1
|
Wang C, Chen B, Ying Z, Shen M, Luo Y, Lin T, Feng D, Yang D, Zhang Z, Wu J. Impact of complement C3 levels on the development of healthcare-associated infections in intensive care patients: a retrospective case-control study. Ann Med 2025; 57:2487631. [PMID: 40193179 PMCID: PMC11980203 DOI: 10.1080/07853890.2025.2487631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 03/21/2025] [Accepted: 03/24/2025] [Indexed: 04/11/2025] Open
Abstract
PURPOSE The immune system serves as a critical line of defence against pathogenic microorganisms. To investigate the impact of immune markers, measured within the first 48 h of intensive care unit (ICU) admission, on the incidence of healthcare-associated infections (HAIs) in ICU patients. METHODS This case-control study included 364 patients admitted from 1 January 2020 to 30 November 2023, receiving immune marker testing within 48 h of ICU admission. Cox proportional hazard models and propensity score matching evaluated immune markers' association with HAIs risk. Log-rank tests compared time-to-event by C3 levels. All data processing and analysis were performed using R version 4.2.0 (R Foundation for Statistical Computing, Vienna, Austria) and Python version 3.11 (Python Software Foundation, Wilmington, DE). RESULTS In total, 258 patients without HAIs (mean [SD] age, 67.24 [17.79] years) and 106 patients with HAIs (mean [SD] age, 73.80 [14.93] years) were included in the final analysis. The HAIs group had older age, longer hospital stay, lower Sequential Organ Failure Assessment (SOFA) scores, and a higher rate of comorbid infections than the non-HAIs group. Also, the HAIs group had a higher proportion of basophils, lymphocytes, monocytes and T suppressor cells (CD3 + CD8+), while the proportion of neutrophils and B cells (CD19+) was lower. After Cox regression analysis and propensity score adjustment, we found that C3 complement levels (HR: 0.40; 95%CI, 0.16-0.98; p = .044) influenced the incidence of HAIs. Patients were then divided into high C3 and low C3 groups based on a cut-off value of 0.455 for C3. A time-to-event plot showed that the median time to HAIs occurrence was nine days in the high C3 group and six days in the low C3 group (p = .048). CONCLUSIONS Elevated complement C3 levels may associat with a reduced incidence of HAIs in ICU patients.
Collapse
Affiliation(s)
- Chenjuan Wang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Binhao Chen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Zhekai Ying
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Mengyuan Shen
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Yiling Luo
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Tianchen Lin
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Dandan Feng
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Dongdong Yang
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiannong Wu
- The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| |
Collapse
|
2
|
Sato Y. Immune Aging and Its Implication for Age-Related Disease Progression. Physiology (Bethesda) 2025; 40:0. [PMID: 39887318 DOI: 10.1152/physiol.00051.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 12/18/2024] [Accepted: 01/25/2025] [Indexed: 02/01/2025] Open
Abstract
As life expectancy increases globally, the prevalence and severity of age-related diseases have risen, significantly impacting patients' quality of life and increasing dependency on the healthcare system. Age-related diseases share several pathological commonalities, and emerging evidence suggests that targeting these biological processes ameliorates multiple age-related diseases. Immune aging plays a critical role in the pathogenesis of age-related diseases, given its involvement not only in controlling infection and cancer but also in facilitating tissue homeostasis and repair. Aging causes compositional and functional changes in both innate and adaptive immune cells, thereby significantly contributing to the pathogenesis of age-related disease and systemic low-grade inflammation, termed "inflammaging." This review article aims to describe the current understanding of immune aging and its impact on age-related diseases with particular emphasis on kidney and autoimmune diseases. In addition, this review highlights tertiary lymphoid structures (TLS) as a hallmark of immune aging, exploring their roles in inflammation, tissue damage, and potential therapeutic targeting.
Collapse
Affiliation(s)
- Yuki Sato
- Department of Medicine, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Immunology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota, United States
- Department of Nephrology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| |
Collapse
|
3
|
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
|
4
|
Ashraf U, Lee A, Gao Q, Gonzalez JC, Scallan C, Chinthrajah RS, Maecker HT, Davis MM, Nadeau KC, Chakraborty S, Wang TT. Impact of age and prior COVID-19 on the response to influenza a components in the 2020-2021 Fluzone vaccine. Vaccine 2025; 56:127171. [PMID: 40328185 DOI: 10.1016/j.vaccine.2025.127171] [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: 12/31/2024] [Revised: 04/07/2025] [Accepted: 04/19/2025] [Indexed: 05/08/2025]
Abstract
Understanding how age and prior COVID-19 infection influenced influenza vaccine responses during the early SARS-CoV-2 pandemic is important for identifying factors that affect vaccine efficacy and for optimizing immunization strategies in diverse populations amid co-circulating respiratory viruses. In this study, participants were enrolled during the 2020-2021 season to receive the Fluzone vaccine, and their humoral responses to the influenza A components were analyzed in relation to age and COVID-19 history. Anti-H1 hemagglutinin (HA) responses were assessed at baseline and multiple time points post-vaccination using neutralizing antibody assays against a contemporary H1-expressing pseudovirus, measurements of H1 HA-specific memory B cells, and profiling of anti-H1 IgG glycosylation. Anti-H3 antibody responses were evaluated using a hemagglutination inhibition (HI) assay. While prior COVID-19 infection was not associated with notable differences in the humoral response in this cohort, older age consistently correlated with reduced responses across multiple readouts. These findings highlight the need for targeted approaches to improve influenza vaccine effectiveness in older adults, who remain at elevated risk for severe outcomes from both influenza virus and SARS-CoV-2 infections.
Collapse
Affiliation(s)
- Usama Ashraf
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Alexandra Lee
- Department of Pathology, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - Qinshan Gao
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - Joseph C Gonzalez
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Program in Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Courtney Scallan
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA
| | - R Sharon Chinthrajah
- Department of Pathology, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - Holden T Maecker
- Institute for Immunity, Transplantation and Infection, Stanford University, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Mark M Davis
- Institute for Immunity, Transplantation and Infection, Stanford University, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA, USA
| | - Kari C Nadeau
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA; Department of Pathology, Sean N. Parker Center for Allergy and Asthma Research, Stanford, CA 94305, USA
| | - Saborni Chakraborty
- Institute for Immunity, Transplantation and Infection, Stanford University, Stanford, CA 94305, USA.
| | - Taia T Wang
- Institute for Immunity, Transplantation and Infection, Department of Medicine, Division of Infectious Diseases, Stanford University, Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford University, Stanford, CA, USA.
| |
Collapse
|
5
|
Feng Y, Wu Z, Hu K, Yuan S, Li J, Wang Y, Wang Z, Yang H, Luo ZH, Zhou J. Inflammatory metabolite 7α,25-OHC promotes TIMP1 expression in COVID-19 monocytes through synergy effect of SMARCC1/JUND/H3K27ac. Cell Mol Life Sci 2025; 82:208. [PMID: 40399563 PMCID: PMC12095718 DOI: 10.1007/s00018-025-05721-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 03/24/2025] [Accepted: 04/19/2025] [Indexed: 05/23/2025]
Abstract
Chromatin remodeling factors are involved in the inflammatory responses, contributing to tissue damage and multi-organ dysfunction in COVID-19 patients. However, the underlying mechanisms remain unclear. In this study, high-dimensional analyses of single-cell RNA sequencing and single-cell ATAC sequencing data revealed increased chromatin accessibility at the promoters or enhancers of the pro-inflammatory cytokine tissue inhibitor of metalloproteinase-1 (TIMP1), as well as altered gene transcription profiles in monocytes from COVID-19 patients. Motif enrichment and positive regulators analyses identified SMARCC1, the core subunit of the chromatin remodeling complex, and the transcription factor JUND as positive regulators to co-modulate TIMP1 expression. In-vitro experiments, co-immunoprecipitation and chromatin immunoprecipitation (ChIP)-qPCR, and others, demonstrated the collaboration of SMARCC1 and JUND. Increased 7α,25-dihydroxycholesterol (7α,25-OHC) enhanced SMARCC1-JUND interactions to co-regulate TIMP1 expression. Further investigation indicated that 7α,25-OHC promoted the expression of SMARCC1 and its co-localization with H3K27ac, which involved in the expression of TIMP1 and inflammatory responses. Our study highlights the critical roles of SMARCC1 and JUND in COVID-19 inflammation, and offers the potential targets for the prevention and treatment of COVID-19.
Collapse
Affiliation(s)
- Ying Feng
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zheng Wu
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Kefan Hu
- Department of Biology, Faculty of Science, Hong Kong Baptist University, Hong Kong, SAR, China
| | - Shenzhen Yuan
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Jun Li
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Yi Wang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Zhongyi Wang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China
| | - Han Yang
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Zhi-Hui Luo
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| | - Jingjiao Zhou
- Department of Biology and Genetics, The College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
6
|
Bi Z, Ren W, Zeng H, Zhou Y, Liu J, Chen Z, Zhang X, He X, Lu G, Wei Y, Wei X. LL-37 Inhibits TMPRSS2-Mediated S2' Site Cleavage and SARS-CoV-2 Infection but Not Omicron Variants. Cell Prolif 2025:e70060. [PMID: 40375579 DOI: 10.1111/cpr.70060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/10/2025] [Accepted: 04/29/2025] [Indexed: 05/18/2025] Open
Abstract
Continual evolution of SARS-CoV-2 spike drives the emergence of Omicron variants that show increased spreading and immune evasion. Understanding how the variants orientate themselves towards host immune defence is crucial for controlling future pandemics. Herein, we demonstrate that human cathelicidin LL-37, a crucial component of innate immunity, predominantly binds to the S2 subunit of SARS-CoV-2 spike protein, occupying sites where TMPRSS2 typically binds. This binding impedes TMPRSS2-mediated priming at site S2' and subsequent membrane fusion processes. The mutation N764K within S2 subunit of Omicron variants reduces affinity for LL-37 significantly, thereby diminishing binding capacity and inhibitory effects on membrane fusion. Moreover, the early humoral immune response enhanced by LL-37 is observed in mice against SARS-CoV-2 spike but not Omicron BA.4/5 spike. These findings reveal the mechanism underlying interactions amongst LL-37, TMPRSS2 and SARS-CoV-2 and VOCs, and highlight the distinct mutation for Omicron variants to evade the fusion activity inhibition by host innate immunity.
Collapse
Affiliation(s)
- Zhenfei Bi
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
- Department of Hepatobiliary Surgery, Xinqiao Hospital, Army Medical University, Chongqing, China
| | - Wenyan Ren
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Hao Zeng
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanyuan Zhou
- Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Science, Shenzhen, China
| | - Jian Liu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Zimin Chen
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xindan Zhang
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xuemei He
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Guangwen Lu
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Yuquan Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| | - Xiawei Wei
- Laboratory of Aging Research and Cancer Drug Target, State Key Laboratory of Biotherapy, National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
7
|
Edae CK, Tufa Bedada A, Teklemariam MD, Gudisa AB, Adamu A, Gebre SG. Immunological response and implications of Ad26.COV2. S (Janssen COVID-19 vaccine) vaccine in diabetic patients: a prospective cohort study in Ethiopia. Front Immunol 2025; 16:1546114. [PMID: 40406137 PMCID: PMC12095280 DOI: 10.3389/fimmu.2025.1546114] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 04/11/2025] [Indexed: 05/26/2025] Open
Abstract
Introduction Patients with Diabetes are at increased risk of severe COVID-19 and death, thus, it is imperative to provide them with vaccination. Ad26.COV2. S vaccine has proven its efficacy. However, the immunological response of the patients with diabetes in Ethiopia has not been well studied. Methods This prospective cohort study assessed immune responses after vaccination with a single dose of the Ad26.COV2.S. The subjects were enrolled diabetic patients who were 18 years old and above and attended a diabetes clinic at Adama Hospital Medical College. A sufficient blood sample was collected from each participant, following established standard protocols. We evaluated correlations among selected immunological parameters (IgG, IgM, CRP, IL6, IFN-Y) and employed statistical techniques such as chi-square tests, independent t-tests, and Generalized Estimating Equations (GEE) to analyze differences between given vaccinated and non-vaccinated cohorts. Generalized Estimating Equations (GEE) are a statistical method for modeling longitudinal or clustered data, particularly useful when dealing with non-normal data like binary or count data, by estimating parameters of a generalized linear model while accounting for potential correlations between observations. Results It was found that vaccinated subjects showed significant alterations in the immune response with IgM elevation and a temporary increase of inflammatory biomarkers CRP and IL-6. Younger age and females were associated with lower inflammatory markers, and no significant effects of lifestyle factors (alcohol, chat, smoking) on immunological outcomes were observed. This vaccine elicited significant immunological responses in diabetic patients, characterized by initial increases in inflammatory markers and subsequent stabilization, and with implications for the healthcare policies to design tailored approaches for diabetic groups.
Collapse
Affiliation(s)
- Chala Kenenisa Edae
- Department of Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Abdisa Tufa Bedada
- Department of Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Maria Degef Teklemariam
- Department of Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| | - Abera Botore Gudisa
- Public Health Emergency and Management Directorate, Oromia Health Bureau, Addis Ababa, Ethiopia
| | - Abdurahman Adamu
- Center for Advanced Research, Education and Training (CAMRET), Usmanu Danfodiyo University of Sokoto (UDUS), Sokoto, Nigeria
| | - Solomon Genet Gebre
- Department of Biochemistry, School of Medicine, College of Health Sciences, Addis Ababa University, Addis Ababa, Ethiopia
| |
Collapse
|
8
|
Celestra D, Nguyen NNL, Laberthonniere C, Pang KC, Saffery R, Davey RA, Mhlanga M, Cheung AS, Novakovic B. Epigenetic remodeling by sex hormone receptors and implications for gender affirming hormone therapy. Front Immunol 2025; 16:1501959. [PMID: 40406098 PMCID: PMC12095348 DOI: 10.3389/fimmu.2025.1501959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 04/17/2025] [Indexed: 05/26/2025] Open
Abstract
Sex differences in immune system development and response to pathogens has been well documented, with females exhibiting more favorable outcomes for certain infections but a higher incidence of autoimmune disease compared to males. At least some of these sex differences are mediated by sex hormones, which signal through sex hormone receptors to remodel the regulatory chromatin landscape of cells. Here, we summarize the current knowledge of how sex hormone receptors remodel chromatin structure and epigenetic marks in different contexts in humans. As the epigenome is fundamental to specifying cell identity and function, and reflects past exposures, epigenetic variation can influence cellular responses to future stimuli. This has implications for susceptibility to infection and complex inflammatory disease in a range of hormone therapy settings, including gender-affirming hormone therapy in transgender people. Therefore, profiling of epigenetic marks in the context of gender-affirming hormone therapy is an important unexplored field of research.
Collapse
Affiliation(s)
- Den Celestra
- Murdoch Children’s Research Institute and Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Nhi N. L. Nguyen
- Murdoch Children’s Research Institute and Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Camille Laberthonniere
- Radboud Institute for Molecular Life Sciences RIMLS, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ken C. Pang
- Murdoch Children’s Research Institute and Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
- Department of Adolescent Medicine, Royal Children’s Hospital, Parkville, VIC, Australia
| | - Richard Saffery
- Murdoch Children’s Research Institute and Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
| | - Rachel A. Davey
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, VIC, Australia
| | - Musa Mhlanga
- Radboud Institute for Molecular Life Sciences RIMLS, Radboud University Medical Center, Nijmegen, Netherlands
| | - Ada S. Cheung
- Department of Medicine Austin Health, University of Melbourne, Heidelberg, VIC, Australia
- Department of Endocrinology, Austin Health, Melbourne, VIC, Australia
| | - Boris Novakovic
- Murdoch Children’s Research Institute and Department of Pediatrics, The University of Melbourne, Parkville, VIC, Australia
| |
Collapse
|
9
|
Li J, Wang H, Xia S. Hematopoietic stem and progenitor cells fine-tuning the "sweet" of trained immunity. J Leukoc Biol 2025; 117:qiaf043. [PMID: 40233187 DOI: 10.1093/jleuko/qiaf043] [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: 01/14/2025] [Revised: 03/19/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
Recent studies have challenged the traditional view of innate immunity as nonspecific and transient by demonstrating that innate immune cells can develop immune memory in response to various activating factors, a phenomenon known as trained immunity. This process involves epigenetic modifications, such as changes in chromatin accessibility, and metabolic reprogramming, which can provide protection against unrelated pathogens but may also trigger immune-mediated damage. This review summarizes the current understanding of innate immune memory, with a particular focus on recent findings regarding the training of innate immune cells at the hematopoietic stem and progenitor cell stage. We present observations of trained immunity in innate immune cells, summarize key activating factors and underlying mechanisms, and propose potential host-directed immunotherapeutic strategies and preventive measures based on trained immunity. Our aim is to highlight the biological significance of trained immunity and its potential applications in enhancing long-term immunity, improving vaccine efficacy, and preventing immune-related diseases.
Collapse
Affiliation(s)
- Jiawei Li
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Hui Wang
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| | - Sheng Xia
- Department of Immunology, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang 212000, China
| |
Collapse
|
10
|
Cui X, Dong Y, Zhan Q, Huang Y, Zhu Q, Zhang Z, Yang G, Wang L, Shen S, Zhao J, Lin Z, Sun J, Su Z, Xiao Y, Zhang C, Liang Y, Shen L, Ji L, Zhang X, Yin J, Wang H, Chen Z, Ju Z, Jiang C, Le R, Gao S. Altered 3D genome reorganization mediates precocious myeloid differentiation of aged hematopoietic stem cells in inflammation. SCIENCE CHINA. LIFE SCIENCES 2025; 68:1209-1225. [PMID: 39754007 DOI: 10.1007/s11427-024-2754-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 10/09/2024] [Indexed: 05/23/2025]
Abstract
Inflammation is a driving force of hematopoietic stem cells (HSCs) aging, causing irreversible exhaustion of functional HSCs. However, the underlying mechanism of HSCs erosion by inflammatory insult remains poorly understood. Here, we find that transient LPS exposure primes aged HSCs to undergo accelerated differentiation at the expense of self-renewal, leading to depletion of HSCs. Meanwhile, the central regulator nuclear factor kappa B (NF-κB) mediating functional impairment by inflammation insult induces differential transcriptional response in aged HSCs compared with young HSCs, with precocious activation of myeloid lineage genes. Altered compartmentalization and chromatin loop formation are associated with aging-related differential transcriptional response in HSCs upon lipopolysaccharide (LPS) stimulation. Mechanistically, enhancer and promoter regions of myeloid lineage genes in aged HSCs are more accessible and display more rapid and prominent CTCF occupancy upon LPS stimulation. Our study provides comprehensive resources for the three-dimensional (3D) genome structure of HSCs and sheds light into the ordered genome organization and the associated transcriptome signature underlying HSCs aging.
Collapse
Affiliation(s)
- Xinyu Cui
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yu Dong
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qiang Zhan
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China
| | - Yixin Huang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Qianshu Zhu
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zihao Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Guang Yang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Liping Wang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Shijun Shen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jia Zhao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhiyi Lin
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Jiatong Sun
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhongqu Su
- Shandong Key Laboratory of Animal Bioengineering and Disease Prevention, College of Animal Science and Technology, Shandong Agricultural University, Taian, 271018, China
| | - Yihan Xiao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Chuyu Zhang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Yuwei Liang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lu Shen
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Lichen Ji
- Department of Joint Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200092, China
| | - Xuguang Zhang
- Shanghai Institute of Nutrition and Health, Chinese Academy of Sciences, Shanghai, 200031, China
| | - Jiqing Yin
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Hong Wang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China
| | - Zhiyang Chen
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Zhenyu Ju
- Key Laboratory of Regenerative Medicine of Ministry of Education, Institute of Aging and Regenerative Medicine, Department of Developmental & Regenerative Medicine, College of Life Science and Technology, Jinan University, Guangzhou, 510632, China.
| | - Cizhong Jiang
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
- Key Laboratory of Spine and Spinal Cord Injury Repair and Regeneration of Ministry of Education, Orthopaedic Department of Tongji Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Rongrong Le
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| | - Shaorong Gao
- Clinical and Translational Research Center of Shanghai First Maternity & Infant Hospital, Frontier Science Center for Stem Cells, School of Life Sciences and Technology, Tongji University, Shanghai, 200092, China.
| |
Collapse
|
11
|
Feng SN, Akbar AF, Zhou AL, Kalra A, Agbor-Enoh S, Merlo CA, Bush EL. Increasing risk of postlung transplant hospitalizations for infection: An analysis of recent trends. JHLT OPEN 2025; 8:100231. [PMID: 40144725 PMCID: PMC11935489 DOI: 10.1016/j.jhlto.2025.100231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/06/2025] [Accepted: 02/12/2025] [Indexed: 03/28/2025]
Abstract
Background Despite advancements in lung transplantation (LT), infection remains a major cause of morbidity and mortality following LT. We examined trends in hospitalizations for infection in the first year after LT. Methods We identified adult LT recipients in the United States (March 1, 2018-March 9, 2023) using the Organ Procurement and Transplantation Network database. We categorized transplants into 3 eras to account for the Composite Allocation Score allocation policy change and coronavirus disease 2019: March 2018 to March 2020, March 2020 to March 2022, and March 2022 to March 9, 2023. One-year post-LT survival was compared using Kaplan-Meier survival analysis and Cox proportional hazards regression. Hospitalizations for infection were compared using multivariable logistic regression, adjusted for era and donor and recipient characteristics. Results Of 12,388 LT recipients (median age = 62, male = 61.2%), hospitalization for infection in the first-year post transplant was 5.2% for patients transplanted from March 2018 to March 2020 (N = 5,031), 7.6% from March 2020 to March 2022 (N = 4,659), and 13.2% post-March 2022 (N = 3,640) (p < 0.001). Compared to March 2018 to March 2020, patients transplanted from March 2020 to March 2022 (adjusted aoods ratio [aOR] = 1.50, 95% confidence interval [CI] = 1.26-1.79) and post-March 2022 (aOR = 2.89, 95% CI = 2.29-3.65) were more likely to be hospitalized for an infection. After adjustment, we found no significant difference in risk of death following LT for recipients transplanted between March 2020 and March 2022 (aHR = 1.09, 95% CI = 0.96-1.23, p = 0.175) compared to March 2018 and March 2020. Post-March 2022 risk of death was elevated (aHR = 1.21, 95% CI = 1.04, 1.40, p = 0.014). Conclusions Odds of hospitalization for infection in the first year after LT performed between March 2020 and March 2022 and post-March 2022 were 1.50 and 2.89 times as high, respectively, as LT performed between March 2018 and March 2020. IRB NUMBERS IRB00352819.
Collapse
Affiliation(s)
- Shi Nan Feng
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Armaan F. Akbar
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alice L. Zhou
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Andrew Kalra
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Sean Agbor-Enoh
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Christian A. Merlo
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Errol L. Bush
- Department of Surgery, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
12
|
Bian H, Chen L, Zhang Z, Wen AD, Zheng ZH, Song LQ, Yao MY, Liu YX, Zhang XJ, Dong HL, Lian JQ, Pan L, Liu Y, Gu X, Zhao H, Wang JW, Wang QY, Zhang K, Jia JF, Xie RH, Luo X, Fu XH, Jia YY, Hou JN, Tan QY, Chen XX, Yang LQ, Lin YL, Wang XX, Zhang L, Zeng QJ, Li WJ, Wang RX, Zhang Y, Sun XX, Wang B, Yang X, Jiang JL, Li L, Wu J, Yang XM, Zhang H, Shi Y, Chen XC, Tang H, Shi HW, Liu SS, Yang Y, Yang TY, Wei D, Chen ZN, Zhu P. Meplazumab, a CD147 antibody, for severe COVID-19: a double-blind, randomized, placebo-controlled, phase 3 clinical trial. Signal Transduct Target Ther 2025; 10:119. [PMID: 40222976 PMCID: PMC11994814 DOI: 10.1038/s41392-025-02208-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 02/17/2025] [Accepted: 03/13/2025] [Indexed: 04/15/2025] Open
Abstract
Meplazumab, a humanized CD147 antibody, showed favorable safety and clinical benefits in phase 1 and phase 2/3 seamless clinical studies. Further evaluation of its therapeutic efficacy in patients with severe COVID-19 is needed. In this phase 3 add-on study, we randomized patients with severe COVID-19 in a 1:1 ratio to receive 0.2 mg/kg meplazumab or placebo via intravenous injection, and evaluated efficacy and safety within 56 days. Between February 2023 and November 2023, 108 patients with severe COVID-19 were randomized to two groups, with their baseline characteristics generally balanced. The primary endpoint, 28-day all-cause mortality was 1.96% in the meplazumab group vs 7.69% in the placebo group (P = 0.1703). Supplementary analysis using composite strategy indicated a significant reduction of 28-day all-cause mortality in meplazumab compared to placebo (3.92% vs 15.38%, P = 0.044). Meplazumab also significantly reduced the mortality in smoking subjects on day 28 (P = 0.047) compared to placebo in supplementary analysis. The secondary endpoint, 56-day all-cause mortality, was 1.96% in the meplazumab group and 11.54% in the placebo group (P = 0.048), which was 3.92% and 15.38%, respectively (P = 0.044) by supplementary analysis. Additional secondary endpoints showed potential benefits, including increased hospital discharge rates, improved clinical outcomes, and improved viral nucleotide conversion rate. Meplazumab demonstrated good safety and tolerability, with no grade ≥ 3 TEAEs observed. These promising results indicate that meplazumab reduces mortality and enhances clinical benefits in severe COVID-19 patients with a good safety profile, providing effective and specific therapeutics for severe COVID-19 (the trial was registered at ClinicalTrials.gov (NCT05679479)).
Collapse
Affiliation(s)
- Huijie Bian
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China.
| | - Liang Chen
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
- School of Medicine, Shanghai University, Shanghai, China
| | - Zheng Zhang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Ai-Dong Wen
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Zhao-Hui Zheng
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Li-Qiang Song
- Department of Pulmonary and Critical Care Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Meng-Ying Yao
- Department of Pulmonary, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ying-Xia Liu
- Department of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Xi-Jing Zhang
- Department of Critical Care Medicine, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Hong-Lin Dong
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jian-Qi Lian
- The Center for Diagnosis and Treatment of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Lei Pan
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Yu Liu
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xing Gu
- Department of Respiratory and Critical Care Medicine, Xi'an Chest Hospital, Xi'an, China
| | - Hui Zhao
- Department of Vascular Surgery, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Jing-Wen Wang
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Qing-Yi Wang
- School of Basic Medicine, The Fourth Military Medical University, Xi'an, China
| | - Kui Zhang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Jun-Feng Jia
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Rong-Hua Xie
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Xing Luo
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Xiang-Hui Fu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Yan-Yan Jia
- Department of Pharmacy, Xijing Hospital, The Fourth Military Medical University, Xi'an, China
| | - Jun-Na Hou
- Department of Pulmonary, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qiu-Yue Tan
- Department of Pulmonary, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xiao-Xia Chen
- Department of Pulmonary and Critical Care Medicine, Tangdu Hospital, The Fourth Military Medical University, Xi'an, China
| | - Liu-Qing Yang
- Department of Liver Disease, Shenzhen Third People's Hospital, Shenzhen, China
| | - Yuan-Long Lin
- Department of Infectious Diseases, Shenzhen Third People's Hospital, Shenzhen, China
| | - Xiao-Xia Wang
- Department of Rheumatology and Immunology, Second Hospital of Shanxi Medical University, Taiyuan, China
| | - Lei Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qin-Jing Zeng
- Department of Critical Care Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wen-Jie Li
- Department of Respiratory and Critical Care Medicine, Xi'an Chest Hospital, Xi'an, China
| | - Rui-Xuan Wang
- Department of Respiratory and Critical Care Medicine, Xi'an Chest Hospital, Xi'an, China
| | - Yang Zhang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Xiu-Xuan Sun
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Bin Wang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Xu Yang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Jian-Li Jiang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Ling Li
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Jiao Wu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Xiang-Min Yang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Hai Zhang
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Ying Shi
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China
| | - Xiao-Chun Chen
- Jiangsu Pacific Meinuoke Biopharmaceutical Co. Ltd, Changzhou, China
| | - Hao Tang
- Jiangsu Pacific Meinuoke Biopharmaceutical Co. Ltd, Changzhou, China
| | - Hong-Wei Shi
- Jiangsu Pacific Meinuoke Biopharmaceutical Co. Ltd, Changzhou, China
| | - Shuang-Shuang Liu
- Jiangsu Pacific Meinuoke Biopharmaceutical Co. Ltd, Changzhou, China
| | - Yong Yang
- Jiangsu Pacific Meinuoke Biopharmaceutical Co. Ltd, Changzhou, China
| | - Tian-Yi Yang
- Jiangsu Pacific Meinuoke Biopharmaceutical Co. Ltd, Changzhou, China
| | - Ding Wei
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China.
| | - Zhi-Nan Chen
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China.
| | - Ping Zhu
- Department of Cell Biology of National Translational Science Center for Molecular Medicine and Department of Clinical Immunology of Xijing Hospital, The Fourth Military Medical University, Xi'an, China.
- State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, Xi'an, China.
| |
Collapse
|
13
|
Liao W, Zai X, Zhang J, Xu J. Hematopoietic stem cell state and fate in trained immunity. Cell Commun Signal 2025; 23:182. [PMID: 40229653 PMCID: PMC11995595 DOI: 10.1186/s12964-025-02192-1] [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: 02/19/2025] [Accepted: 04/06/2025] [Indexed: 04/16/2025] Open
Abstract
Trained immunity serves as a de facto memory for innate immune responses, resulting in long-term functional reprogramming of innate immune cells. It enhances resistance to pathogens and augments immunosurveillance under physiological conditions. Given that innate immune cells typically have a short lifespan and do not divide, persistent innate immune memory may be mediated by epigenetic and metabolic changes in long-lived hematopoietic stem cells (HSCs) in the bone marrow. HSCs fine-tune their state and fate in various training conditions, thereby generating functionally adapted progeny cells that orchestrate innate immune plasticity. Notably, both beneficial and maladaptive trained immunity processes can comprehensively influence HSC state and fate, leading to divergent hematopoiesis and immune outcomes. However, the underlying mechanisms are still not fully understood. In this review, we summarize recent advances regarding HSC state and fate in the context of trained immunity. By elucidating the stem cell-intrinsic and extrinsic regulatory network, we aim to refine current models of innate immune memory and provide actionable insights for developing targeted therapies against infectious diseases and chronic inflammation. Furthermore, we propose a conceptual framework for engineering precision-trained immunity through HSC-targeted interventions.
Collapse
Affiliation(s)
- Weinian Liao
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Xiaodong Zai
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Jun Zhang
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China
| | - Junjie Xu
- Laboratory of Advanced Biotechnology, Beijing Institute of Biotechnology, Beijing, 100071, China.
| |
Collapse
|
14
|
Weinand K, Langan EM, Curtis M, Raychaudhuri S. Defining effective strategies to integrate multi-sample single-nucleus ATAC-seq datasets via a multimodal-guided approach. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.04.02.646871. [PMID: 40236024 PMCID: PMC11996549 DOI: 10.1101/2025.04.02.646871] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Background Chromatin accessibility, measured via single-nucleus Assay for Transposase-Accessible Chromatin with sequencing (snATAC-seq), can reveal the underpinnings of transcriptional regulation across heterogeneous cell states. As the number and scale of snATAC-seq datasets increases, we need robust computational pipelines to integrate samples within a dataset and datasets across studies. These integration pipelines should correct cell-state-obfuscating technical effects while conserving underlying biological cell states, as has been shown for single-cell RNA-seq (scRNA-seq) pipelines. However, scRNA-seq integration methods have performed inconsistently on snATAC-seq datasets, potentially due to sparsity and genomic feature differences. Results Using single-nucleus multimodal datasets profiling ATAC and RNA simultaneously, we can measure snATAC-seq integration method performance by comparison to independently integrated snRNA-seq gold standard embeddings and annotations. Here, we benchmark 58 pipelines, incorporating 7 integration methods plus 1 embedding correction method with 5 feature sets. Using our command-line tool, we assessed 5 multimodal datasets at 3 different resolutions using 2 novel metrics to determine the best practices for multi-sample snATAC-seq integration. ATAC features outperformed Gene Activity Score (GAS) features, and embedding correction with Harmony was generally useful. SnapATAC2, PeakVI, and ArchR's iterative Latent Semantic Indexing (LSI) performed well. Conclusions We recommend SnapATAC2 + Harmony with pre-defined ENCODE candidate cis -regulatory element (cCRE) features as a first-pass pipeline given its metric performance, generalizability of features, and method resource-efficiency. This and other high-performing pipelines will guide future comprehensive gene regulation maps.
Collapse
|
15
|
Cai M, Xu E, Xie Y, Al-Aly Z. Rates of infection with other pathogens after a positive COVID-19 test versus a negative test in US veterans (November, 2021, to December, 2023): a retrospective cohort study. THE LANCET. INFECTIOUS DISEASES 2025:S1473-3099(24)00831-4. [PMID: 40185115 DOI: 10.1016/s1473-3099(24)00831-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 12/07/2024] [Accepted: 12/12/2024] [Indexed: 04/07/2025]
Abstract
BACKGROUND SARS-CoV-2 infection leads to post-acute sequelae that can affect nearly every organ system, including the immune system. However, whether an infection with SARS-CoV-2 is associated with increased risk of future infections with other pathogens is not yet fully characterised. In this study, we aimed to test the association between a positive test for COVID-19, compared with a negative test, and rates of future infections with other pathogens. METHODS We used the US Department of Veterans Affairs health-care databases to build a spatiotemporally aligned cohort of 231 899 people with a positive COVID-19 test and 605 014 with a negative COVID-19 test (test-negative control group) between Nov 1, 2021, and Dec 31, 2023. We first did a discovery approach to map the associations between those with a positive COVID-19 test versus a negative test and laboratory-based outcomes of infectious illnesses. We then compared rates of a prespecified set of infectious disease outcomes between those with and without a positive COVID-19 test. To evaluate the specificity of the findings to COVID-19, we compared the rates of a prespecified set of infectious disease outcomes in a spatiotemporally aligned cohort of people admitted to hospital for COVID-19 (n=12 450) versus those admitted for seasonal influenza (n=3293). Outcomes were ascertained 30 days after the date of the first test until the end of follow-up (365 days after the first test plus 30 days, death, or July 18, 2024, whichever came first). An inverse probability weighting approach was used to balance demographic and health characteristics across cohorts. Log-binomial regression models were used to estimate risk ratios (RRs) and 95% CIs. FINDINGS In the 12 months of follow-up, compared with participants who had a negative test for COVID-19, people with COVID-19 who did not require admission to hospital during the acute phase of infection had increased test positivity rates for bacterial infections (in blood, urine, and respiratory cultures) and viral diseases (including Epstein-Barr virus, herpes simplex virus reactivation, and respiratory viral infections). People who were positive for COVID-19 and admitted to hospital also had increased rates of bacterial infections in blood, respiratory, and urine biospecimens, and viral infections in blood and respiratory biospecimens. Analyses of prespecified outcomes showed that, compared with the test-negative control group, participants with a positive COVID-19 test who were not admitted to hospital had significantly increased rates of outpatient diagnosis of infectious illnesses (RR 1·17 [95% CI 1·15-1·19]), including bacterial, fungal, and viral infections; outpatient respiratory infections (1·46 [1·43-1·50]); and admission to hospital for infectious illnesses (1·41 [1·37-1·45]), including for sepsis and respiratory infections; the rates of prespecified outcomes were generally higher among those who were admitted to hospital for COVID-19 during the acute phase. Compared with people admitted to hospital for seasonal influenza, those admitted for COVID-19 had higher rates of admission to hospital for infectious illnesses (1·24 [1·10-1·40]), admission to hospital for sepsis (RR 1·35 [1·11-1·63]), and in-hospital use of antimicrobials (1·23 [1·10-1·37]). INTERPRETATION Our results suggest that a positive test for COVID-19 (vs a negative test) was associated with increased rates of diagnosis of various infections in the 12 months following an acute SARS-CoV-2 infection. The putative long-term effects of COVID-19 on the immune system and the propensity for infection with other pathogens should be further evaluated in future studies. FUNDING US Department of Veterans Affairs.
Collapse
Affiliation(s)
- Miao Cai
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, MO, USA; Veterans Research and Education Foundation of Saint Louis, Saint Louis, MO, USA
| | - Evan Xu
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, MO, USA; Veterans Research and Education Foundation of Saint Louis, Saint Louis, MO, USA
| | - Yan Xie
- Division of Pharmacoepidemiology, VA Saint Louis Health Care System, Saint Louis, MO, USA; Veterans Research and Education Foundation of Saint Louis, Saint Louis, MO, USA; Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Saint Louis, MO, USA
| | - Ziyad Al-Aly
- Clinical Epidemiology Center, Research and Development Service, VA Saint Louis Health Care System, Saint Louis, MO, USA; Nephrology Section, Medicine Service, VA Saint Louis Health Care System, Saint Louis, MO, USA; Veterans Research and Education Foundation of Saint Louis, Saint Louis, MO, USA; Department of Medicine, Washington University School of Medicine, Saint Louis, MO, USA; Institute for Public Health, Washington University in Saint Louis, Saint Louis, MO, USA.
| |
Collapse
|
16
|
Simonis A, Theobald SJ, Koch AE, Mummadavarapu R, Mudler JM, Pouikli A, Göbel U, Acton R, Winter S, Albus A, Holzmann D, Albert MC, Hallek M, Walczak H, Ulas T, Koch M, Tessarz P, Hänsel-Hertsch R, Rybniker J. Persistent epigenetic memory of SARS-CoV-2 mRNA vaccination in monocyte-derived macrophages. Mol Syst Biol 2025; 21:341-360. [PMID: 40133533 PMCID: PMC11965535 DOI: 10.1038/s44320-025-00093-6] [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: 06/15/2023] [Revised: 02/10/2025] [Accepted: 02/24/2025] [Indexed: 03/27/2025] Open
Abstract
Immune memory plays a critical role in the development of durable antimicrobial immune responses. How precisely mRNA vaccines train innate immune cells to shape protective host defense mechanisms remains unknown. Here we show that SARS-CoV-2 mRNA vaccination significantly establishes histone H3 lysine 27 acetylation (H3K27ac) at promoters of human monocyte-derived macrophages, suggesting epigenetic memory. However, we found that two consecutive vaccinations were required for the persistence of H3K27ac, which matched with pro-inflammatory innate immune-associated transcriptional changes and antigen-mediated cytokine secretion. H3K27ac at promoter regions were preserved for six months and a single mRNA booster vaccine potently restored their levels and release of macrophage-derived cytokines. Interestingly, we found that H3K27ac at promoters is enriched for G-quadruplex DNA secondary structure-forming sequences in macrophage-derived nucleosome-depleted regions, linking epigenetic memory to nucleic acid structure. Collectively, these findings reveal that mRNA vaccines induce a highly dynamic and persistent training of innate immune cells enabling a sustained pro-inflammatory immune response.
Collapse
Affiliation(s)
- Alexander Simonis
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Sebastian J Theobald
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany
| | - Anna E Koch
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Ram Mummadavarapu
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Julie M Mudler
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Andromachi Pouikli
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
| | - Ulrike Göbel
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Richard Acton
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Babraham Institute, Cambridge, UK
| | - Sandra Winter
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Alexandra Albus
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Dmitriy Holzmann
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Marie-Christine Albert
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Michael Hallek
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany
| | - Henning Walczak
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Center for Cell Death, Cancer and Inflammation, UCL Cancer Institute, University College London, London, United Kingdom
| | - Thomas Ulas
- Systems Medicine, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
- PRECISE Plattform for Single Cell Genomics and Epigenomics, DZNE, University of Bonn, Bonn and West German Genome Center, Bonn, Germany
- Genomics and Immunoregulation, Life & Medical Sciences (LIMES) Institute, University of Bonn, Bonn, Germany
| | - Manuel Koch
- Institute of Biochemistry I, Faculty of Medicine, University Hospital Cologne, University of Cologne, Cologne, Germany
- Institute for Dental Research and Oral Musculoskeletal Biology, Center for Dental, Oral and Maxillofacial Medicine (central facilities), Medical Faculty and University of Cologne, Cologne, Germany
| | - Peter Tessarz
- Max Planck Research Group "Chromatin and Ageing", Max Planck Institute for Biology of Ageing, Joseph-Stelzmann-Str. 9b, Cologne, 50931, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Department of Human Biology, Radboud Institute for Molecular Life Sciences, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - Robert Hänsel-Hertsch
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany.
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany.
- Department of Translational Genomics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany.
- Institute of Human Genetics, University Hospital Cologne, Cologne, Germany.
| | - Jan Rybniker
- Department I of Internal Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50937, Germany.
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, 50931, Germany.
- German Center for Infection Research (DZIF), Partner Site Bonn-Cologne, Cologne, Germany.
| |
Collapse
|
17
|
Lei Y, Tsang JS. Systems Human Immunology and AI: Immune Setpoint and Immune Health. Annu Rev Immunol 2025; 43:693-722. [PMID: 40279304 DOI: 10.1146/annurev-immunol-090122-042631] [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: 04/27/2025]
Abstract
The immune system, critical for human health and implicated in many diseases, defends against pathogens, monitors physiological stress, and maintains tissue and organismal homeostasis. It exhibits substantial variability both within and across individuals and populations. Recent technological and conceptual progress in systems human immunology has provided predictive insights that link personal immune states to intervention responses and disease susceptibilities. Artificial intelligence (AI), particularly machine learning (ML), has emerged as a powerful tool for analyzing complex immune data sets, revealing hidden patterns across biological scales, and enabling predictive models for individualistic immune responses and potentially personalized interventions. This review highlights recent advances in deciphering human immune variation and predicting outcomes, particularly through the concepts of immune setpoint, immune health, and use of the immune system as a window for measuring health. We also provide a brief history of AI; review ML modeling approaches, including their applications in systems human immunology; and explore the potential of AI to develop predictive models and personal immune state embeddings to detect early signs of disease, forecast responses to interventions, and guide personalized health strategies.
Collapse
Affiliation(s)
- Yona Lei
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
| | - John S Tsang
- Yale Center for Systems and Engineering Immunology and Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA;
- Department of Biomedical Engineering, Yale University, New Haven, Connecticut, USA
- Chan Zuckerberg Biohub NY, New Haven, Connecticut, USA
| |
Collapse
|
18
|
Humer B, Dik WA, Versnel MA. Advocating the role of trained immunity in the pathogenesis of ME/CFS: a mini review. Front Immunol 2025; 16:1483764. [PMID: 40201181 PMCID: PMC11975576 DOI: 10.3389/fimmu.2025.1483764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 03/11/2025] [Indexed: 04/10/2025] Open
Abstract
Myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is a complex chronic disease of which the underlying (molecular) mechanisms are mostly unknown. An estimated 0.89% of the global population is affected by ME/CFS. Most patients experience a multitude of symptoms that severely affect their lives. These symptoms include post-exertional malaise, chronic fatigue, sleep disorder, impaired cognitive functions, flu-like symptoms, and chronic immune activation. Therapy focusses on symptom management, as there are no drugs available. Approximately 60% of patients develop ME/CFS following an acute infection. Such a preceding infection may induce a state of trained immunity; defined as acquired, nonspecific, immunological memory of innate immune cells. Trained immune cells undergo long term epigenetic reprogramming, which leads to changes in chromatin accessibility, metabolism, and results in a hyperresponsive phenotype. Initially, trained immunity has only been demonstrated in peripheral blood monocytes and macrophages. However, more recent findings indicate that hematopoietic stem cells in the bone marrow are required for long-term persistence of trained immunity. While trained immunity is beneficial to combat infections, a disproportionate response may cause disease. We hypothesize that pronounced hyperresponsiveness of innate immune cells to stimuli could account for the aberrant activation of various immune pathways, thereby contributing to the pathophysiology of ME/CFS. In this mini review, we elaborate on the concept of trained immunity as a factor involved in the pathogenesis of ME/CFS by presenting evidence from other post-infectious diseases with symptoms that closely resemble those of ME/CFS.
Collapse
Affiliation(s)
- Bart Humer
- Department of Immunology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Willem A. Dik
- Laboratory Medical Immunology, Department of Immunology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Marjan A. Versnel
- Department of Immunology, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
19
|
Pan S, Tang H, Yao B, Tian G, Sun B, Hu Y, Chen Y, Li J, Xu X, Zhang C, Ying S. Decoding the ontogeny of myeloid lineage diversity by cross-species and developmental analyses of hematopoietic progenitor atlases. Cell Rep 2025; 44:115406. [PMID: 40057952 DOI: 10.1016/j.celrep.2025.115406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Revised: 12/23/2024] [Accepted: 02/15/2025] [Indexed: 03/29/2025] Open
Abstract
Myeloid cells play vital roles in homeostasis and immune responses in vertebrates, but the developmental pathway underlying their lineage diversity remains elusive. Here, we construct a single-cell transcriptional map of myeloid progenitors from mouse bone marrow and conduct cross-species and developmental analyses across human, monkey, mouse, and zebrafish. We uncover a conserved specification program separating the eosinophil-basophil-mast cell (EBM) lineage and neutrophil-monocyte (NM) lineage, reclassifying myeloid cells beyond the conventional granulocytic and monocytic framework. By generating Ikzf2-EGFP reporter mice, we identify IKZF2 as a priming marker for EBM lineage specification. Ikzf2-EGFP+ and Ikzf2-EGFP- granulocyte-monocyte progenitors (GMPs) exhibit distinct potential to generate EBM and NM lineages, and Ikzf2-EGFP expression robustly distinguishes their progenies. Additionally, we demonstrate that lineage specification emerges early during myelopoiesis. These findings provide a redefined perspective on myeloid lineage ontogeny, highlighting the conservation of lineage specification and offering insights into the understanding and therapeutic development of myelopoiesis.
Collapse
Affiliation(s)
- Sheng Pan
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Haoyu Tang
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China
| | - Bingpeng Yao
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Guoxiong Tian
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Beibei Sun
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Yangmingzi Hu
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Yan Chen
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China
| | - Jiaqian Li
- Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China.
| | - Chao Zhang
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China; Department of Anatomy, Zhejiang University School of Medicine, Hangzhou 310058, China.
| | - Songmin Ying
- Department of Pharmacy, Center for Regeneration and Aging Medicine, the Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Zhejiang-Denmark Joint Laboratory of Regeneration and Aging Medicine, Yiwu 322000, China; Key Laboratory of Respiratory Disease of Zhejiang Province, Department of Respiratory and Critical Care Medicine, Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang 310009, China; Department of Pharmacology, Zhejiang University School of Medicine, Hangzhou 310058, China.
| |
Collapse
|
20
|
Khan R, Ji W, Guzman Rivera J, Madhvi A, Andrews T, Richlin B, Suarez C, Gaur S, Hasan UN, Cuddy W, Singh AR, Bukulmez H, Kaelber D, Kimura Y, Ganapathi U, Michailidis IE, Ukey R, Moroso-Fela S, Kuster JK, Casseus M, Roy J, Burns JC, Kleinman LC, Horton DB, Lakhani SA, Gennaro ML. A genetically modulated Toll-like receptor-tolerant phenotype in peripheral blood cells of children with multisystem inflammatory syndrome. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2025; 214:vkaf006. [PMID: 40101747 PMCID: PMC11952872 DOI: 10.1093/jimmun/vkaf006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 01/02/2025] [Indexed: 03/20/2025]
Abstract
Dysregulated innate immune responses contribute to multisystem inflammatory syndrome in children (MIS-C), characterized by gastrointestinal, mucocutaneous, and/or cardiovascular injury occurring weeks after severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) exposure. To investigate innate immune functions, we stimulated ex vivo peripheral blood cells from MIS-C patients with agonists of Toll-like receptors (TLR), key innate immune response initiators. We found severely dampened cytokine responses and elevated gene expression of negative regulators of TLR signaling. Increased plasma levels of zonulin, a gut leakage marker, were also detected. These effects were also observed in fully convalescent children months after MIS-C recovery. When we investigated the genetic background of patients in relation to TLR responsiveness, we found that cells from MIS-C children carrying rare heterozygous variants of lysosomal trafficking regulator (LYST) were less refractory to TLR stimulation and exhibited lysosomal and mitochondrial abnormalities with altered energy metabolism. Moreover, these rare LYST variant heterozygous carriers tended to exhibit unfavorable clinical laboratory indicators of inflammation, including more profound lymphopenia. The results of our observational study have several implications. First, TLR hyporesponsiveness may be associated with hyperinflammation and/or excessive or prolonged stimulation with gut-originated TLR ligands. Second, TLR hyporesponsiveness during MIS-C may be protective, since LYST variant heterozygous carriers exhibited reduced TLR hyporesponsiveness and unfavorable clinical laboratory indicators of inflammation. Thus, links may exist between genetic background, ability to establish a refractory immune state, and MIS-C clinical spectrum. Third, the possibility exists that prolonged TLR hyporesponsiveness is one of the mechanisms driving long coronavirus disease (COVID), which highlights the need to monitor long-term consequences of MIS-C.
Collapse
Affiliation(s)
- Rehan Khan
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Weizhen Ji
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Jeisac Guzman Rivera
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Abhilasha Madhvi
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Tracy Andrews
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Benjamin Richlin
- Pediatric Clinical Research Center, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Christian Suarez
- Pediatric Clinical Research Center, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Sunanda Gaur
- Department of Pediatrics, Clinical Research Center, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Uzma N Hasan
- Department of Pediatrics, Cooperman Barnabas Medical Center, Livingston, NJ, United States
| | - William Cuddy
- Maria Fareri Children’s Hospital, Valhalla, NY, United States
| | - Aalok R Singh
- Maria Fareri Children’s Hospital, Valhalla, NY, United States
- New York Medical College, Touro University, Valhalla, NY, United States
| | - Hulya Bukulmez
- Department of Pediatrics, Division of Rheumatology, MetroHealth System, Cleveland, OH, United States
| | - David Kaelber
- Department of Pediatrics, Division of Rheumatology, MetroHealth System, Cleveland, OH, United States
- Center for Clinical Informatics Research and Education, MetroHealth System, Cleveland, OH, United States
- Department of Internal Medicine, Pediatrics, and Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland, OH, United States
| | - Yukiko Kimura
- Hackensack University Medical Center, Hackensack Meridian School of Medicine, Nutley, NJ, United States
| | - Usha Ganapathi
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Ioannis E Michailidis
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Rahul Ukey
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| | - Sandra Moroso-Fela
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - John K Kuster
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Myriam Casseus
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
| | - Jason Roy
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Jane C Burns
- Department of Pediatrics, University of California, San Diego, CA, United States
- Rady Children’s Hospital-San Diego, University of California, San Diego School of Medicine, San Diego, CA, United States
| | - Lawrence C Kleinman
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
- Department of Global Urban Health, Rutgers School of Public Health, Piscataway, NJ, United States
| | - Daniel B Horton
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, NJ, United States
- Department of Pediatrics, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, United States
- Rutgers Center for Pharmacoepidemiology and Treatment Science, Institute for Health, Health Care Policy and Aging Research, New Brunswick, NJ, United States
| | - Saquib A Lakhani
- Pediatric Genomics Discovery Program, Department of Pediatrics, Yale University School of Medicine, New Haven, CT, United States
| | - Maria Laura Gennaro
- Public Health Research Institute, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
- Department of Medicine, Rutgers New Jersey Medical School, Rutgers Biomedical and Health Sciences, Newark, NJ, United States
| |
Collapse
|
21
|
Kato Y, Kumanogoh A. The immune memory of innate immune systems. Int Immunol 2025; 37:195-202. [PMID: 39588905 DOI: 10.1093/intimm/dxae067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/24/2024] [Indexed: 11/27/2024] Open
Abstract
Immune memory has long been considered a function specific to adaptive immune systems; however, adaptive immune memory alone has not fully explained the mechanism by which vaccines exert their protective effects against nontarget pathogens. Recently, trained immunity, in which human monocytes vaccinated with bacillus Calmette-Guérin become highly responsive to pathogens other than Mycobacterium tuberculosis, has been reported. However, a phenomenon called endotoxin tolerance is also known, in which monocyte responsiveness is attenuated after the first lipopolysaccharide stimulation. These phenomena represent an altered innate immune response after the initial exposure to the stimulus, indicating that memories are formed in the innate immune system. In this review, we discuss trained immunity and endotoxin tolerance, known as innate immune memory, and innate immune memory formation by mRNA vaccines, which have been newly used in the coronavirus disease 2019 (COVID-19) pandemic and are considered important vaccine modalities in the future.
Collapse
Affiliation(s)
- Yasuhiro Kato
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Atsushi Kumanogoh
- Department of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka, Japan
- Department of Respiratory Medicine and Clinical Immunology, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
- Center for Infectious Diseases Education and Research, Osaka University, Suita, Osaka, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka, Japan
- Japan Agency for Medical Research and Development-Core Research for Evolutional Science and Technology (AMED-CREST), Osaka University, Suita, Osaka, Japan
- Center for Advanced Modalities and DDS, Osaka University, Suita, Osaka, Japan
| |
Collapse
|
22
|
Du Y, Faz-Lopez B, Ah Kioon MD, Cenac C, Pierides M, Lakin KS, Spiera RF, Chaumeil J, Truchetet ME, Gordon JK, Guéry JC, Barrat FJ. Altered X-chromosome inactivation of the TLR7/8 locus and heterogeneity of pDCs in systemic sclerosis. J Exp Med 2025; 222:e20231809. [PMID: 39670995 PMCID: PMC11639950 DOI: 10.1084/jem.20231809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 09/26/2024] [Accepted: 11/21/2024] [Indexed: 12/14/2024] Open
Abstract
Systemic sclerosis (SSc) is an autoimmune disease that has a strong female predominance. Both the X-linked TLR7 and TLR8 can induce type I IFN (IFN-I) by plasmacytoid DCs (pDCs), which can promote fibrosis. We identified five subclusters of pDCs, including ISGhigh clusters that were over-represented in SSc patients. We observed that both TLR7 and TLR8 genes escape from X chromosome inactivation (XCI) at higher frequency in pDCs of SSc patients, which was associated with changes in TLR7 protein profile. Combined DNA/RNA FISH analysis revealed that the TLR7/8 locus is preferentially located outside of the inactive X (Xi) territory when TLR7 is expressed, suggesting that higher-order loop formation is linked to TLR7/8 expression from the Xi. Furthermore, the expression levels of XIST and the transcriptional repressor SPEN were reduced in SSc pDCs. Hence, our data revealed the heterogeneity of pDCs in SSc and suggested that altered XCI at the TLR7/8 locus may contribute to the chronic IFN-I activity of pDCs in female SSc patients.
Collapse
Affiliation(s)
- Yong Du
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| | - Bérénice Faz-Lopez
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Marie Dominique Ah Kioon
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Claire Cenac
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Michael Pierides
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
| | - Kimberly S. Lakin
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Robert F. Spiera
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Julie Chaumeil
- Institut Cochin, Université Paris Cité, CNRS, INSERM, Paris, France
| | - Marie-Elise Truchetet
- ImmunoConcEpt, CNRS, UMR 5164, University of Bordeaux, Talence, France
- Rheumatology Department, CHU de Bordeaux, Bordeaux, France
| | - Jessica K. Gordon
- Division of Rheumatology and Scleroderma and Vasculitis Center, Department of Medicine, Hospital for Special Surgery, New York, NY, USA
| | - Jean-Charles Guéry
- Institut Toulousain des Maladies Infectieuses et Inflammatoires, Université de Toulouse, INSERM, CNRS, UPS, Toulouse, France
| | - Franck J. Barrat
- HSS Research Institute and David Z. Rosensweig Genomics Research Center, Inflammation and Autoimmunity Program, Hospital for Special Surgery, New York, NY, USA
- Department of Microbiology and Immunology, Weill Cornell Medical College of Cornell University, New York, NY, USA
| |
Collapse
|
23
|
Li Y, Wang L, Chen S. An overview of the progress made in research into the Mpox virus. Med Res Rev 2025; 45:788-812. [PMID: 39318037 DOI: 10.1002/med.22085] [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: 03/26/2024] [Revised: 08/05/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
Mpox is a zoonotic illness caused by the Mpox virus (MPXV), a member of the Orthopoxvirus family. Although a few cases have been reported outside Africa, it was originally regarded as an endemic disease limited to African countries. However, the Mpox outbreak of 2022 was remarkable in that the infection spread to more than 123 countries worldwide, causing thousands of infections and deaths. The ongoing Mpox outbreak has been declared as a public health emergency of international concern by the World Health Organization. For a better management and control of the epidemic, this review summarizes the research advances and important scientific findings on MPXV by reviewing the current literature on epidemiology, clinical characteristics, diagnostic methods, prevention and treatment measures, and animal models of MPXV. This review provides useful information to raise awareness about the transmission, symptoms, and protective measures of MPXV, serving as a theoretical guide for relevant institutions to control MPXV.
Collapse
Affiliation(s)
- Yansheng Li
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Lianrong Wang
- Department of Respiratory Diseases, Institute of Pediatrics, Shenzhen Children's Hospital, Shenzhen, Guangdong, China
| | - Shi Chen
- Shenzhen Key Laboratory of Microbiology in Genomic Modification & Editing and Application, Medical Innovation Technology Transformation Center of Shenzhen Second People's Hospital, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound lmaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Department of Critical Care Medicine, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen Institute of Translational Medicine, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| |
Collapse
|
24
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
25
|
Unterberger S, Terrazzini N, Sacre S. Convalescent COVID-19 monocytes exhibit altered steady-state gene expression and reduced TLR2, TLR4 and RIG-I induced cytokine expression. Hum Immunol 2025; 86:111249. [PMID: 39922089 DOI: 10.1016/j.humimm.2025.111249] [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: 08/23/2024] [Revised: 01/12/2025] [Accepted: 01/21/2025] [Indexed: 02/10/2025]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), which causes COVID-19, can induce trained immunity in monocytes. Trained immunity is the result of metabolic and epigenetic reprogramming of progenitor cells leading to an altered inflammatory response to subsequent activation. To investigate the monocyte response 3-6 months post SARS-CoV-2 infection, steady-state gene expression and innate immune receptor stimulation were investigated in monocytes from unvaccinated SARS-CoV-2 naïve individuals and convalescent COVID-19 participants. The differentially expressed genes (DEGs) identified were involved in the regulation of innate immune signalling pathways associated with anti-viral defence. COVID-19 participants who had experienced severe symptoms exhibited a larger number of DEGs than participants that had mild symptoms. Interestingly, genes encoding receptors that recognise SARS-CoV-2 RNA were downregulated. DDX58, encoding retinoic-acid inducible gene I (RIG-I), was downregulated which corresponded with a reduced response to RIG-I activation. Furthermore, toll-like receptor (TLR)1/2 and TLR4 activation also exhibited reduced cytokine secretion from convalescent COVID-19 monocytes. These data suggest that following SARS-CoV-2 infection, monocytes exhibit altered steady-state gene expression and reduced responsiveness to innate immune receptor activation. As both RIG-I and TLRs recognise components of SARS-CoV-2, this may lead to a moderated inflammatory response to SARS-CoV-2 reinfection in the months following the initial infection.
Collapse
Affiliation(s)
- Sarah Unterberger
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK
| | - Nadia Terrazzini
- Centre for Regenerative Medicine and Devices, School of Applied Sciences, University of Brighton, Brighton, UK
| | - Sandra Sacre
- Brighton and Sussex Medical School, University of Sussex, Brighton, UK.
| |
Collapse
|
26
|
Chu Y, Wang J, Xiong Y, Gao Y, Liu X, Luo G, Gao X, Zhao M, Huang C, Qiu Z, Meng X. Point-annotation supervision for robust 3D pulmonary infection segmentation by CT-based cascading deep learning. Comput Biol Med 2025; 187:109760. [PMID: 39923589 DOI: 10.1016/j.compbiomed.2025.109760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/23/2025] [Accepted: 01/27/2025] [Indexed: 02/11/2025]
Abstract
Infected region segmentation is crucial for pulmonary infection diagnosis, severity assessment, and monitoring treatment progression. High-performance segmentation methods rely heavily on fully annotated, large-scale training datasets. However, manual labeling for pulmonary infections demands substantial investments of time and labor. While weakly supervised learning can greatly reduce annotation efforts, previous developments have focused mainly on natural or medical images with distinct boundaries and consistent textures. These approaches is not applicable to pulmonary infection segmentation, which should contend with high topological and intensity variations, irregular and ambiguous boundaries, and poor contrast in 3D contexts. In this study, we propose a cascading point-annotation framework to segment pulmonary infections, enabling optimization on larger datasets and superior performance on external data. Via comparing the representation of annotated points and unlabeled voxels, as well as establishing global uncertainty, we develop two regularization strategies to constrain the network to a more holistic lesion pattern understanding under sparse annotations. We further encompass an enhancement module to improve global anatomical perception and adaptability to spatial anisotropy, alongside a texture-aware variational module to determine more regionally consistent boundaries based on common textures of infection. Experiments on a large dataset of 1,072 CT volumes demonstrate our method outperforming state-of-the-art weakly-supervised approaches by approximately 3%-6% in dice score and is comparable to fully-supervised methods on external datasets. Moreover, our approach demonstrates robust performance even when applied to an unseen infection subtype, Mycoplasma pneumoniae, which was not included in the training datasets. These results collectively underscore rapid and promising applicability for emerging pulmonary infections.
Collapse
Affiliation(s)
- Yuetan Chu
- Center of Excellence for Smart Health (KCSH), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Jianpeng Wang
- The Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yaxin Xiong
- The Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yuan Gao
- The Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Xin Liu
- The Department of Prosthodontics, First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Gongning Luo
- Center of Excellence for Smart Health (KCSH), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Xin Gao
- Center of Excellence for Smart Health (KCSH), King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Mingyan Zhao
- The Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Chao Huang
- Ningbo Institute of Information Technology Application, Chinese Academy of Sciences (CAS), Ningbo, China.
| | - Zhaowen Qiu
- College of Computer and Control Engineering, Northeast Forestry University, Harbin, China.
| | - Xianglin Meng
- The Department of Critical Care Medicine, First Affiliated Hospital of Harbin Medical University, Harbin, China; The Cancer Institute and Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Shanghai, China.
| |
Collapse
|
27
|
Asaba CN, Bitazar R, Labonté P, Bukong TN. Bronchoalveolar lavage single-cell transcriptomics reveals immune dysregulations driving COVID-19 severity. PLoS One 2025; 20:e0309880. [PMID: 39928675 PMCID: PMC11809808 DOI: 10.1371/journal.pone.0309880] [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: 08/20/2024] [Accepted: 12/30/2024] [Indexed: 02/12/2025] Open
Abstract
The continuous threats posed by Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2), the virus that causes COVID-19, including the emergence of potentially more infectious and deadly variants, necessitate ongoing studies to uncover novel and detailed mechanisms driving disease severity. Using single-cell transcriptomics, we conducted a secondary data analysis of bronchoalveolar lavage fluid (BALF) from COVID-19 patients of varying severities and healthy controls to comprehensively examine immune responses. We observed significant immune cell alterations correlating with disease severity. In severe cases, macrophages showed upregulation of pro-inflammatory genes TNFα and IL1β, contributing to severe inflammation and tissue damage. Neutrophils exhibited increased activation, marked by S100A8, CXCL8, and IL1β expression, with extended viability and reduced phagocytosis. Genes such as MCL1 and HIF1α supported extended viability, while MSR1 and MRC1 indicated reduced phagocytosis. Enhanced formation of neutrophil extracellular traps (NETs) and reduced clearance, indicated by NET-associated markers, were linked to thrombo-inflammation and organ damage. Both macrophages and neutrophils in severe cases showed impaired efferocytosis, indicated by decreased expression of MSR1 and TREM2 in macrophages and downregulation of FCGR3B in neutrophils, leading to the accumulation of apoptotic cells and exacerbating inflammation. Severe cases were characterized by M1 macrophages with high TNFα and IL1β, while milder cases had M2 macrophages with elevated PPARγ. Dendritic cells (DCs) in severe cases exhibited reduced proportions and attenuated expression of MHC class I genes (HLA-A, HLA-B, HLA-C) and co-stimulatory molecules (CD80, CD86), alongside increased cytochrome c expression, indicating impaired antigen presentation and enhanced apoptosis. NK and T cells in severe cases demonstrated altered receptor and gene expression, with increased activation markers IFNγ and ISG15, suggesting a paradoxical state of activation and exhaustion. This analysis highlights the critical role of dysregulated neutrophil, macrophage, dendritic cell, NK, and T cell responses in severe COVID-19, identifying potential therapeutic targets and providing novel insights into the disease.
Collapse
Affiliation(s)
- Clinton Njinju Asaba
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Razieh Bitazar
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Patrick Labonté
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| | - Terence Ndonyi Bukong
- Armand-Frappier Santé Biotechnologie Research Center, Institut National de la Recherche Scientifique, Laval, Québec, Canada
| |
Collapse
|
28
|
Lim HX, Khalid K, Abdullah ADI, Lee LH, Raja Ali RA. Subphenotypes of Long COVID and the clinical applications of probiotics. Biomed Pharmacother 2025; 183:117855. [PMID: 39862702 DOI: 10.1016/j.biopha.2025.117855] [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: 10/21/2024] [Revised: 12/25/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
As the number of infections and deaths attributable to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection continues to rise, it is now becoming apparent that the health impacts of the Coronavirus disease (COVID-19) may not be limited to infection and the subsequent resolution of symptoms. Reports have shown that patients with SARS-CoV-2 infection may experience multiple symptoms across different organ systems that are associated with adverse health outcomes and develop new cardiac, renal, respiratory, musculoskeletal, and nervous conditions, a condition known as Long COVID or the post-acute sequelae of COVID-19 (PASC). This review provides insights into distinct subphenotypes of Long COVID and identifies microbiota dysbiosis as a common theme and crucial target for future therapies. Another important finding is that Long COVID is associated with prolonged and increased inflammation, potentially attributable to immune system dysfunction. A promising solution lies in the potential of probiotics to mitigate Long COVID symptoms by restoring gut microbiota balance and modulating the immune response. By evaluating the current clinical development landscape of the use of probiotics to treat Long COVID symptoms, this paper provides recommendations for future research by stressing the need to understand the modulation of bacterium strains followed by probiotic therapy to understand the association of microbiota dysbiosis with Long COVID symptoms. This will facilitate the development of effective probiotic formulations that could serve as reliable therapies against Long COVID.
Collapse
Affiliation(s)
- Hui Xuan Lim
- Sunway Microbiome Centre, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia.
| | - Kanwal Khalid
- Centre for Virus and Vaccine Research, School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia.
| | | | - Learn-Han Lee
- Microbiome Research Group, Research Centre for Life Science and Healthcare, Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute (CBI), University of Nottingham, Ningbo 315000, China
| | - Raja Affendi Raja Ali
- School of Medical and Life Sciences, Sunway University, Petaling Jaya, Selangor 47500, Malaysia.
| |
Collapse
|
29
|
Saha A, Ganguly A, Kumar A, Srivastava N, Pathak R. Harnessing Epigenetics: Innovative Approaches in Diagnosing and Combating Viral Acute Respiratory Infections. Pathogens 2025; 14:129. [PMID: 40005506 PMCID: PMC11858160 DOI: 10.3390/pathogens14020129] [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: 01/09/2025] [Revised: 01/26/2025] [Accepted: 01/28/2025] [Indexed: 02/27/2025] Open
Abstract
Acute respiratory infections (ARIs) caused by viruses such as SARS-CoV-2, influenza viruses, and respiratory syncytial virus (RSV), pose significant global health challenges, particularly for the elderly and immunocompromised individuals. Substantial evidence indicates that acute viral infections can manipulate the host's epigenome through mechanisms like DNA methylation and histone modifications as part of the immune response. These epigenetic alterations can persist beyond the acute phase, influencing long-term immunity and susceptibility to subsequent infections. Post-infection modulation of the host epigenome may help distinguish infected from uninfected individuals and predict disease severity. Understanding these interactions is crucial for developing effective treatments and preventive strategies for viral ARIs. This review highlights the critical role of epigenetic modifications following viral ARIs in regulating the host's innate immune defense mechanisms. We discuss the implications of these modifications for diagnosing, preventing, and treating viral infections, contributing to the advancement of precision medicine. Recent studies have identified specific epigenetic changes, such as hypermethylation of interferon-stimulated genes in severe COVID-19 cases, which could serve as biomarkers for early detection and disease progression. Additionally, epigenetic therapies, including inhibitors of DNA methyltransferases and histone deacetylases, show promise in modulating the immune response and improving patient outcomes. Overall, this review provides valuable insights into the epigenetic landscape of viral ARIs, extending beyond traditional genetic perspectives. These insights are essential for advancing diagnostic techniques and developing innovative treatments to address the growing threat of emerging viruses causing ARIs globally.
Collapse
Affiliation(s)
- Ankita Saha
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India;
| | - Anoop Kumar
- Molecular Diagnostic Laboratory, National Institute of Biologicals, Noida 201309, India;
| | - Nityanand Srivastava
- Department of Cell Biology, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; (A.S.); (N.S.)
| | - Rajiv Pathak
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| |
Collapse
|
30
|
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
|
31
|
Hajishengallis G, Netea MG, Chavakis T. Trained immunity in chronic inflammatory diseases and cancer. Nat Rev Immunol 2025:10.1038/s41577-025-01132-x. [PMID: 39891000 DOI: 10.1038/s41577-025-01132-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
A decade after the term 'trained immunity' (TRIM) was coined to reflect the long-lasting hyper-responsiveness of innate immune cells with an epigenetically imprinted 'memory' of earlier stimuli, our understanding has broadened to include the potential implications of TRIM in health and disease. Here, after summarizing the well-documented beneficial effects of TRIM against infections, we discuss emerging evidence that TRIM is also a major underlying mechanism in chronic inflammation-related disorders such as periodontitis, rheumatoid arthritis and cardiovascular disease. Furthermore, mounting evidence indicates that the induction of TRIM by certain agonists confers protective antitumour responses. Although the mechanisms underlying TRIM require further study, the current knowledge enables the experimental development of innovative therapeutic approaches to stimulate or inhibit TRIM in a context-appropriate manner, such as the stimulation of TRIM in cancer or its inhibition in inflammatory disorders.
Collapse
Affiliation(s)
- George Hajishengallis
- Department of Basic and Translational Sciences, Penn Dental Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| | - Mihai G Netea
- Department of Internal Medicine and Radboud Center for Infectious Diseases, Radboud University Medical Center, Nijmegen, the Netherlands.
- Department of Immunology and Metabolism, LIMES, University of Bonn, Bonn, Germany.
| | - Triantafyllos Chavakis
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.
| |
Collapse
|
32
|
Marzetti E, Coelho-Júnior HJ, Calvani R, Girolimetti G, Di Corato R, Ciciarello F, Galluzzo V, Di Mario C, Tolusso B, Santoro L, Giampaoli O, Tomassini A, Aureli W, Tosato M, Landi F, Bucci C, Guerra F, Picca A. Mitochondria-Derived Vesicles and Inflammatory Profiles of Adults with Long COVID Supplemented with Red Beetroot Juice: Secondary Analysis of a Randomized Controlled Trial. Int J Mol Sci 2025; 26:1224. [PMID: 39940991 PMCID: PMC11818272 DOI: 10.3390/ijms26031224] [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: 01/02/2025] [Revised: 01/27/2025] [Accepted: 01/29/2025] [Indexed: 02/16/2025] Open
Abstract
In a recent clinical trial, beetroot juice supplementation for 14 days yielded positive effects on systemic inflammation in adults with long COVID. Here, we explored the relationship between circulating markers of mitochondrial quality and inflammation in adults with long COVID as well as the impact of beetroot administration on those markers. We conducted secondary analyses of a placebo-controlled randomized clinical trial testing beetroot juice supplementation as a remedy against long COVID. Analyses were conducted in 25 participants, 10 assigned to placebo (mean age: 40.2 ± 11.5 years, 60% women) and 15 allocated to beetroot juice (mean age: 38.3 ± 7.7 years, 53.3% women). Extracellular vesicles were purified from serum by ultracentrifugation and assayed for components of the electron transport chain and mitochondrial DNA (mtDNA) by Western blot and droplet digital polymerase chain reaction (ddPCR), respectively. Inflammatory markers and circulating cell-free mtDNA were quantified in serum through a multiplex immunoassay and ddPCR, respectively. Beetroot juice administration for 14 days decreased serum levels of interleukin (IL)-1β, IL-8, and tumor necrosis factor alpha, with no effects on circulating markers of mitochondrial quality control. Significant negative associations were observed between vesicular markers of mitochondrial quality control and the performance on the 6 min walk test and flow-mediated dilation irrespective of group allocation. These findings suggest that an amelioration of mitochondrial quality, possibly mediated by mitochondria-derived vesicle recycling, may be among the mechanisms supporting improvements in physical performance and endothelial function during the resolution of long COVID.
Collapse
Affiliation(s)
- Emanuele Marzetti
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Hélio José Coelho-Júnior
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Riccardo Calvani
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Giulia Girolimetti
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni, 73100 Lecce, Italy; (G.G.); (C.B.)
| | - Riccardo Di Corato
- Institute for Microelectronics and Microsystems (IMM), CNR, Via Provinciale Lecce-Monteroni, 73100 Lecce, Italy;
- Center for Biomolecular Nanotechnologies, Istituto Italiano di Tecnologia, 73010 Arnesano, Italy
| | - Francesca Ciciarello
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
| | - Vincenzo Galluzzo
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
| | - Clara Di Mario
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
| | - Barbara Tolusso
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
| | - Luca Santoro
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
| | - Ottavia Giampaoli
- Department of Environmental Biology, Sapienza University of Rome, Piazzale Aldo Moro 5, 00185 Rome, Italy;
- NMR-Based Metabolomics Laboratory (NMLab), Sapienza University of Rome, 00185 Rome, Italy
| | - Alberta Tomassini
- R&D, Aureli Mario S.S. Agricola, Via Mario Aureli 7, 67050 Ortucchio, Italy; (A.T.); (W.A.)
| | - Walter Aureli
- R&D, Aureli Mario S.S. Agricola, Via Mario Aureli 7, 67050 Ortucchio, Italy; (A.T.); (W.A.)
| | - Matteo Tosato
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
| | - Francesco Landi
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
- Department of Geriatrics, Orthopedics and Rheumatology, Università Cattolica del Sacro Cuore, 00168 Rome, Italy
| | - Cecilia Bucci
- Department of Experimental Medicine, University of Salento, Via Provinciale Lecce-Monteroni, 73100 Lecce, Italy; (G.G.); (C.B.)
| | - Flora Guerra
- Department of Biological and Environmental Sciences and Technologies, Università del Salento, 73100 Lecce, Italy;
| | - Anna Picca
- Fondazione Policlinico Universitario “Agostino Gemelli” IRCCS, 00168 Rome, Italy; (E.M.); (H.J.C.-J.); (F.C.); (V.G.); (C.D.M.); (B.T.); (L.S.); (M.T.); (F.L.); (A.P.)
- Department of Medicine and Surgery, LUM University, Str. Statale 100 Km 18, 70010 Casamassima, Italy
| |
Collapse
|
33
|
O’Farrell A, Niu Z, Li J, Van Eyndhoven LC, Sarma K, Raj A. Innate Immune Memory is Stimulus Specific. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.634275. [PMID: 39896612 PMCID: PMC11785108 DOI: 10.1101/2025.01.22.634275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Innate immune memory (also termed trained immunity) is defined in part by its ability to cross-protect against heterologous pathogens, and can be generated by many different stimuli, suggesting a "universal" trained state. However, different stimuli could form distinct memories, leading to stimulus-specific trained responses. Here, we use primary human monocyte-derived macrophages to demonstrate phenotypic and epigenetic stimulus specificity of innate immune memory six days after initial exposure. Quantification of cytokine production with single-molecule RNA imaging demonstrates stimulus-specific patterns of response to restimulation at the single cell level. Differential licensing of inflammatory transcription factors is associated with encoding of specificities in chromatin. Trained cells show stronger responses to secondary stimuli that are more similar to the initial stimulus they experienced, suggesting a functional role for these stimulus-specific memories. Rather than activating a universal training state, our findings demonstrate that different stimuli impart specific memories that generate distinct training phenotypes in macrophages.
Collapse
Affiliation(s)
- Aoife O’Farrell
- Department of Bioengineering, School of Engineering and Applied Sciences University of Pennsylvania, Philadelphia, PA, USA
| | - Zijian Niu
- Department of Chemistry, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Department of Physics and Astronomy, School of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
- Computational and Systems Biology Program, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Jingxin Li
- Genetics and Epigenetics, Cell and Molecular Biology Graduate Group, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Laura C. Van Eyndhoven
- Department of Bioengineering, School of Engineering and Applied Sciences University of Pennsylvania, Philadelphia, PA, USA
| | - Kavitha Sarma
- The Wistar Institute, Gene Expression and Regulation Program, Philadelphia, PA, USA
- Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Arjun Raj
- Department of Bioengineering, School of Engineering and Applied Sciences University of Pennsylvania, Philadelphia, PA, USA
- Department of Genetics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
34
|
Aldridge DL, Lanzar Z, Phan AT, Christian DA, Pardy R, Min B, Kedl RM, Hunter CA. IL-27 limits HSPC differentiation during infection and protects from stem cell exhaustion. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.15.633135. [PMID: 39868131 PMCID: PMC11761129 DOI: 10.1101/2025.01.15.633135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Many inflammatory stimuli can induce progenitor cells in the bone marrow to produce increased numbers of myeloid cells as part of the process of emergency myelopoiesis. These events are associated with innate training and can have long-term impacts on hematopoietic stem and progenitor cell (HSPC) development but can also compromise their function. While many cytokines support emergency myelopoiesis, less is known about the mechanisms that temper these events. When mice that lack the cytokine IL-27 were infected with Toxoplasma gondii, there was enhanced generation of monocyte progenitors and increased numbers of inflammatory monocytes. In the bone marrow of infected mice there was increased production of IL-27 that localized with HSPCs and a survey of cytokine receptor expression highlighted that HSPCs were uniquely poised to respond to IL-27. Furthermore, the use of in vitro differentiation assays and mixed bone marrow chimeras revealed that HSPCs from IL-27 deficient mice are pre-disposed towards the monocyte lineage. Additional studies highlighted that after infection loss of the IL-27R resulted in reduced HSPC fitness that manifested as reduced proliferative responses and a decreased ability to reconstitute the hematopoietic system. Thus, the ability of IL-27 to act on HSPC provides a regulatory brake on differentiation to limit monocyte induction and preserve HSPC stemness.
Collapse
Affiliation(s)
- Daniel L Aldridge
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Zachary Lanzar
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Anthony T Phan
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - David A Christian
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Ryan Pardy
- University of Pennsylvania School of Veterinary Medicine, Philadelphia, PA, USA
| | - Booki Min
- Department of Microbiology and Immunology, Feinber School of Medicine, Northwestern University, Chicago, IL, USA
| | - Ross M Kedl
- University of Colorado, Anschuitz Medical Campus, Aurora, CO, USA
| | | |
Collapse
|
35
|
Marchand V, Laplane L, Valensi L, Plo I, Aglave M, Silvin A, Pasquier F, Porteu F, Vainchenker W, Selimoglu-Buet D, Droin N, Raslova H, Marcel V, Diaz JJ, Fontenay M, Solary E. Monocytes generated by interleukin-6-treated human hematopoietic stem and progenitor cells secrete calprotectin that inhibits erythropoiesis. iScience 2025; 28:111522. [PMID: 39811665 PMCID: PMC11732210 DOI: 10.1016/j.isci.2024.111522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/02/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
Elevated circulating levels of calprotectin (CAL), the S100A8/A9 heterodimer, are biomarkers of severe systemic inflammation. Here, we investigate the effects of CAL on early human hematopoiesis. CAL demonstrates limited impact on gene expression in stem and progenitor cells, in contrast with interleukin-6 (IL6), which promotes the expression of the S100A8 and S100A9 genes in hematopoietic progenitors and the generation of monocytes that release CAL. The main target of CAL is an erythroid-megakaryocyte progenitor (EMP) subset. CAL prevents both erythropoietin-driven differentiation of healthy progenitors and JAK2-V617F-driven erythropoiesis. In the context of JAK2-V617F, CAL also promotes the expression of S100A8 and S100A9 genes in monocytes. The signature of CAL effects is detected in the bone marrow progenitors of patients with myeloid malignancy or severe infection. These results position CAL as a mediator of IL6 effects on triggering anemia during inflammation, an effect that is amplified in the context of JAK2-V617F-driven hematopoiesis.
Collapse
Affiliation(s)
- Valentine Marchand
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Lucie Laplane
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- CNRS 8590, Université Paris 1 Panthéon-Sorbonne, Paris, France
| | - Louis Valensi
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Isabelle Plo
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Marine Aglave
- AMMICa, INSERM US 23, CNRS UMS 3655, Gustave Roussy Cancer Center, Villejuif, France
| | - Aymeric Silvin
- INSERM U1108, Gustave Roussy Cancer Center, Villejuif, France
| | - Florence Pasquier
- Department of Hematology, Gustave Roussy Cancer Center, Villejuif, France
| | - Françoise Porteu
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - William Vainchenker
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | | | - Nathalie Droin
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- AMMICa, INSERM US 23, CNRS UMS 3655, Gustave Roussy Cancer Center, Villejuif, France
| | - Hana Raslova
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
| | - Virginie Marcel
- Inserm U1052, CNRS UMR5286 Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Jean-Jacques Diaz
- Inserm U1052, CNRS UMR5286 Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Michaela Fontenay
- Université Paris Cité, Institut Cochin, CNRS UMR 8104, INSERM U1016, Paris, France
- Laboratory of Excellence for Red Blood Cells, GR-Ex, Paris, France
| | - Eric Solary
- INSERM U1287, Université Paris-Saclay, Gustave Roussy Cancer Center, Villejuif, France
- Université Paris-Saclay, Faculté de Médecine, Le Kremlin-Bicêtre, France
| |
Collapse
|
36
|
Jarras H, Blais I, Goyer B, Bazié WW, Rabezanahary H, Thériault M, Santerre K, Langlois MA, Masson JF, Pelletier JN, Brousseau N, Boudreau D, Trottier S, Baz M, Gilbert C. Impact of SARS-CoV-2 vaccination and of seasonal variations on the innate immune inflammatory response. Front Immunol 2025; 15:1513717. [PMID: 39877354 PMCID: PMC11772892 DOI: 10.3389/fimmu.2024.1513717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Accepted: 12/27/2024] [Indexed: 01/31/2025] Open
Abstract
Introduction The innate immune response is an important first checkpoint in the evolution of an infection. Although adaptive immunity is generally considered the immune component that retains antigenic memory, innate immune responses can also be affected by previous stimulations. This study evaluated the impact of vaccination on innate cell activation by TLR7/8 agonist R848, as well as seasonal variations. Methods To this end, blood samples from a cohort of 304 food and retail workers from the Quebec City region were collected during three visits at 12-week intervals. Peripheral blood mononuclear cells and polymorphonuclear neutrophils were isolated during the first and third visits and were stimulated with R848 to assess the innate immune response. Results Our results show that IL-8 production after stimulation decreased after vaccination. In addition, the IL-8 response was significantly different depending on the season when the visit occurred, for both COVID-19 vaccinated and unvaccinated individuals. Discussion This study highlights that innate immune responses can be affected by SARS-CoV-2 vaccination and fluctuate seasonally.
Collapse
Affiliation(s)
- Hend Jarras
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Isalie Blais
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Benjamin Goyer
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Wilfried W. Bazié
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Programme de Recherche sur les Maladies Infectieuses, Centre Muraz, Institut National de Santé Publique, Bobo-Dioulasso, Houet, Burkina Faso
| | - Henintsoa Rabezanahary
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Mathieu Thériault
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Kim Santerre
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
| | - Marc-André Langlois
- Department of Biochemistry, Microbiology and Immunology, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Jean-François Masson
- Department of Chemistry, Institut Courtois, Quebec Center for Advanced Materials, Regroupement québécois sur les matériaux de pointe, and Centre Interdisciplinaire de Recherche sur le Cerveau et l’Apprentissage, Université de Montréal, Montreal, QC, Canada
| | - Joelle N. Pelletier
- Department of Chemistry, Department of Biochemistry, Université de Montréal, Montreal, QC, Canada
- PROTEO — The Québec Network for Research on Protein Function, Engineering, and Applications, Quebec City, QC, Canada
| | - Nicholas Brousseau
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Biological Risks Department, Institut national de santé publique du Québec, Quebec City, QC, Canada
| | - Denis Boudreau
- Département de Chimie et Center for Optics, Photonics and Lasers (COPL), Université Laval, Quebec City, QC, Canada
| | - Sylvie Trottier
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Mariana Baz
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| | - Caroline Gilbert
- Axe de Recherche Maladies Infectieuses et Immunitaires, Centre de Recherche du CHU de Québec-Université Laval, Quebec City, QC, Canada
- Département de Microbiologie-Infectiologie et d’Immunologie, Faculté de Médecine, Université Laval, Quebec City, QC, Canada
| |
Collapse
|
37
|
Ucar D, Thibodeau A, Mejias A, Nehar-Belaid D, Marches R, Xu Z, Eryilmaz G, Josefowicz S, Paust S, Pascual V, Banchereau J, Ramilo O. Infants display reduced NK cell responses in RSV and increased inflammatory responses in SARS-CoV-2 infections. RESEARCH SQUARE 2025:rs.3.rs-5640872. [PMID: 39877087 PMCID: PMC11774461 DOI: 10.21203/rs.3.rs-5640872/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/31/2025]
Abstract
Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infection hospitalizations in infants and poses a significantly higher risk of respiratory failure than SARS-CoV-2. The mechanisms underlying these differences remain unclear. We analyzed blood samples from infants (median age 2.3 months) with SARS-CoV-2 (n = 30), RSV (n = 19), and healthy controls (n = 17) using single-cell transcriptomics and epigenomics, and cytokine profiling. Both viruses triggered comparable interferon responses across PBMC subsets but differed in NK cell and inflammatory responses. Severe RSV cases showed reduced NK cell frequencies, lower IFNG expression, and decreased chromatin accessibility at T-BET and EOMES binding sites. RSV infections were also associated with increased CD4+ TEMRA, memory Treg and transitional B cells. In contrast, SARS-CoV-2 was characterized by stronger pro-inflammatory signatures, including increased NFKB pathway activity and higher serum TNF concentrations. These findings highlight distinct immune responses to RSV and SARS-CoV-2, providing insights that may inform clinical decisions.
Collapse
Affiliation(s)
| | | | - Asuncion Mejias
- Department of Infectious Diseases, St. Jude Children's Research Hospital
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
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
|
39
|
Kim GS, Kwak DY, Kim HW, Shin S, Ko MK, Hwang SY, Park SH, Kim DH, Park JH, Kim SM, Lee MJ. Levamisole, as a viral vaccine adjuvant, induces robust host defense through the modulation of innate and adaptive immune responses. Front Microbiol 2025; 15:1493561. [PMID: 39845058 PMCID: PMC11751227 DOI: 10.3389/fmicb.2024.1493561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/17/2024] [Indexed: 01/24/2025] Open
Abstract
Introduction An effective vaccination policy must be implemented to prevent foot-and-mouth disease (FMD). However, the currently used vaccines for FMD have several limitations, including induction of humoral rather than cellular immune responses. Methods To overcome these shortcomings, we assessed the efficacy of levamisole, a small-molecule immunomodulator, as an adjuvant for the FMD vaccine. We conducted in vitro studies using murine peritoneal exudate cells (PECs) and porcine peripheral blood mononuclear cells (PBMCs) and in vivo studies using mice (experimental animals) and pigs (target animals). We evaluated levamisole-mediated modulation of the innate and adaptive immune responses; early, mid-term, and long-term immune-inducing effects; modes of action; and host defense against viral infection. Results Levamisole treatment promoted IFNγ secretion in murine PECs and porcine PBMCs. Additionally, it induced robust and long-lasting immune responses by eliciting high antibody titers and high virus-neutralizing antibody titers. By activating downstream signaling pathways of various pattern-recognition receptors, levamisole stimulated the expression of multiple cytokines and costimulatory molecules. Owing to these immunostimulatory effects, levamisole elicited host defense against viral infections in pigs. Our findings demonstrate the potential of levamisole as an immunostimulatory agent. Discussion The results also indicate that levamisole, as an adjuvant for animal vaccines, can elicit robust innate and adaptive immune responses, thereby enhancing host defense against viral infections. This study provides a promising approach for the development of improved FMD vaccine strategies in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Min Ja Lee
- Center for Foot-and-Mouth Disease Vaccine Research, Animal and Plant Quarantine Agency, Gimcheon-si, Republic of Korea
| |
Collapse
|
40
|
Tang Z, Chen Y, Ouyang Y, Peng Y, Man X. COVID-19 related epigenetic changes and atopic dermatitis: An exploratory analysis. World Allergy Organ J 2025; 18:101022. [PMID: 39867872 PMCID: PMC11758953 DOI: 10.1016/j.waojou.2024.101022] [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: 05/21/2024] [Revised: 09/13/2024] [Accepted: 12/06/2024] [Indexed: 01/28/2025] Open
Abstract
Background While epidemiological data suggest a connection between atopic dermatitis (AD) and COVID-19, the molecular mechanisms underlying this relationship remain unclear. Objective To investigate whether COVID-19-related CpGs may contribute to AD development and whether this association is mediated through the regulation of specific genes' expression. Methods We combined Mendelian randomization and transcriptome analysis for data-driven explorations. Results Among the 172 CpGs -associated with COVID-19 infection, merely 3 of them exhibited significant impacts on the risk of AD, including cg04543273, cg11916609, and cg10636246. In the following analysis of the causal effects of CpGs and their related gene expression, cg04543273 inhibited LMAN2 expression. However, there was not a significant impact of cg11916609 and cg10636246 on the expression of their corresponding genes. Besides, transcriptome analysis suggested that LMAN2 expression was significantly upregulated among the COVID-19-infected population, and LMAN2 expression was obviously correlated with Type 2 helper cells across different post-infection time points. Conclusion Overall, this study provides new insights of the COVID-19-related onset and exacerbation of AD-COVID-19-related epigenetic changes and their regulatory impact on transcription. A novel role of LMAN2 was proposed in the relationship between viral infection and AD. More studies are warranted to further explore the mechanism of LMAN2-related immunopathology.
Collapse
Affiliation(s)
- Zhenwei Tang
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Chen
- Clinical Medicine Eight-year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Yuzhen Ouyang
- Clinical Medicine Eight-year Program, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Peng
- Department of Rheumatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyong Man
- Department of Dermatology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
41
|
Cáceres E, Divani AA, Viñan-Garces AE, Olivella-Gomez J, Quintero-Altare A, Pérez S, Reyes LF, Sasso N, Biller J. Tackling persistent neurological symptoms in patients following acute COVID-19 infection: an update of the literature. Expert Rev Neurother 2025; 25:67-83. [PMID: 39715694 DOI: 10.1080/14737175.2024.2440543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 12/06/2024] [Indexed: 12/25/2024]
Abstract
INTRODUCTION The COVID-19 pandemic has taught myriad lessons and left several questions we are yet to comprehend. Initially, the scientific community was concerned with the management of acute disease and immunization. Once the peak of the pandemic receded, it became clear that a proportion of patients were far from fully recovered. Researchers started to recognize those persisting symptoms as a new entity termed 'Long COVID,' where neurological symptoms are evident and have a major impact on quality of life. AREAS COVERED The main purpose of this narrative review is to analyze and synthesize the current literature regarding Long COVID, its relation to the nervous system, and to explore the evidence on treatments for persistent neurological symptoms. The most common reported and observed neurologic manifestations include fatigue, cognitive impairment, pain, polyneuropathy, and neuropsychiatric disorders. A variety of pharmacologic and non-pharmacologic therapies have been evaluated and yielded mixed results. Many of them focused on immunomodulation and none currently have U.S. FDA approval. EXPERT OPINION Challenges remain in terms of clinical characterization and prognosis of Long COVID, besides understanding its pathophysiology. Standardization of biomarkers and diagnostic criteria will allow the use of common nomenclature and data elements in the design of future clinical studies.
Collapse
Affiliation(s)
- Eder Cáceres
- Unisabana Center for Translational Science, School of Medicine, Universidad de La Sabana, Chía, Colombia
- School of Engineering, Universidad de La Sabana, Chía, Colombia
- Department of Critical Care, Clínica Universidad de La Sabana, Chía, Colombia
| | - Afshin A Divani
- Department of Neurology, The University of New Mexico, Albuquerque, NM, USA
| | | | - Juan Olivella-Gomez
- Department of Critical Care, Clínica Universidad de La Sabana, Chía, Colombia
| | | | - Sebastián Pérez
- Department of Critical Care, Clínica Universidad de La Sabana, Chía, Colombia
| | - Luis F Reyes
- Unisabana Center for Translational Science, School of Medicine, Universidad de La Sabana, Chía, Colombia
- Pandemic Sciences Institute, University of Oxford, Oxford, UK
| | - Nicholas Sasso
- Department of Neurology, Loyola University Stritch School of Medicine, Loyola University Health System, Maywood, IL, USA
| | - Jose Biller
- Department of Neurology, Loyola University Stritch School of Medicine, Loyola University Health System, Maywood, IL, USA
| |
Collapse
|
42
|
Camici M, Del Duca G, Brita AC, Antinori A. Connecting dots of long COVID-19 pathogenesis: a vagus nerve- hypothalamic-pituitary- adrenal-mitochondrial axis dysfunction. Front Cell Infect Microbiol 2024; 14:1501949. [PMID: 39735263 PMCID: PMC11671747 DOI: 10.3389/fcimb.2024.1501949] [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: 09/25/2024] [Accepted: 11/25/2024] [Indexed: 12/31/2024] Open
Abstract
The pathogenesis of long COVID (LC) still presents many areas of uncertainty. This leads to difficulties in finding an effective specific therapy. We hypothesize that the key to LC pathogenesis lies in the presence of chronic functional damage to the main anti-inflammatory mechanisms of our body: the three reflexes mediated by the vagus nerve, the hypothalamic-pituitary-adrenal (HPA) hormonal axis, and the mitochondrial redox status. We will illustrate that this neuro-endocrine-metabolic axis is closely interconnected and how the SARS-CoV-2 can damage it at all stages through direct, immune-inflammatory, epigenetic damage mechanisms, as well as through the reactivation of neurotropic viruses. According to our theory, the direct mitochondrial damage carried out by the virus, which replicates within these organelles, and the cellular oxidative imbalance, cannot be countered in patients who develop LC. This is because their anti-inflammatory mechanisms are inconsistent due to reduced vagal tone and direct damage to the endocrine glands of the HPA axis. We will illustrate how acetylcholine (ACh) and cortisol, with its cytoplasmatic and cellular receptors respectively, are fundamental players in the LC process. Both Ach and cortisol play multifaceted and synergistic roles in reducing inflammation. They achieve this by modulating the activity of innate and cell-mediated immunity, attenuating endothelial and platelet activation, and modulating mitochondrial function, which is crucial for cellular energy production and anti-inflammatory mechanisms. In our opinion, it is essential to study the sensitivity of the glucocorticoids receptor in people who develop LC and whether SARS-CoV-2 can cause long-term epigenetic variations in its expression and function.
Collapse
Affiliation(s)
- Marta Camici
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Giulia Del Duca
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Anna Clelia Brita
- Department of Clinical Psychology, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| | - Andrea Antinori
- Clinical and Research Infectious Diseases Department, National Institute for Infectious Diseases Lazzaro Spallanzani Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Rome, Italy
| |
Collapse
|
43
|
Mu Z, Randolph HE, Aguirre-Gamboa R, Ketter E, Dumaine A, Locher V, Brandolino C, Liu X, Kaufmann DE, Barreiro LB, Li YI. Impact of disease-associated chromatin accessibility QTLs across immune cell types and contexts. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.05.24318552. [PMID: 39711700 PMCID: PMC11661428 DOI: 10.1101/2024.12.05.24318552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2024]
Abstract
Only a third of immune-associated loci from genome-wide association studies (GWAS) colocalize with expression quantitative trait loci (eQTLs). To learn about causal genes and mechanisms at the remaining loci, we created a unified single-cell chromatin accessibility (scATAC-seq) map in peripheral blood comprising a total of 282,424 cells from 48 individuals. Clustering and topic modeling of scATAC data identified discrete cell-types and continuous cell states, which helped reveal disease-relevant cellular contexts, and allowed mapping of genetic effects on chromatin accessibility across these contexts. We identified 37,390 chromatin accessibility QTLs (caQTL) at 10% FDR across eight cell groups and observed extensive sharing of caQTLs across immune cell contexts, finding that fewer than 20% of caQTLs are specific to a single cell type. Notably, caQTLs colocalized with ∼50% more GWAS loci compared to eQTLs, helping to nominate putative causal genes for many unexplained loci. However, most GWAS-caQTL colocalizations had no detectable downstream regulatory effects on gene expression levels in the same cell type. We find evidence that the higher rates of colocalization between caQTLs and GWAS signals reflect missing disease-relevant cellular contexts among existing eQTL studies. Thus, there remains a pressing need for identifying disease-causing cellular contexts and for mapping gene regulatory variation in these cells.
Collapse
|
44
|
Ruffinatto L, Groult Y, Iacono J, Sarrazin S, de Laval B. Hematopoietic stem cell a reservoir of innate immune memory. Front Immunol 2024; 15:1491729. [PMID: 39720722 PMCID: PMC11666435 DOI: 10.3389/fimmu.2024.1491729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 10/30/2024] [Indexed: 12/26/2024] Open
Abstract
Hematopoietic stem cells (HSCs) are a rare, long-lived and multipotent population that give rise to majority of blood cells and some tissue-resident immune cells. There is growing evidence that inflammatory stimuli can trigger persistent reprogramming in HSCs that enhances or inhibits the cellular functions of these HSCs and their progeny in response to subsequent infections. This newly discovered property makes HSCs a reservoir for innate immune memory. The molecular mechanisms underlying innate immune memory in HSCs are similar to those observed in innate immune cells, although their full elucidation is still pending. In this review, we examine the current state of knowledge on how an inflammatory response leads to reprogramming of HSCs. Understanding the full spectrum of consequences of reshaping early hematopoiesis is critical for assessing the potential benefits and risks under physiological and pathological conditions.
Collapse
Affiliation(s)
| | | | | | | | - Bérengère de Laval
- Aix Marseille University, Centre National de la Recherche Scientifique (CNRS), Institut
National de la Santé et de la Recherche Médicale (INSERM), Centre d’Immunologie de Marseille-Luminy (CIML), Marseille, France
| |
Collapse
|
45
|
Yang J, Cui S, Shao B, Zhao Y, Wang Z, Liu Q, Zhang Y, Yang D. ScRNA-seq reveals trained immunity-engaged Th17 cell activation against Edwardsiella piscicida-induced intestinal inflammation in teleost. Microbiol Res 2024; 289:127912. [PMID: 39326350 DOI: 10.1016/j.micres.2024.127912] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/19/2024] [Accepted: 09/13/2024] [Indexed: 09/28/2024]
Abstract
Mucosal immunity typically involves innate and adaptive immune cells, while the cellular mechanism of teleost's intestinal immune cells that engages gut homeostasis against bacterial infection remains largely unknown. Taking advantage of the enteric fish pathogen (Edwardsiella piscicida) infection-induced intestinal inflammation in turbot (Scophthalmus maximus), we find that β-glucan training could mitigate the bacterial infection-induced intestinal inflammation. Through single-cell transcriptome profiling and cellular function analysis, we identify that E. piscicida infection could tune down the activation of intestinal Th17 cells, while β-glucan-training could preserve the potential to amplify and restore the function of intestinal Th17 cells. Moreover, through pharmacological inhibitor treatment, we identify that Th17 cells are essential for ameliorating bacterial infection-induced intestinal inflammation in teleost. Taken together, these results suggest a new concept of trained immunity activation to regulate the intestinal Th17 cells' function, which might contribute to better developing strategies for maintaining gut homeostasis against bacterial infection.
Collapse
Affiliation(s)
- Jin Yang
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China
| | - Shu Cui
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China
| | - Boning Shao
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China
| | - Yanbo Zhao
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China
| | - Zhuang Wang
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Yuanxing Zhang
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China; Southern Marine Science and Engineering Guangdong Laboratory (Zhuhai), Zhuhai, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China
| | - Dahai Yang
- State Key Laboratory of Bioreactor Engineering, Laboratory for Aquatic Animal Diseases, East China University of Science and Technology, Shanghai 200237, China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao, China; Shanghai Engineering Research Center of Maricultured Animal Vaccines, Shanghai 200237, China.
| |
Collapse
|
46
|
Luo EY, Chuen-Chung Chang R, Gilbert-Jaramillo J. SARS-CoV-2 infection in microglia and its sequelae: What do we know so far? Brain Behav Immun Health 2024; 42:100888. [PMID: 39881814 PMCID: PMC11776083 DOI: 10.1016/j.bbih.2024.100888] [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] [Revised: 09/22/2024] [Accepted: 10/05/2024] [Indexed: 01/31/2025] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) caused the COVID-19 pandemic. After the success of therapeutics and worldwide vaccination, the long-term sequelae of SARS-CoV-2 infections are yet to be determined. Common symptoms of COVID-19 include the loss of taste and smell, suggesting SARS-CoV-2 infection has a potentially detrimental effect on neurons within the olfactory/taste pathways, with direct access to the central nervous system (CNS). This could explain the detection of SARS-CoV-2 antigens in the brains of COVID-19 patients. Different viruses display neurotropism that causes impaired neurodevelopment and/or neurodegeneration. Hence, it is plausible that COVID-19-associated neuropathologies are directly driven by SARS-CoV-2 infection in the CNS. Microglia, resident immune cells of the brain, are constantly under investigation as their surveillance role has been suggested to act as a friend or a foe impacting the progression of neurological disorders. Herein, we review the current literature suggesting microglia potentially been a susceptible target by SARS-CoV-2 virions and their role in viral dissemination within the CNS. Particular attention is given to the different experimental models and their translational potential.
Collapse
Affiliation(s)
- Echo Yongqi Luo
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Raymond Chuen-Chung Chang
- Laboratory of Neurodegenerative Diseases, School of Biomedical Sciences, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
- State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Javier Gilbert-Jaramillo
- James & Lillian Martin Centre, Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford, OX1 3RE, UK
| |
Collapse
|
47
|
Capriotti Z, Klase Z. Innate immune memory in chronic HIV and HIV-associated neurocognitive disorders (HAND): potential mechanisms and clinical implications. J Neurovirol 2024; 30:451-476. [PMID: 39733092 PMCID: PMC11846772 DOI: 10.1007/s13365-024-01239-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 11/26/2024] [Accepted: 12/13/2024] [Indexed: 12/30/2024]
Abstract
Although antiretroviral therapy (ART) has dramatically improved the outlook of the HIV/AIDS pandemic, people living with HIV (PLWH) on suppressive therapy are still at higher risk for a range of comorbidities including cardiovascular disease (CVD) and HIV-associated neurocognitive disorders (HAND), among others. Chronic inflammation and immune activation are thought to be an underlying cause of these comorbidities. Many of the factors thought to drive chronic inflammation and immune activation in HIV overlap with factors known to induce trained immunity. Trained immunity is a form of innate immune memory that metabolically and epigenetically reprograms innate immune cells to mount enhanced inflammatory responses upon secondary encounter with unrelated inflammatory stimuli. While this phenotype has been characterized in a variety of disease states in animals and humans, very little is known about its potential contribution to chronic HIV pathogenesis. In this review, a broad overview of innate immune memory in the periphery and the central nervous system (CNS) is provided and the evidence for trained immunity in the context of HIV is considered. In PLWH on ART, this phenotype could contribute to the chronic inflammation and immune activation associated with HIV comorbidities and could complicate HIV cure strategies due to the potential persistence of the phenotype after eradication of the virus. Further research into this immune state in the context of HIV may open the door for new therapeutics aimed at treating HIV comorbidities like HAND.
Collapse
Affiliation(s)
- Zachary Capriotti
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA
- Molecular and Cell Biology and Genetics Graduate Program, Drexel University College of Medicine, Philadelphia, PA, USA
| | - Zachary Klase
- Department of Pharmacology and Physiology, Drexel University College of Medicine, Philadelphia, PA, 19102, USA.
- Center for Neuroimmunology and CNS Therapeutics, Institute for Molecular Medicine and Infectious Disease, Drexel University College of Medicine, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA, 19102, USA.
| |
Collapse
|
48
|
Jarczak J, Bujko K, Ratajczak MZ, Kucia M. scRNA-seq revealed transcriptional signatures of human umbilical cord primitive stem cells and their germ lineage origin regulated by imprinted genes. Sci Rep 2024; 14:29264. [PMID: 39587190 PMCID: PMC11589151 DOI: 10.1038/s41598-024-79810-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 11/12/2024] [Indexed: 11/27/2024] Open
Abstract
A population of CD133+lin-CD45- and CD34+lin-CD45- very small embryonic-like stem cells (VSELs) has been identified in postnatal human tissues, including bone marrow (BM), mobilized peripheral blood (mPB) and umbilical cord blood (UCB). Under appropriate conditions, VSELs in vitro and in vivo differentiate into tissue-committed stem cells for all three germ layers. Molecular analysis of adult murine BM-purified VSELs revealed that these rare cells deposited during development in adult tissues (i) express a similar transcriptome as embryonic stem cells, (ii) share several markers characteristic for epiblast and migratory primordial germ cells (PGCs), (iii) highly express a polycomb group protein enhancer of zeste drosophila homolog 2 (Ezh2) and finally (iv) display a unique pattern of imprinting at crucial paternally inherited genes that promotes their quiescence. Here, by employing single-cell RNA sequencing we demonstrate for the first time that purified from UCB human VSELs defined by expression of CD34 or CD133 antigens and lack of lineage markers, including CD45 antigen express similar molecular signature as murine BM-derived VSELs. Specifically, unsupervised clustering revealed numerous subpopulations of VSELs including ones i) annotated to germline compartments, ii) regulated by parental imprinting, iii) responding to early developmental fate decisions, iv) transcription factors involved in differentiation and development, including homeobox family of genes, and v) expressing innate immunity and purinergic signaling genes.
Collapse
Affiliation(s)
- Justyna Jarczak
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
| | - Kamila Bujko
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
| | - Mariusz Z Ratajczak
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland
- Stem Cell Institute at Brown Cancer Center, University of Louisville, 500 S. Floyd Street, Rm. 107, Louisville, KY, 40202, USA
| | - Magdalena Kucia
- Laboratory of Regenerative Medicine, Center for Preclinical Studies and Technology, Medical University of Warsaw, Ul. Banacha 1B, Warsaw, Poland.
| |
Collapse
|
49
|
Antar AAR, Cox AL. Translating insights into therapies for Long Covid. Sci Transl Med 2024; 16:eado2106. [PMID: 39536116 DOI: 10.1126/scitranslmed.ado2106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Long Covid is defined by a wide range of symptoms that persist after the acute phase of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. Commonly reported symptoms include fatigue, weakness, postexertional malaise, and cognitive dysfunction, with many other symptoms reported. Symptom range, duration, and severity are highly variable and partially overlap with symptoms of myalgic encephalomyelitis/chronic fatigue syndrome and other post-acute infectious syndromes, highlighting opportunities to define shared mechanisms of pathogenesis. Potential mechanisms of Long Covid are diverse, including persistence of viral reservoirs, dysregulated immune responses, direct viral damage of tissues targeted by SARS-CoV-2, inflammation driven by reactivation of latent viral infections, vascular endothelium activation or dysfunction, and subsequent thromboinflammation, autoimmunity, metabolic derangements, microglial activation, and microbiota dysbiosis. The heterogeneity of symptoms and baseline characteristics of people with Long Covid, as well as the varying states of immunity and therapies given at the time of acute infection, have made etiologies of Long Covid difficult to determine. Here, we examine progress on preclinical models for Long Covid and review progress being made in clinical trials, highlighting the need for large human studies and further development of models to better understand Long Covid. Such studies will inform clinical trials that will define treatments to benefit those living with this condition.
Collapse
Affiliation(s)
- Annukka A R Antar
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Andrea L Cox
- Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
50
|
Lercher A, Cheong JG, Bale MJ, Jiang C, Hoffmann HH, Ashbrook AW, Lewy T, Yin YS, Quirk C, DeGrace EJ, Chiriboga L, Rosenberg BR, Josefowicz SZ, Rice CM. Antiviral innate immune memory in alveolar macrophages following SARS-CoV-2 infection ameliorates secondary influenza A virus disease. Immunity 2024; 57:2530-2546.e13. [PMID: 39353439 PMCID: PMC11563926 DOI: 10.1016/j.immuni.2024.08.018] [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: 08/02/2023] [Revised: 05/16/2024] [Accepted: 08/28/2024] [Indexed: 10/04/2024]
Abstract
Pathogen encounter can result in epigenetic remodeling that shapes disease caused by heterologous pathogens. Here, we examined innate immune memory in the context of commonly circulating respiratory viruses. Single-cell analyses of airway-resident immune cells in a disease-relevant murine model of SARS-CoV-2 recovery revealed epigenetic reprogramming in alveolar macrophages following infection. Post-COVID-19 human monocytes exhibited similar epigenetic signatures. In airway-resident macrophages, past SARS-CoV-2 infection increased activity of type I interferon (IFN-I)-related transcription factors and epigenetic poising of antiviral genes. Viral pattern recognition and canonical IFN-I signaling were required for the establishment of this innate immune memory and augmented secondary antiviral responses. Antiviral innate immune memory mounted by airway-resident macrophages post-SARS-CoV-2 was necessary and sufficient to ameliorate secondary disease caused by influenza A virus and curtailed hyperinflammatory dysregulation and mortality. Our findings provide insights into antiviral innate immune memory in the airway that may facilitate the development of broadly effective therapeutic strategies.
Collapse
Affiliation(s)
- Alexander Lercher
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| | - Jin-Gyu Cheong
- Department of Pathology and Laboratory Medicine, Laboratory of Epigenetics and Immunity, Weill Cornell Medicine, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Michael J Bale
- Department of Pathology and Laboratory Medicine, Laboratory of Epigenetics and Immunity, Weill Cornell Medicine, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY 10065, USA
| | - Chenyang Jiang
- Department of Pathology and Laboratory Medicine, Laboratory of Epigenetics and Immunity, Weill Cornell Medicine, New York, NY 10065, USA; BCMB Allied Program, Weill Cornell Graduate School of Medical Sciences, New York, NY 10065, USA
| | - Hans-Heinrich Hoffmann
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Alison W Ashbrook
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Tyler Lewy
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Yue S Yin
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Corrine Quirk
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA
| | - Emma J DeGrace
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Luis Chiriboga
- Department of Pathology, New York University Medical Center, New York, NY 10016, USA; Center for Biospecimen Research and Development, New York, NY 10016, USA
| | - Brad R Rosenberg
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Steven Z Josefowicz
- Department of Pathology and Laboratory Medicine, Laboratory of Epigenetics and Immunity, Weill Cornell Medicine, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Weill Cornell Medicine, New York, NY 10065, USA.
| | - Charles M Rice
- Laboratory of Virology and Infectious Disease, The Rockefeller University, New York, NY 10065, USA.
| |
Collapse
|