVID-19 followed by an increase for some cell types in severe COVID-19; and (iii) the human embryo cells transcriptomically rejuvenated at the morulae and blastocyst stages. In summary, here we demonstrate that single-cell transcriptomic clocks are useful tools to investigate aging and rejuvenation at the single-cell level.
Collapse
Affiliation(s)
- Enikő Zakar-Polyák
- Institute for Computer Science and Control (SZTAKI), Hungarian Research Network (HUN-REN), Budapest, Hungary.
- Doctoral School of Informatics, Eötvös Loránd University, Budapest, Hungary.
| | - Attila Csordas
- AgeCurve Limited, Cambridge, UK
- Doctoral School of Clinical Medicine, University of Szeged, Szeged, Hungary
| | - Róbert Pálovics
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Csaba Kerepesi
- Institute for Computer Science and Control (SZTAKI), Hungarian Research Network (HUN-REN), Budapest, Hungary.
| |
Collapse
|
29
|
Kelly MS, Shi P, Boiditswe SC, Qin E, Steenhoff AP, Mazhani T, Patel MZ, Cunningham CK, Rawls JF, Luinstra K, Gilchrist J, Maciejewski J, Hurst JH, Seed PC, Bulir D, Smieja M. The role of the microbiota in respiratory virus-bacterial pathobiont relationships in the upper respiratory tract. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.10.22.24315478. [PMID: 39502658 PMCID: PMC11537323 DOI: 10.1101/2024.10.22.24315478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/15/2024]
Abstract
The mechanisms by which respiratory viruses predispose to secondary bacterial infections remain poorly characterized. Using 2,409 nasopharyngeal swabs from 300 infants in Botswana, we performed a detailed analysis of factors that influence the dynamics of bacterial pathobiont colonization during infancy. We quantify the extent to which viruses increase the acquisition of Haemophilus influenzae, Moraxella catarrhalis, and Streptococcus pneumoniae. We provide evidence of cooperative interactions between these pathobionts while identifying host characteristics and environmental exposures that influence the odds of pathobiont colonization during early life. Using 16S rRNA gene sequencing, we demonstrate that respiratory viruses result in losses of putatively beneficial Corynebacterium and Streptococcus species that are associated with a lower odds of pathobiont acquisition. These findings provide novel insights into viral-bacterial relationships in the URT of direct relevance to respiratory infections and suggest that the URT bacterial microbiota is a potentially modifiable mechanism by which viruses promote bacterial respiratory infections.
Collapse
Affiliation(s)
- Matthew S. Kelly
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Division of Pediatric Infectious Diseases, Duke University, Durham, North Carolina, United States
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States
| | - Pixu Shi
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, United States
| | | | - Emily Qin
- Department of Biostatistics and Bioinformatics, Duke University, Durham, North Carolina, United States
| | - Andrew P. Steenhoff
- Botswana-University of Pennsylvania Partnership, Gaborone, Botswana
- Global Health Center, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
- Division of Pediatric Infectious Diseases, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States
| | - Tiny Mazhani
- University of Botswana School of Medicine, Gaborone, Botswana
| | | | - Coleen K. Cunningham
- Division of Pediatric Infectious Diseases, University of California, Irvine, Children’s Hospital of Orange County, Orange, California, United States
| | - John F. Rawls
- Department of Molecular Genetics and Microbiology, Duke University, Durham, North Carolina, United States
| | - Kathy Luinstra
- Infectious Disease Research Group, Research Institute of St. Joe’s Hamilton, Hamilton, Ontario, Canada
| | - Jodi Gilchrist
- Infectious Disease Research Group, Research Institute of St. Joe’s Hamilton, Hamilton, Ontario, Canada
| | - Julia Maciejewski
- Infectious Disease Research Group, Research Institute of St. Joe’s Hamilton, Hamilton, Ontario, Canada
| | - Jillian H. Hurst
- Division of Pediatric Infectious Diseases, Duke University, Durham, North Carolina, United States
| | - Patrick C. Seed
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, Illinois, USA
| | - David Bulir
- Department of Chemical Engineering, McMaster University, Hamilton, Ontario, Canada
| | - Marek Smieja
- Infectious Disease Research Group, Research Institute of St. Joe’s Hamilton, Hamilton, Ontario, Canada
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ontario, Canada
| |
Collapse
|
30
|
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
|
31
|
Hartmann CR, Khan R, Schöning J, Richter M, Willers M, Pirr S, Heckmann J, Dirks J, Morbach H, Konrad M, Fries E, Winkler M, Büchel J, Seidenspinner S, Fischer J, Vollmuth C, Meinhardt M, Marissen J, Schmolke M, Haid S, Pietschmann T, Backes S, Dölken L, Löber U, Keil T, Heuschmann PU, Wöckel A, Sagar, Ulas T, Forslund-Startceva SK, Härtel C, Viemann D. A clinical protocol for a German birth cohort study of the Maturation of Immunity Against respiratory viral Infections (MIAI). Front Immunol 2024; 15:1443665. [PMID: 39355253 PMCID: PMC11442434 DOI: 10.3389/fimmu.2024.1443665] [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: 06/05/2024] [Accepted: 08/29/2024] [Indexed: 10/03/2024] Open
Abstract
Introduction Respiratory viral infections (RVIs) are a major global contributor to morbidity and mortality. The susceptibility and outcome of RVIs are strongly age-dependent and show considerable inter-population differences, pointing to genetically and/or environmentally driven developmental variability. The factors determining the age-dependency and shaping the age-related changes of human anti-RVI immunity after birth are still elusive. Methods We are conducting a prospective birth cohort study aiming at identifying endogenous and environmental factors associated with the susceptibility to RVIs and their impact on cellular and humoral immune responses against the influenza A virus (IAV), respiratory syncytial virus (RSV) and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The MIAI birth cohort enrolls healthy, full-term neonates born at the University Hospital Würzburg, Germany, with follow-up at four defined time-points during the first year of life. At each study visit, clinical metadata including diet, lifestyle, sociodemographic information, and physical examinations, are collected along with extensive biomaterial sampling. Biomaterials are used to generate comprehensive, integrated multi-omics datasets including transcriptomic, epigenomic, proteomic, metabolomic and microbiomic methods. Discussion The results are expected to capture a holistic picture of the variability of immune trajectories with a focus on cellular and humoral key players involved in the defense of RVIs and the impact of host and environmental factors thereon. Thereby, MIAI aims at providing insights that allow unraveling molecular mechanisms that can be targeted to promote the development of competent anti-RVI immunity in early life and prevent severe RVIs. Clinical trial registration https://drks.de/search/de/trial/, identifier DRKS00034278.
Collapse
Affiliation(s)
- Carina R. Hartmann
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Robin Khan
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Jennifer Schöning
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Maximilian Richter
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Maike Willers
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
| | - Sabine Pirr
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Julia Heckmann
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Johannes Dirks
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Henner Morbach
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- Center for Primary Immunodeficiencies and Autoinflammatory Diseases (CIDA), University Hospital Würzburg, Würzburg, Germany
| | - Monika Konrad
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Elena Fries
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Magdalene Winkler
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Johanna Büchel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | | | - Jonas Fischer
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Claudia Vollmuth
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Martin Meinhardt
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Mirco Schmolke
- Department of Microbiology and Molecular Medicine, University of Geneva, Geneva, Switzerland
- Center for Inflammation Research, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Sibylle Haid
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research (TWINCORE), a joint venture between the Helmholtz Centre for Infection Research and The Hannover Medical School, Hannover, Germany
| | - Thomas Pietschmann
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Institute for Experimental Virology, Centre for Experimental and Clinical Infection Research (TWINCORE), a joint venture between the Helmholtz Centre for Infection Research and The Hannover Medical School, Hannover, Germany
- German Centre for Infection Research, Partner Site Braunschweig-Hannover, Braunschweig, Germany
| | - Simone Backes
- Institute for Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | - Lars Dölken
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Institute of Virology, Hannover Medical School, Hannover, Germany
| | - Ulrike Löber
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation of Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- German Centre for Cardiovascular Research (DZHK), Partner site Berlin, Berlin, Germany
| | - Thomas Keil
- Institute of Social Medicine, Epidemiology and Health Economics, Charité - Universitätsmedizin Berlin, Berlin, Germany
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- State Institute of Health I, Bavarian Health and Food Safety Authority, Erlangen, Germany
| | - Peter U. Heuschmann
- Institute of Clinical Epidemiology and Biometry, University of Würzburg, Würzburg, Germany
- Institute for Medical Data Science, University Hospital Würzburg, Würzburg, Germany
- Clinical Trial Centre Würzburg, University Hospital Würzburg, Würzburg, Germany
| | - Achim Wöckel
- Department of Gynecology and Obstetrics, University Hospital Würzburg, Würzburg, Germany
| | - Sagar
- Department of Medicine II (Gastroenterology, Hepatology, Endocrinology, and Infectious Diseases), Freiburg University Medical Center, University of Freiburg, Freiburg, Germany
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE) and University of Bonn, Bonn, Germany
- Genomics and Immunoregulation, Life and Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Sofia K. Forslund-Startceva
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Experimental and Clinical Research Center, a cooperation of Charité - Universitätsmedizin Berlin and Max-Delbrück-Center for Molecular Medicine, Berlin, Germany
- Charité - Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Dorothee Viemann
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- Department of Pediatric Pneumology, Allergology and Neonatology, Hannover Medical School, Hannover, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
- Center for Infection Research, University Würzburg, Würzburg, Germany
| |
Collapse
|
32
|
Zanoni I. Kids' noses resist COVID-19. J Exp Med 2024; 221:e20241027. [PMID: 38949639 PMCID: PMC11215540 DOI: 10.1084/jem.20241027] [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] [Indexed: 07/02/2024] Open
Abstract
Children resist COVID-19, and previous studies reported increased innate immunity in their upper airways. A new paper by Watkins et al. (https://doi.org/10.1084/jem.20230911) shows that the nasal mucosa of children is characterized by often asymptomatic viral and/or bacterial infections that dynamically regulate distinct innate immune programs.
Collapse
Affiliation(s)
- Ivan Zanoni
- Division of Immunology and Division of Gastroenterology, Harvard Medical School and Boston Children’s Hospital, Boston, MA, USA
| |
Collapse
|
33
|
Watkins TA, Green AB, Amat JA, Cheemarla NR, Hänsel K, Lozano R, Dudgeon SN, Germain G, Landry ML, Schulz WL, Foxman EF. High burden of viruses and bacterial pathobionts drives heightened nasal innate immunity in children. J Exp Med 2024; 221:e20230911. [PMID: 38949638 PMCID: PMC11215523 DOI: 10.1084/jem.20230911] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 04/17/2024] [Accepted: 06/12/2024] [Indexed: 07/02/2024] Open
Abstract
Studies during the COVID-19 pandemic showed that children had heightened nasal innate immune responses compared with adults. To evaluate the role of nasal viruses and bacteria in driving these responses, we performed cytokine profiling and comprehensive, symptom-agnostic testing for respiratory viruses and bacterial pathobionts in nasopharyngeal samples from children tested for SARS-CoV-2 in 2021-22 (n = 467). Respiratory viruses and/or pathobionts were highly prevalent (82% of symptomatic and 30% asymptomatic children; 90 and 49% for children <5 years). Virus detection and load correlated with the nasal interferon response biomarker CXCL10, and the previously reported discrepancy between SARS-CoV-2 viral load and nasal interferon response was explained by viral coinfections. Bacterial pathobionts correlated with a distinct proinflammatory response with elevated IL-1β and TNF but not CXCL10. Furthermore, paired samples from healthy 1-year-olds collected 1-2 wk apart revealed frequent respiratory virus acquisition or clearance, with mucosal immunophenotype changing in parallel. These findings reveal that frequent, dynamic host-pathogen interactions drive nasal innate immune activation in children.
Collapse
Affiliation(s)
- Timothy A. Watkins
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Alex B. Green
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Julien A.R. Amat
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Nagarjuna R. Cheemarla
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| | - Katrin Hänsel
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Richard Lozano
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Sarah N. Dudgeon
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Gregory Germain
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Marie L. Landry
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Wade L. Schulz
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
| | - Ellen F. Foxman
- Department of Laboratory Medicine, Yale School of Medicine, New Haven, CT, USA
- Department of Immunobiology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
34
|
Li R, Sone N, Gotoh S, Sun X, Hagood JS. Contemporary and emerging technologies for research in children's rare and interstitial lung disease. Pediatr Pulmonol 2024; 59:2349-2359. [PMID: 37204232 DOI: 10.1002/ppul.26490] [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: 01/13/2023] [Revised: 05/04/2023] [Accepted: 05/09/2023] [Indexed: 05/20/2023]
Abstract
Although recent decades have seen the identification, classification and discovery of the genetic basis of many children's interstitial and rare lung disease (chILD) disorders, detailed understanding of pathogenesis and specific therapies are still lacking for most of them. Fortunately, a revolution of technological advancements has created new opportunities to address these critical knowledge gaps. High-throughput sequencing has facilitated analysis of transcription of thousands of genes in thousands of single cells, creating tremendous breakthroughs in understanding normal and diseased cellular biology. Spatial techniques allow analysis of transcriptomes and proteomes at the subcellular level in the context of tissue architecture, in many cases even in formalin-fixed, paraffin-embedded specimens. Gene editing techniques allow creation of "humanized" animal models in a shorter time frame, for improved knowledge and preclinical therapeutic testing. Regenerative medicine approaches and bioengineering advancements facilitate the creation of patient-derived induced pluripotent stem cells and their differentiation into tissue-specific cell types which can be studied in multicellular "organoids" or "organ-on-a-chip" approaches. These technologies, singly and in combination, are already being applied to gain new biological insights into chILD disorders. The time is ripe to systematically apply these technologies to chILD, together with sophisticated data science approaches, to improve both biological understanding and disease-specific therapy.
Collapse
Affiliation(s)
- Rongbo Li
- Department of Pediatrics, Division of Respiratory Medicine, UC-San Diego, La Jolla, California, USA
| | - Naoyuki Sone
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Shimpei Gotoh
- Center for iPS Cell Research and Application, Kyoto University, Kyoto, Japan
| | - Xin Sun
- Department of Pediatrics, Division of Respiratory Medicine, UC-San Diego, La Jolla, California, USA
| | - James S Hagood
- Department of Pediatrics, Pulmonology Division, Program for Rare and Interstitial Lung Disease, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
35
|
Carter MJ, Carrol ED, Ranjit S, Mozun R, Kissoon N, Watson RS, Schlapbach LJ. Susceptibility to childhood sepsis, contemporary management, and future directions. THE LANCET. CHILD & ADOLESCENT HEALTH 2024; 8:682-694. [PMID: 39142742 DOI: 10.1016/s2352-4642(24)00141-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 05/27/2024] [Accepted: 06/04/2024] [Indexed: 08/16/2024]
Abstract
Sepsis disproportionally affects children across all health-care settings and is one of the leading causes of morbidity and mortality in neonatal and paediatric age groups. As shown in the first paper in this Series, the age-specific incidence of sepsis is highest during the first years of life, before approaching adult incidence rates during adolescence. In the second paper in this Series, we focus on the unique susceptibility of paediatric patients to sepsis and how the underlying dysregulated host response relates to developmental aspects of children's immune system, genetic, perinatal, and environmental factors, and comorbidities and socioeconomic determinants of health, which often differ between children and adults. State-of-the-art clinical management of paediatric sepsis is organised around three treatment pillars-diagnosis, early resuscitation, and titration of advanced care-and we examine available treatment guidelines and the limitations of their supporting evidence. Serious evidence gaps remain in key areas of paediatric sepsis care, especially surrounding recognition, common interventions, and survivor support, and to this end we offer a research roadmap for the next decade that could accelerate targeted diagnostics and personalised use of immunomodulation. However, improving outcomes for children with sepsis relies fundamentally on systematic quality improvement in both recognition and treatment, which is the theme of the third paper in this Series. Digital health, as shown in the fourth and final paper of this Series, holds promising potential in breaking down the barriers that hinder progress in paediatric sepsis care and, ultimately, global child health.
Collapse
Affiliation(s)
- Michael J Carter
- Centre for Human Genetics, University of Oxford, Oxford, UK; Paediatric Intensive Care unit, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | - Enitan D Carrol
- Department of Clinical Infection, Microbiology and Immunology, University of Liverpool Institute of Infection, Veterinary and Ecological Sciences, Liverpool, UK
| | | | - Rebeca Mozun
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Niranjan Kissoon
- Global Child Health Department of Pediatrics and Emergency Medicine, British Columbia Women and Children's Hospital and the University of British Columbia, Vancouver, BC, Canada
| | - R Scott Watson
- Seattle Children's Hospital, University of Washington School of Medicine, Seattle, WA, USA
| | - Luregn J Schlapbach
- Department of Intensive Care and Neonatology, and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland; Child Health Research Centre, The University of Queensland, Brisbane, QLD, Australia.
| |
Collapse
|
36
|
Okuducu YK, Mall MA, Yonker LM. COVID-19 in Pediatric Populations. Clin Chest Med 2024; 45:675-684. [PMID: 39069330 DOI: 10.1016/j.ccm.2024.02.019] [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] [Indexed: 07/30/2024]
Abstract
The COVID-19 pandemic reshaped the landscape of respiratory viral illnesses, causing common viruses to fade as SARS-CoV-2 took precedence. By 2023, more than 96% of the children in the United States were estimated to have been infected with SARS-CoV-2, with certain genetic predispositions and underlying health conditions posing risk factors for severe disease in children. Children, in general though, exhibit immunity advantages, protecting against aspects of the SARS-CoV-2 infection known to drive increased severity in older adults. Post-COVID-19 complications such as multisystem inflammatory syndrome in children and long COVID have emerged, underscoring the importance of vaccination. Here, we highlight the risks of severe pediatric COVID-19, age-specific immunoprotection, comparisons of SARS-CoV-2 with other respiratory viruses, and factors contributing to post-COVID-19 complications in children.
Collapse
Affiliation(s)
- Yanki K Okuducu
- Department of Pediatrics, Pulmonary Division, Massachusetts General Hospital, 175 Cambridge Street, 5(th) floor, Boston, MA 02114, USA; Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA
| | - Marcus A Mall
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité - Universitätsmedizin Berlin Augustenburger Platz 1, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin 13353, Germany; German Center for Lung Research (DZL), Berlin, Germany; Berlin Institute of Health at Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Lael M Yonker
- Department of Pediatrics, Pulmonary Division, Massachusetts General Hospital, 175 Cambridge Street, 5(th) floor, Boston, MA 02114, USA; Mucosal Immunology and Biology Research Center, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
37
|
Kuthning D, Raafat D, Holtfreter S, Gramenz J, Wittmann N, Bröker BM, Meyer-Bahlburg A. Variant-specific antibody profiling for tracking SARS-CoV-2 variant infections in children and adolescents. Front Immunol 2024; 15:1434291. [PMID: 39257574 PMCID: PMC11384586 DOI: 10.3389/fimmu.2024.1434291] [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: 05/17/2024] [Accepted: 08/05/2024] [Indexed: 09/12/2024] Open
Abstract
Monitoring the seroprevalence of SARS-CoV-2 in children and adolescents can provide valuable information for effective SARS-CoV-2 surveillance, and thus guide vaccination strategies. In this study, we quantified antibodies against the spike S1 domains of several SARS-CoV-2 variants (wild-type, Alpha, Delta, and Omicron variants) as well as endemic human coronaviruses (HCoVs) in 1,309 children and adolescents screened between December 2020 and March 2023. Their antibody binding profiles were compared with those of 22 pre-pandemic samples from children and adolescents using an in-house Luminex®-based Corona Array (CA). The primary objectives of this study were to (i) monitor SARS-CoV-2-specific antibodies in children and adolescents, (ii) evaluate whether the S1-specific antibody response can identify the infecting variant of concern (VoC), (iii) estimate the prevalence of silent infections, and (iv) test whether vaccination or infection with SARS-CoV-2 induce HCoV cross-reactive antibodies. Both SARS-CoV-2 infection and vaccination induced a robust antibody response against the S1 domain of WT and VoCs in children and adolescents. Antibodies specific for the S1 domain were able to distinguish between SARS-CoV-2 VoCs in infected children. The serologically identified VoC was typically the predominant VoC at the time of infection. Furthermore, our highly sensitive CA identified more silent SARS-CoV-2 infections than a commercial ELISA (12.1% vs. 6.3%, respectively), and provided insights into the infecting VoC. Seroconversion to endemic HCoVs occurred in early childhood, and vaccination or infection with SARS-CoV-2 did not induce HCoV S1 cross-reactive antibodies. In conclusion, the antibody response to the S1 domain of the spike protein of SARS-CoV-2 is highly specific, providing information about the infecting VoC and revealing clinically silent infections.
Collapse
Affiliation(s)
- Daniela Kuthning
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Dina Raafat
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
- Department of Microbiology and Immunology, Faculty of Pharmacy, Alexandria University, Alexandria, Egypt
| | - Silva Holtfreter
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Jana Gramenz
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Nico Wittmann
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Barbara M Bröker
- Institute of Immunology, University Medicine Greifswald, Greifswald, Germany
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology, Department of Pediatric and Adolescent Medicine, University Medicine Greifswald, Greifswald, Germany
| |
Collapse
|
38
|
Frasson I, Diamante L, Zangrossi M, Carbognin E, Pietà AD, Penna A, Rosato A, Verin R, Torrigiani F, Salata C, Dizanzo MP, Vaccaro L, Cacchiarelli D, Richter SN, Montagner M, Martello G. Identification of druggable host dependency factors shared by multiple SARS-CoV-2 variants of concern. J Mol Cell Biol 2024; 16:mjae004. [PMID: 38305139 PMCID: PMC11411213 DOI: 10.1093/jmcb/mjae004] [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: 02/03/2023] [Revised: 06/23/2023] [Accepted: 01/31/2024] [Indexed: 02/03/2024] Open
Abstract
The high mutation rate of SARS-CoV-2 leads to the emergence of multiple variants, some of which are resistant to vaccines and drugs targeting viral elements. Targeting host dependency factors, e.g. cellular proteins required for viral replication, would help prevent the development of resistance. However, it remains unclear whether different SARS-CoV-2 variants induce conserved cellular responses and exploit the same core host factors. To this end, we compared three variants of concern and found that the host transcriptional response was conserved, differing only in kinetics and magnitude. Clustered regularly interspaced short palindromic repeats screening identified host genes required for each variant during infection. Most of the genes were shared by multiple variants. We validated our hits with small molecules and repurposed the US Food and Drug Administration-approved drugs. All the drugs were highly active against all the tested variants, including new variants that emerged during the study (Delta and Omicron). Mechanistically, we identified reactive oxygen species production as a key step in early viral replication. Antioxidants such as N-acetyl cysteine (NAC) were effective against all the variants in both human lung cells and a humanized mouse model. Our study supports the use of available antioxidant drugs, such as NAC, as a general and effective anti-COVID-19 approach.
Collapse
Affiliation(s)
- Ilaria Frasson
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Linda Diamante
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Manuela Zangrossi
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Elena Carbognin
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| | - Anna Dalla Pietà
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Alessandro Penna
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
| | - Antonio Rosato
- Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua 35128, Italy
- Veneto Institute of Oncology IOV-IRCCS, Padua 35128, Italy
| | - Ranieri Verin
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Filippo Torrigiani
- Department of Comparative Biomedicine and Food Science, University of Padua, Padua 35020, Italy
| | - Cristiano Salata
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | | | - Lorenzo Vaccaro
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
| | - Davide Cacchiarelli
- Telethon Institute of Genetics and Medicine (TIGEM), Armenise/Harvard Laboratory of Integrative Genomics, Pozzuoli 80078, Italy
- Department of Translational Medicine, University of Naples Federico II, Naples 80138, Italy
- School for Advanced Studies, Genomics and Experimental Medicine Program, University of Naples Federico II, Naples 80138, Italy
| | - Sara N Richter
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
- Microbiology and Virology Unit, Padua University Hospital, Padua 35128, Italy
| | - Marco Montagner
- Department of Molecular Medicine, University of Padua, Padua 35121, Italy
| | - Graziano Martello
- Department of Biology, Armenise/Harvard Pluripotent Stem Cell Biology Laboratory, University of Padua, Padua 35131, Italy
| |
Collapse
|
39
|
Gay ACA, Banchero M, Carpaij O, Kole TM, Apperloo L, van Gosliga D, Fajar PA, Koppelman GH, Bont L, Hendriks RW, van den Berge M, Nawijn MC. Airway epithelial cell response to RSV is mostly impaired in goblet and multiciliated cells in asthma. Thorax 2024; 79:811-821. [PMID: 38373824 PMCID: PMC11347251 DOI: 10.1136/thorax-2023-220230] [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: 03/10/2023] [Accepted: 11/27/2023] [Indexed: 02/21/2024]
Abstract
BACKGROUND In patients with asthma, respiratory syncytial virus (RSV) infections can cause disease exacerbation by infecting the epithelial layer of the airways, inducing subsequent immune response. The type I interferon antiviral response of epithelial cells upon RSV infection is found to be reduced in asthma in most-but not all-studies. Moreover, the molecular mechanisms causing the differences in the asthmatic bronchial epithelium in response to viral infection are poorly understood. METHODS Here, we investigated the transcriptional response to RSV infection of primary bronchial epithelial cells (pBECs) from patients with asthma (n=8) and healthy donors (n=8). The pBECs obtained from bronchial brushes were differentiated in air-liquid interface conditions and infected with RSV. After 3 days, cells were processed for single-cell RNA sequencing. RESULTS A strong antiviral response to RSV was observed for all cell types, for all samples (p<1e-48). Most (1045) differentially regulated genes following RSV infection were found in cells transitioning to secretory cells. Goblet cells from patients with asthma showed lower expression of genes involved in the interferon response (false discovery rate <0.05), including OASL, ICAM1 and TNFAIP3. In multiciliated cells, an impairment of the signalling pathways involved in the response to RSV in asthma was observed. CONCLUSION Our results highlight that the response to RSV infection of the bronchial epithelium in asthma and healthy airways was largely similar. However, in asthma, the response of goblet and multiciliated cells is impaired, highlighting the need for studying airway epithelial cells at high resolution in the context of asthma exacerbation.
Collapse
Affiliation(s)
- Aurore C A Gay
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Martin Banchero
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Orestes Carpaij
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Tessa M Kole
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Leonie Apperloo
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Djoke van Gosliga
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Putri Ayu Fajar
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| | - Gerard H Koppelman
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Louis Bont
- Department of Pediatric Infectious Diseases and Immunology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, the Netherlands
- Division of Infectious Diseases, Department of Pediatrics, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Rudi W Hendriks
- Department of Pulmonary Medicine, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Maarten van den Berge
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
- Department of Pulmonology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Martijn C Nawijn
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
- GRIAC research institute, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
40
|
Fujimoto K, Kameda Y, Nagano Y, Deguchi S, Yamamoto T, Krol RP, Gee P, Matsumura Y, Okamoto T, Nagao M, Takayama K, Yokokawa R. SARS-CoV-2-induced disruption of a vascular bed in a microphysiological system caused by type-I interferon from bronchial organoids. LAB ON A CHIP 2024; 24:3863-3879. [PMID: 38252025 DOI: 10.1039/d3lc00768e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2024]
Abstract
Blood vessels show various COVID-19-related conditions including thrombosis and cytokine propagation. Existing in vitro blood vessel models cannot represent the consequent changes in the vascular structure or determine the initial infection site, making it difficult to evaluate how epithelial and endothelial tissues are damaged. Here, we developed a microphysiological system (MPS) that co-culture the bronchial organoids and the vascular bed to analyze infection site and interactions. In this system, virus-infected organoids caused damage in vascular structure. However, vasculature was not damaged or infected when the virus was directly introduced to vascular bed. The knockout of interferon-related genes and inhibition of the JAK/STAT pathway reduced the vascular damage, indicating the protective effect of interferon response suppression. The results demonstrate selective infection of bronchial epithelial cells and vascular damage by cytokines and also indicate the applicability of MPS to investigate how the infection influences vascular structure and functions.
Collapse
Affiliation(s)
- Kazuya Fujimoto
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Yoshikazu Kameda
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Yuta Nagano
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| | - Sayaka Deguchi
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Takuya Yamamoto
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
- Institute for the Advanced Study of Human Biology (WPI-ASHBi), Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Medical-risk Avoidance based on iPS Cells Team, RIKEN Center for Advanced Intelligence Project (AIP), Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Rafal P Krol
- Research and Development Center, CiRA Foundation, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8397, Japan
| | - Peter Gee
- MaxCyte Inc., Gaithersburg, MD 20878, USA
| | - Yasufumi Matsumura
- Department of Clinical Laboratory medicine, Kyoto University Graduate School of Medicine, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Toru Okamoto
- Department of Microbiology, School of Medicine, Juntendo University, Hongo 2-1-1, Bunkyo-ku, Tokyo, 113-8421, Japan
| | - Miki Nagao
- Department of Clinical Laboratory medicine, Kyoto University Graduate School of Medicine, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Kazuo Takayama
- Center for iPS cell Research and Application (CiRA), Kyoto University, Shogoin-Kawahara-cho 53, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Ryuji Yokokawa
- Department of Micro Engineering, Kyoto University, Kyoto daigaku-Katsura, Nishikyo-ku, Kyoto 615-8540, Japan.
| |
Collapse
|
41
|
Rothoeft T, Maier C, Talarico A, Hoffmann A, Schlegtendal A, Lange B, Petersmann A, Denz R, Timmesfeld N, Toepfner N, Vidal-Blanco E, Pfaender S, Lücke T, Brinkmann F. Natural and hybrid immunity after SARS-CoV-2 infection in children and adolescents. Infection 2024; 52:1449-1458. [PMID: 38499828 PMCID: PMC11288991 DOI: 10.1007/s15010-024-02225-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Accepted: 02/24/2024] [Indexed: 03/20/2024]
Abstract
PURPOSE In contrast to adults, immune protection against SARS-CoV-2 in children and adolescents with natural or hybrid immunity is still poorly understood. The aim of this study was to analyze different immune compartments in different age groups and whether humoral immune reactions correlate with a cellular immune response. METHODS 72 children and adolescents with a preceding SARS-CoV-2 infection were recruited. 37 were vaccinated with an RNA vaccine (BNT162b2). Humoral immunity was analyzed 3-26 months (median 10 months) after infection by measuring Spike protein (S), nucleocapsid (NCP), and neutralizing antibodies (nAB). Cellular immunity was analyzed using a SARS-CoV-2-specific interferon-γ release assay (IGRA). RESULTS All children and adolescents had S antibodies; titers were higher in those with hybrid immunity (14,900 BAU/ml vs. 2118 BAU/ml). NCP antibodies were detectable in > 90%. Neutralizing antibodies (nAB) were more frequently detected (90%) with higher titers (1914 RLU) in adolescents with hybrid immunity than in children with natural immunity (62.5%, 476 RLU). Children with natural immunity were less likely to have reactive IGRAs (43.8%) than adolescents with hybrid immunity (85%). The amount of interferon-γ released by T cells was comparable in natural and hybrid immunity. CONCLUSION Spike antibodies are the most reliable markers to monitor an immune reaction against SARS-CoV-2. High antibody titers of spike antibodies and nAB correlated with cellular immunity, a phenomenon found only in adolescents with hybrid immunity. Hybrid immunity is associated with markedly higher antibody titers and a higher probability of a cellular immune response than a natural immunity.
Collapse
Affiliation(s)
- T Rothoeft
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany.
| | - C Maier
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Talarico
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Hoffmann
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - A Schlegtendal
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - B Lange
- Department of Epidemiology, Helmholtz Centre for Infection Research, Brunswick, Germany
| | - A Petersmann
- University Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Oldenburg, Oldenburg, Germany
- University Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - R Denz
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - N Timmesfeld
- Department of Medical Informatics, Biometry and Epidemiology, Ruhr-University Bochum, Bochum, Germany
| | - N Toepfner
- Department of Pediatrics, Faculty of Medicine, University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - E Vidal-Blanco
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - S Pfaender
- Department of Molecular and Medical Virology, Ruhr University Bochum, Bochum, Germany
| | - T Lücke
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
| | - F Brinkmann
- University Hospital of Pediatrics and Adolescent Medicine, St. Josef-Hospital, Ruhr-University, Bochum, Germany
- University Children's Hospital, Lübeck, Germany
- Airway Research Center North (ARCN), German Center for Lung Research (DZL), Lübeck, Germany
| |
Collapse
|
42
|
Buytaert M, El Kaddouri R, Hoste L, Meertens B, Tavernier SJ, Claes K, Debacker V, Dehoorne J, Haerynck F. Age-Dependent Signature of Serum Inflammatory Cytokines in Healthy Children and Young Adults. J Interferon Cytokine Res 2024; 44:372-378. [PMID: 38934089 DOI: 10.1089/jir.2024.0053] [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] [Indexed: 06/28/2024] Open
Abstract
The study of sensitive and specific biomarkers, such as blood inflammatory cytokines, could provide an answer to the challenges faced in the differential diagnosis of patients with systemic inflammation. Limited data exist on the impact of age on serum levels of inflammatory cytokines. We collected serum samples of 42 healthy children and young adults (1 month to 21 years). Serum levels of interleukin 1 receptor antagonist (IL-1Ra), IL-1β, IL-6, IL-18, tumor necrosis factor-alpha (TNF-α), CXCL9, and CXCL10 were measured. Data were analyzed for three different age groups (<6, 6-17, and 18-21 years). IL-18, TNF-α, and CXCL9 values varied significantly according to age group. Median values of IL-18 and TNF-α decline with age, whereas CXCL9 and CXCL10 are lowest at 6-17 years. IL-1Ra is stable among age groups. In the majority of cases, IL-1β and IL-6 are not measurable above the lower limit of quantification. A scoping literature review revealed highly variable data on IL-1Ra, IL-18, TNF-α, and CXCL10. For CXCL9, pediatric reference data are scarce. In conclusion, we report an age-dependent signature of multiple inflammatory cytokines measured in the serum of healthy children and young adults, suggesting the need to use age-specific reference values in future pediatric studies.
Collapse
Affiliation(s)
- Maarten Buytaert
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Liver Research Centre Ghent, Ghent University, Ghent, Belgium
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | | | - Levi Hoste
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
| | - Bram Meertens
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Primary Immune Deficiency Research Laboratory, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, European Reference Network RITA, Ghent University, Ghent, Belgium
| | - Simon Jan Tavernier
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatrics, Ghent University Hospital, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Primary Immune Deficiency Research Laboratory, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, European Reference Network RITA, Ghent University, Ghent, Belgium
| | - Karlien Claes
- Department of Internal Medicine and Pediatrics, Primary Immune Deficiency Research Laboratory, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, European Reference Network RITA, Ghent University, Ghent, Belgium
| | - Veronique Debacker
- Department of Internal Medicine and Pediatrics, Primary Immune Deficiency Research Laboratory, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, European Reference Network RITA, Ghent University, Ghent, Belgium
| | - Jo Dehoorne
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Pediatric Nephrology and Rheumatology, European Reference Network RITA, European Reference Network for Rare Immunodeficiency, Autoinflammatory and Autoimmune Diseases, Ghent University Hospital, Ghent, Belgium
| | - Filomeen Haerynck
- Department of Internal Medicine and Pediatrics, Ghent University, Ghent, Belgium
- Department of Internal Medicine and Pediatrics, Primary Immune Deficiency Research Laboratory, Centre for Primary Immunodeficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, European Reference Network RITA, Ghent University, Ghent, Belgium
- Department of Pediatric Pulmonology, Infectious Diseases and Immune Deficiency, Centre for Primary Immune Deficiency Ghent, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
43
|
Kozawa K, Higashimoto Y, Kawamura Y, Miura H, Hattori F, Mihara Y, Nakai H, Nishimura N, Ozaki T, Ihira M, Yoshikawa T. Temporal patterns of asymptomatic SARS-CoV-2 infection among pediatric population in Japan: A 3-year analysis. J Med Virol 2024; 96:e29847. [PMID: 39105394 DOI: 10.1002/jmv.29847] [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: 04/02/2024] [Revised: 07/02/2024] [Accepted: 07/27/2024] [Indexed: 08/07/2024]
Abstract
To elucidate the seroprevalence and rate of asymptomatic infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) in Japanese children, serological analysis was performed using serum samples collected from March 2020 to February 2023. A total of 1493 serum samples were collected during the first study period (March 2020 to February 2021). None of the serum samples was positive for SARS-CoV-2 antibody. In the second period (March 2021 to February 2022), seven of the 1055 patients (0.7%) experienced SARS-CoV-2 infection. The third period (March 2022 to February 2023) was divided into three terms: from March to June 30, 2022; from July to October 2022; and from November 2022 to February 2023. The seroprevalence gradually increased throughout this period, with rates of 6.0%, 18.6%, and 30.4% in the three terms, respectively. Pediatric cases of asymptomatic SARS-CoV-2 infection occurred after the surge of Omicron variants. Since none of the SARS-CoV-2 antibody-positive patients had a previous history of coronavirus disease 2019, the seroprevalence rate in this study may represent the rate of asymptomatic infection.
Collapse
Affiliation(s)
- Kei Kozawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Yuki Higashimoto
- Department of Clinical Microbiology, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yoshiki Kawamura
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Hiroki Miura
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| | - Fumihiko Hattori
- Department of Pediatrics, Toyokawa City Hospital, Toyokawa, Aichi, Japan
| | - Yuka Mihara
- Department of Pediatrics, Kariya Toyota General Hospital, Kariya, Aichi, Japan
| | - Hidetaka Nakai
- Department of Pediatrics, Toyokawa City Hospital, Toyokawa, Aichi, Japan
| | - Naoko Nishimura
- Department of Pediatrics, Konan Kosei Hospital, Konan, Aichi, Japan
| | - Takao Ozaki
- Department of Pediatrics, Konan Kosei Hospital, Konan, Aichi, Japan
| | - Masaru Ihira
- Department of Clinical Science for Biological Monitoring, Fujita Health University School of Medical Sciences, Toyoake, Aichi, Japan
| | - Tetsushi Yoshikawa
- Department of Pediatrics, Fujita Health University School of Medicine, Toyoake, Aichi, Japan
| |
Collapse
|
44
|
Shepard RM, Ghebremedhin A, Pratumchai I, Robinson SR, Betts C, Hu J, Sasik R, Fisch KM, Zak J, Chen H, Paradise M, Rivera J, Amjad M, Uchiyama S, Seo H, Campos AD, Dayao DA, Tzipori S, Piedra-Mora C, Das S, Hasteh F, Russo H, Sun X, Xu L, Crotty Alexander L, Duran JM, Odish M, Pretorius V, Kirchberger NC, Chin SM, Von Schalscha T, Cheresh D, Morrey JD, Alargova R, O’Connell B, Martinot TA, Patel SP, Nizet V, Martinot AJ, Coussens LM, Teijaro JR, Varner JA. PI3Kγ inhibition circumvents inflammation and vascular leak in SARS-CoV-2 and other infections. Sci Transl Med 2024; 16:eadi6887. [PMID: 38959328 PMCID: PMC11272418 DOI: 10.1126/scitranslmed.adi6887] [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: 05/11/2023] [Accepted: 06/04/2024] [Indexed: 07/05/2024]
Abstract
Virulent infectious agents such as severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and methicillin-resistant Staphylococcus aureus (MRSA) induce tissue damage that recruits neutrophils, monocyte, and macrophages, leading to T cell exhaustion, fibrosis, vascular leak, epithelial cell depletion, and fatal organ damage. Neutrophils, monocytes, and macrophages recruited to pathogen-infected lungs, including SARS-CoV-2-infected lungs, express phosphatidylinositol 3-kinase gamma (PI3Kγ), a signaling protein that coordinates both granulocyte and monocyte trafficking to diseased tissues and immune-suppressive, profibrotic transcription in myeloid cells. PI3Kγ deletion and inhibition with the clinical PI3Kγ inhibitor eganelisib promoted survival in models of infectious diseases, including SARS-CoV-2 and MRSA, by suppressing inflammation, vascular leak, organ damage, and cytokine storm. These results demonstrate essential roles for PI3Kγ in inflammatory lung disease and support the potential use of PI3Kγ inhibitors to suppress inflammation in severe infectious diseases.
Collapse
Affiliation(s)
- Ryan M. Shepard
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | | | | | - Sally R. Robinson
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University North Grafton, MA 01536
- New England Regional Biosafety Laboratory, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536
| | - Courtney Betts
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201
| | - Jingjing Hu
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Roman Sasik
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA 92093
| | - Kathleen M. Fisch
- Center for Computational Biology & Bioinformatics, University of California, San Diego, La Jolla, CA 92093
- Department of Obstetrics, Gynecology & Reproductive Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Jaroslav Zak
- Department of Immunology, The Scripps Research Institute, La Jolla CA 92037
| | - Hui Chen
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Marc Paradise
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Jason Rivera
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Mohammad Amjad
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Satoshi Uchiyama
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Hideya Seo
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Alejandro D. Campos
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Denise Ann Dayao
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University North Grafton, MA 01536
| | - Saul Tzipori
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University North Grafton, MA 01536
| | - Cesar Piedra-Mora
- Department of Comparative Pathobiology, Section of Pathology, Tufts University Cummings School of Veterinary Medicine, North Grafton, MA, USA 01536
| | - Soumita Das
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Farnaz Hasteh
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Hana Russo
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - Xin Sun
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | - Le Xu
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
| | | | - Jason M. Duran
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Mazen Odish
- Department of Medicine, University of California, San Diego, La Jolla, CA 92093
| | - Victor Pretorius
- Department of Surgery, University of California, San Diego, La Jolla, CA 92093
| | - Nell C. Kirchberger
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201
| | - Shao-ming Chin
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
| | - Tami Von Schalscha
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - David Cheresh
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| | - John D. Morrey
- The Institute for Antiviral Research, Animal, Dairy and Veterinary Science, Utah State University, Logan, UT 84322
| | | | | | | | - Sandip P. Patel
- Department of Medicine/Medical Oncology, University of California, San Diego, La Jolla, CA 92093
| | - Victor Nizet
- Department of Pediatrics, University of California, San Diego, La Jolla, CA 92093
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, CA 92093
| | - Amanda J. Martinot
- Department of Infectious Disease and Global Health, Cummings School of Veterinary Medicine, Tufts University North Grafton, MA 01536
- New England Regional Biosafety Laboratory, Cummings School of Veterinary Medicine, Tufts University, North Grafton, MA 01536
- Department of Comparative Pathobiology, Section of Pathology, Tufts University Cummings School of Veterinary Medicine, North Grafton, MA, USA 01536
| | - Lisa M. Coussens
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR 97201
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR 97201
| | - John R. Teijaro
- Department of Immunology, The Scripps Research Institute, La Jolla CA 92037
| | - Judith A. Varner
- Moores Cancer Center, University of California, San Diego, La Jolla, CA 92093
- Department of Pathology, University of California, San Diego, La Jolla, CA 92093
| |
Collapse
|
45
|
Lindeboom RGH, Worlock KB, Dratva LM, Yoshida M, Scobie D, Wagstaffe HR, Richardson L, Wilbrey-Clark A, Barnes JL, Kretschmer L, Polanski K, Allen-Hyttinen J, Mehta P, Sumanaweera D, Boccacino JM, Sungnak W, Elmentaite R, Huang N, Mamanova L, Kapuge R, Bolt L, Prigmore E, Killingley B, Kalinova M, Mayer M, Boyers A, Mann A, Swadling L, Woodall MNJ, Ellis S, Smith CM, Teixeira VH, Janes SM, Chambers RC, Haniffa M, Catchpole A, Heyderman R, Noursadeghi M, Chain B, Mayer A, Meyer KB, Chiu C, Nikolić MZ, Teichmann SA. Human SARS-CoV-2 challenge uncovers local and systemic response dynamics. Nature 2024; 631:189-198. [PMID: 38898278 PMCID: PMC11222146 DOI: 10.1038/s41586-024-07575-x] [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: 02/24/2023] [Accepted: 05/16/2024] [Indexed: 06/21/2024]
Abstract
The COVID-19 pandemic is an ongoing global health threat, yet our understanding of the dynamics of early cellular responses to this disease remains limited1. Here in our SARS-CoV-2 human challenge study, we used single-cell multi-omics profiling of nasopharyngeal swabs and blood to temporally resolve abortive, transient and sustained infections in seronegative individuals challenged with pre-Alpha SARS-CoV-2. Our analyses revealed rapid changes in cell-type proportions and dozens of highly dynamic cellular response states in epithelial and immune cells associated with specific time points and infection status. We observed that the interferon response in blood preceded the nasopharyngeal response. Moreover, nasopharyngeal immune infiltration occurred early in samples from individuals with only transient infection and later in samples from individuals with sustained infection. High expression of HLA-DQA2 before inoculation was associated with preventing sustained infection. Ciliated cells showed multiple immune responses and were most permissive for viral replication, whereas nasopharyngeal T cells and macrophages were infected non-productively. We resolved 54 T cell states, including acutely activated T cells that clonally expanded while carrying convergent SARS-CoV-2 motifs. Our new computational pipeline Cell2TCR identifies activated antigen-responding T cells based on a gene expression signature and clusters these into clonotype groups and motifs. Overall, our detailed time series data can serve as a Rosetta stone for epithelial and immune cell responses and reveals early dynamic responses associated with protection against infection.
Collapse
Affiliation(s)
- Rik G H Lindeboom
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- The Netherlands Cancer Institute, Amsterdam, The Netherlands.
| | - Kaylee B Worlock
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Lisa M Dratva
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK
| | - Masahiro Yoshida
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - David Scobie
- Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| | - Helen R Wagstaffe
- Department of Infectious Disease, Imperial College London, London, UK
| | - Laura Richardson
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Josephine L Barnes
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | | | | | | | - Puja Mehta
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | | | | | - Waradon Sungnak
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Microbiology, Faculty of Science, and Integrative Computational BioScience Center, Mahidol University, Bangkok, Thailand
| | - Rasa Elmentaite
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Ensocell Therapeutics, BioData Innovation Centre, Wellcome Genome Campus, Hinxton, UK
| | - Ni Huang
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Lira Mamanova
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Rakesh Kapuge
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Liam Bolt
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Elena Prigmore
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Ben Killingley
- Department of Infectious Diseases, University College London Hospital, London, UK
| | | | | | | | | | - Leo Swadling
- Division of Infection and Immunity, Institute of Immunity and Transplantation, University College London, London, UK
| | | | - Samuel Ellis
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Claire M Smith
- UCL Great Ormond Street Institute of Child Health, London, UK
| | - Vitor H Teixeira
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Sam M Janes
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Rachel C Chambers
- UCL Respiratory, Division of Medicine, University College London, London, UK
| | - Muzlifah Haniffa
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | | | - Robert Heyderman
- Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| | - Mahdad Noursadeghi
- Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| | - Benny Chain
- Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| | - Andreas Mayer
- Research Department of Infection, Division of Infection and Immunity, University College London, London, UK
| | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Christopher Chiu
- Department of Infectious Disease, Imperial College London, London, UK
| | - Marko Z Nikolić
- UCL Respiratory, Division of Medicine, University College London, London, UK.
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK.
- Theory of Condensed Matter, Cavendish Laboratory, Department of Physics, University of Cambridge, Cambridge, UK.
- Wellcome MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
46
|
Chang JJY, Grimley SL, Tran BM, Deliyannis G, Tumpach C, Nguyen AN, Steinig E, Zhang J, Schröder J, Caly L, McAuley J, Wong SL, Waters SA, Stinear TP, Pitt ME, Purcell D, Vincan E, Coin LJ. Uncovering strain- and age-dependent innate immune responses to SARS-CoV-2 infection in air-liquid-interface cultured nasal epithelia. iScience 2024; 27:110009. [PMID: 38868206 PMCID: PMC11166695 DOI: 10.1016/j.isci.2024.110009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 04/03/2024] [Accepted: 05/14/2024] [Indexed: 06/14/2024] Open
Abstract
Continuous assessment of the impact of SARS-CoV-2 on the host at the cell-type level is crucial for understanding key mechanisms involved in host defense responses to viral infection. We investigated host response to ancestral-strain and Alpha-variant SARS-CoV-2 infections within air-liquid-interface human nasal epithelial cells from younger adults (26-32 Y) and older children (12-14 Y) using single-cell RNA-sequencing. Ciliated and secretory-ciliated cells formed the majority of highly infected cell-types, with the latter derived from ciliated lineages. Strong innate immune responses were observed across lowly infected and uninfected bystander cells and heightened in Alpha-infection. Alpha highly infected cells showed increased expression of protein-refolding genes compared with ancestral-strain-infected cells in children. Furthermore, oxidative phosphorylation-related genes were down-regulated in bystander cells versus infected and mock-control cells, underscoring the importance of these biological functions for viral replication. Overall, this study highlights the complexity of cell-type-, age- and viral strain-dependent host epithelial responses to SARS-CoV-2.
Collapse
Affiliation(s)
- Jessie J.-Y. Chang
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Samantha L. Grimley
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Bang M. Tran
- Department of Infectious Diseases, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Georgia Deliyannis
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Carolin Tumpach
- Department of Infectious Diseases, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - An N.T. Nguyen
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Eike Steinig
- Department of Infectious Diseases, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - JianShu Zhang
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Jan Schröder
- Computational Sciences Initiative (CSI), The Peter Doherty Institute for Infection and Immunity, The University of Melbourne, Melbourne, VIC 3000, Australia
| | - Leon Caly
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Julie McAuley
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Sharon L. Wong
- Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
| | - Shafagh A. Waters
- Molecular and Integrative Cystic Fibrosis Research Centre, University of New South Wales, Sydney, NSW 2052, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW 2052, Australia
- Department of Respiratory Medicine, Sydney Children’s Hospital, Sydney, NSW 2031, Australia
| | - Timothy P. Stinear
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Miranda E. Pitt
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Australian Institute for Microbiology and Infection, University of Technology Sydney, Sydney, NSW 2007, Australia
| | - Damian Purcell
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
| | - Elizabeth Vincan
- Department of Infectious Diseases, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia
| | - Lachlan J.M. Coin
- Department of Microbiology and Immunology, University of Melbourne at The Peter Doherty Institute for Infection and Immunity, Melbourne, VIC 3000, Australia
- Department of Clinical Pathology, University of Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
47
|
Bagala I, Namuganga JF, Nayebare P, Cuu G, Katairo T, Nabende I, Gonahasa S, Nassali M, Tukwasibwe S, Dorsey G, Nankabirwa J, Kitaka SB, Kiguli S, Greenhouse B, Ssewanyana I, Kamya MR, Briggs J. Seroprevalence of SARS-CoV-2 and risk factors for infection among children in Uganda: a serial cross-sectional study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.06.09.24308673. [PMID: 38947039 PMCID: PMC11213087 DOI: 10.1101/2024.06.09.24308673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/02/2024]
Abstract
Background Understanding COVID-19's impact on children is vital for public health policy, yet age-specific data is scarce, especially in Uganda. This study examines SARS-CoV-2 seroprevalence and risk factors among Ugandan children at two timepoints, along with COVID-19-related knowledge and practices in households, including adult vaccination status. Methods Baseline surveys were conducted in 12 communities from April to May 2021 (post-Alpha wave) and follow-up surveys in 32 communities from November 2021 to March 2022 (Omicron wave). Household questionnaires and blood samples were collected to test for malaria by microscopy and for SARS-CoV-2 using a Luminex assay. Seroprevalence was estimated at both the survey and community level. Mixed-effects logistic regression models assessed the association between individual and household factors and SARS-CoV-2 seropositivity in children, adjusting for household clustering. Results More households reported disruptions in daily life at baseline compared to follow-up, though economic impacts lingered. By the follow-up survey, 52.7% of adults had received at least one COVID-19 vaccine dose. Overall seroprevalence in children was higher at follow-up compared to baseline (71.6% versus 19.2%, p < 0.001). Seroprevalence in children ranged across communities from 6-37% at baseline and 50-90% at follow-up. At baseline, children from the poorest households were more likely to be infected. Increasing age remained the only consistent risk factor for SARS-CoV-2 seroconversion at both timepoints. Conclusions Results indicate that a larger number of children were infected by the Delta and Omicron waves of COVID-19 compared to the Alpha wave. This study is the largest seroprevalence survey in children in Uganda, providing evidence that most children were infected with SARS-CoV-2 before the vaccine was widely available to pediatric populations. Pediatric infections were vastly underreported by case counts, highlighting the importance of seroprevalence surveys in assessing disease burden when testing and reporting rates are limited and many cases are mild or asymptomatic.
Collapse
Affiliation(s)
- Irene Bagala
- Makerere University College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Gloria Cuu
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Thomas Katairo
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | - Isaiah Nabende
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Martha Nassali
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | | | - Joaniter Nankabirwa
- Makerere University College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | | - Sarah Kiguli
- Makerere University College of Health Sciences, Kampala, Uganda
| | | | - Isaac Ssewanyana
- Infectious Diseases Research Collaboration, Kampala, Uganda
- Central Public Health Laboratory, Butabika, Uganda
| | - Moses R Kamya
- Makerere University College of Health Sciences, Kampala, Uganda
- Infectious Diseases Research Collaboration, Kampala, Uganda
| | | |
Collapse
|
48
|
Ziegler CGK, Owings AH, Galeas-Pena M, Kazer SW, Miao VN, Navia AW, Tang Y, Bromley JD, Lotfy P, Sloan M, Laird H, Williams HB, George M, Drake RS, Pride Y, Abraham GE, Senitko M, Robinson TO, Diamond G, Lionakis MS, Shalek AK, Ordovas-Montanes J, Horwitz BH, Glover SC. An enhanced IL17 and muted type I interferon nasal epithelial cell state characterizes severe COVID-19 with fungal coinfection. Microbiol Spectr 2024; 12:e0351623. [PMID: 38687064 PMCID: PMC11237666 DOI: 10.1128/spectrum.03516-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Accepted: 04/15/2024] [Indexed: 05/02/2024] Open
Abstract
Recent case reports and epidemiological data suggest that fungal infections represent an underappreciated complication among people with severe COVID-19. However, the frequency of fungal colonization in patients with COVID-19 and associations with specific immune responses in the airways remain incompletely defined. We previously generated a single-cell RNA-sequencing data set characterizing the upper respiratory microenvironment during COVID-19 and mapped the relationship between disease severity and the local behavior of nasal epithelial cells and infiltrating immune cells. Our previous study, in agreement with findings from related human cohorts, demonstrated that a profound deficiency in host immunity, particularly in type I and type III interferon signaling in the upper respiratory tract, is associated with rapid progression to severe disease and worse clinical outcomes. We have now performed further analysis of this cohort and identified a subset of participants with severe COVID-19 and concurrent detection of Candida species-derived transcripts within samples collected from the nasopharynx and trachea. Here, we present the clinical characteristics of these individuals. Using matched single-cell transcriptomic profiles of these individuals' respiratory mucosa, we identify epithelial immune signatures suggestive of IL17 stimulation and anti-fungal immunity. Further, we observe a significant expression of anti-fungal inflammatory cascades in the nasal and tracheal epithelium of all participants who went on to develop severe COVID-19, even among participants without detectable genetic material from fungal pathogens. Together, our data suggest that IL17 stimulation-in part driven by Candida colonization-and blunted interferon signaling represent a common feature of severe COVID-19 infection. IMPORTANCE In this paper, we present an analysis suggesting that symptomatic and asymptomatic fungal coinfections can impact patient disease progression during COVID-19 hospitalization. By looking into the presence of other pathogens and their effect on the host immune response during COVID-19 hospitalizations, we aim to offer insight into an underestimated scenario, furthering our current knowledge of determinants of severity that could be considered for future diagnostic and intervention strategies.
Collapse
Affiliation(s)
- Carly G. K. Ziegler
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Anna H. Owings
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michelle Galeas-Pena
- Department of Medicine, Section of Gastroenterology and Hepatology, Tulane University School of Medicine, New Orleans, Los Angeles, USA
| | - Samuel W. Kazer
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, USA
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Vincent N. Miao
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Andrew W. Navia
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Ying Tang
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Joshua D. Bromley
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Department of Microbiology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Peter Lotfy
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Meredith Sloan
- Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Hannah Laird
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Haley B. Williams
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Micayla George
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Riley S. Drake
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Yilianys Pride
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - George E. Abraham
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Michal Senitko
- Division of Pulmonary, Critical Care, and Sleep Medicine, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Tanya O. Robinson
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - Gill Diamond
- Department of Oral Immunology and Infectious Diseases, University of Louisville, Louisville, Kentucky, USA
| | - Michail S. Lionakis
- Fungal Pathogenesis Section, Laboratory of Clinical Immunology and Microbiology (LCIM), National Institute of Allergy and Infectious Diseases (NIAID), Bethesda, Maryland, USA
| | - Alex K. Shalek
- Program in Health Sciences & Technology, Harvard Medical School & MIT, Boston, Massachusetts, USA
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Harvard Graduate Program in Biophysics, Harvard University, Cambridge, Massachusetts, USA
- Institute for Medical Engineering & Science, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, USA
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Jose Ordovas-Montanes
- Ragon Institute of MGH, MIT, and Harvard, Cambridge, Massachusetts, USA
- Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
- Harvard Stem Cell Institute, Cambridge, Massachusetts, USA
| | - Bruce H. Horwitz
- Program in Immunology, Harvard Medical School, Boston, Massachusetts, USA
- Division of Emergency Medicine, Boston Children’s Hospital, Boston, Massachusetts, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Boston Children’s Hospital, Boston, Massachusetts, USA
| | - Sarah C. Glover
- Division of Digestive Diseases, University of Mississippi Medical Center, Jackson, Mississippi, USA
- Center for Immunology and Microbial Research, Department of Cell & Molecular Biology, University of Mississippi Medical Center, Jackson, Mississippi, USA
| |
Collapse
|
49
|
Stevens J, Tezel O, Bonnefil V, Hapstack M, Atreya MR. Biological basis of critical illness subclasses: from the bedside to the bench and back again. Crit Care 2024; 28:186. [PMID: 38812006 PMCID: PMC11137966 DOI: 10.1186/s13054-024-04959-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Critical illness syndromes including sepsis, acute respiratory distress syndrome, and acute kidney injury (AKI) are associated with high in-hospital mortality and long-term adverse health outcomes among survivors. Despite advancements in care, clinical and biological heterogeneity among patients continues to hamper identification of efficacious therapies. Precision medicine offers hope by identifying patient subclasses based on clinical, laboratory, biomarker and 'omic' data and potentially facilitating better alignment of interventions. Within the previous two decades, numerous studies have made strides in identifying gene-expression based endotypes and clinico-biomarker based phenotypes among critically ill patients associated with differential outcomes and responses to treatment. In this state-of-the-art review, we summarize the biological similarities and differences across the various subclassification schemes among critically ill patients. In addition, we highlight current translational gaps, the need for advanced scientific tools, human-relevant disease models, to gain a comprehensive understanding of the molecular mechanisms underlying critical illness subclasses.
Collapse
Affiliation(s)
- Joseph Stevens
- Division of Immunobiology, Graduate Program, College of Medicine, University of Cincinnati, Cincinnati, OH, 45267, USA
| | - Oğuzhan Tezel
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Valentina Bonnefil
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA
| | - Matthew Hapstack
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Mihir R Atreya
- Division of Critical Care Medicine, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA.
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, 45627, USA.
| |
Collapse
|
50
|
Zhang Z, Kean IRL, Dratva LM, Clark JA, Syrimi E, Khan N, Daubney E, White D, O'Neill L, Chisholm C, Payne C, Benkenstein S, Kupiec K, Galassini R, Wright V, Winmill H, Robbins C, Brown K, Ramnarayan P, Scholefield B, Peters M, Klein N, Montgomery H, Meyer KB, Teichmann SA, Bryant C, Taylor G, Pathan N. Enhanced CD95 and interleukin 18 signalling accompany T cell receptor Vβ21.3+ activation in multi-inflammatory syndrome in children. Nat Commun 2024; 15:4227. [PMID: 38762592 PMCID: PMC11102542 DOI: 10.1038/s41467-024-48699-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: 10/31/2022] [Accepted: 05/10/2024] [Indexed: 05/20/2024] Open
Abstract
Multisystem inflammatory syndrome in children is a post-infectious presentation SARS-CoV-2 associated with expansion of the T cell receptor Vβ21.3+ T-cell subgroup. Here we apply muti-single cell omics to compare the inflammatory process in children with acute respiratory COVID-19 and those presenting with non SARS-CoV-2 infections in children. Here we show that in Multi-Inflammatory Syndrome in Children (MIS-C), the natural killer cell and monocyte population demonstrate heightened CD95 (Fas) and Interleuking 18 receptor expression. Additionally, TCR Vβ21.3+ CD4+ T-cells exhibit skewed differentiation towards T helper 1, 17 and regulatory T cells, with increased expression of the co-stimulation receptors ICOS, CD28 and interleukin 18 receptor. We observe no functional evidence for NLRP3 inflammasome pathway overactivation, though MIS-C monocytes show elevated active caspase 8. This, coupled with raised IL18 mRNA expression in CD16- NK cells on single cell RNA sequencing analysis, suggests interleukin 18 and CD95 signalling may trigger activation of TCR Vβ21.3+ T-cells in MIS-C, driven by increased IL-18 production from activated monocytes and CD16- Natural Killer cells.
Collapse
MESH Headings
- Humans
- Interleukin-18/metabolism
- Child
- Signal Transduction
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- fas Receptor/metabolism
- fas Receptor/genetics
- Monocytes/immunology
- Monocytes/metabolism
- Systemic Inflammatory Response Syndrome/immunology
- Systemic Inflammatory Response Syndrome/metabolism
- COVID-19/immunology
- COVID-19/virology
- COVID-19/metabolism
- COVID-19/complications
- Inflammasomes/metabolism
- Inflammasomes/immunology
- SARS-CoV-2/immunology
- Adolescent
- Male
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Female
- Child, Preschool
- Single-Cell Analysis
- NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
- NLR Family, Pyrin Domain-Containing 3 Protein/genetics
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD28 Antigens/metabolism
- Lymphocyte Activation/immunology
- Receptors, Interleukin-18/metabolism
- Receptors, Interleukin-18/genetics
- Receptors, Interleukin-18/immunology
Collapse
Affiliation(s)
- Zhenguang Zhang
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Iain R L Kean
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Lisa M Dratva
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - John A Clark
- Departments of Paediatrics, University of Cambridge, Cambridge, UK
| | - Eleni Syrimi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Naeem Khan
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Esther Daubney
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK
| | - Deborah White
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK
| | - Lauran O'Neill
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Catherine Chisholm
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Caroline Payne
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Sarah Benkenstein
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | - Klaudia Kupiec
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | | | - Victoria Wright
- Department of Paediatrics, Imperial College London, London, UK
| | - Helen Winmill
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
| | - Ceri Robbins
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
| | - Katherine Brown
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
| | | | - Barnaby Scholefield
- Paediatric Intensive Care Unit, Birmingham Children's Hospital, Birmingham, UK
- Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Mark Peters
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
- Departments of Paediatrics, University College London, London, UK
| | - Nigel Klein
- Paediatric Intensive Care Unit, Great Ormond Street Hospital, London, UK
- Departments of Paediatrics, University College London, London, UK
| | | | - Kerstin B Meyer
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
| | - Sarah A Teichmann
- Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, UK
- Department of Theory of Condensed Matter, Cavendish Laboratory, Department of Physics University of Cambridge, Cambridge, UK
| | - Clare Bryant
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Graham Taylor
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| | - Nazima Pathan
- Departments of Paediatrics, University of Cambridge, Cambridge, UK.
- Paediatric Intensive Care Unit, Addenbrookes Hospital, Cambridge, UK.
| |
Collapse
|