1
|
Xu X, Wang L, Sun Y, Yang C, Wang X, Guo P, Mei D. Unveiling the differences: infection disorders associated with tumor necrosis factor α inhibitors in pediatric patients-a pharmacovigilance study (2004-2023). Eur J Pediatr 2025; 184:324. [PMID: 40317305 DOI: 10.1007/s00431-025-06152-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/15/2025] [Accepted: 04/23/2025] [Indexed: 05/07/2025]
Abstract
The increasing use of tumor necrosis factor inhibitors (TNFi) in pediatric patients has raised concerns about their potential impact on the immune system and related adverse events. Infection-related adverse events (AEs) caused by TNFi have already raised widespread concerns in real-world settings. This study aims to comprehensively analyze and summarize the infection-related AEs associated with TNFi in pediatric patients. A retrospective pharmacovigilance study was conducted to identify cases of TNFi-related infections reported to the FDA Adverse Event Reporting System (FAERS) database between Q1 2004 and Q1 2023. TNFi reports were carefully reviewed to exclude confounding factors like other AEs, concomitant medications, and prescription indications. Proportionality analysis was conducted by comparing TNFi reports to the entire FAERS database to identify infection-related AEs significantly associated with TNFi use. Infection-related AEs accounted for 8.36% of all TNFi-related adverse event reports in the FAERS database. A total of 8050 cases of TNFi-associated infections were identified in the pediatric population, with 2.57% of reports resulting in fatalities. Infliximab and golimumab showed a stronger association with infection-related AEs compared to other TNFi. Notably, only adalimumab shows a lower risk of viral infections, while it exhibits an increased risk of bacterial and mycobacterial infections, similar to other TNFi. CONCLUSIONS This study identified a significant association between TNFi use and infection-related AEs in pediatric patients, providing the foothold for further research. However, due to its retrospective nature, further investigations are warranted to confirm these findings and identify potential risk factors in a controlled, prospective study setting. WHAT IS KNOWN • There is sufficient evidence to demonstrate the infection risk associated with TNFi in adult patients. • Pediatric patients, whose immune systems are still developing, are more vulnerable to certain infections. WHAT IS NEW • There is a significant association between TNFi use and infection-related adverse events in pediatric patients, and different TNFi have distinct infection profiles.
Collapse
Affiliation(s)
- Xiaolin Xu
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Luquan Wang
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Yixin Sun
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Changqing Yang
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiaoling Wang
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China
| | - Peng Guo
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
| | - Dong Mei
- Department of Pharmacy, Beijing Children's Hospital, National Center for Children's Health, Capital Medical University, Beijing, 100045, China.
| |
Collapse
|
2
|
Athina P, Stavroula L, Despoina Zoe MT, Stavroula K, Maria KG. The Clinical Burden of SARS-CoV-2 Compared to Influenza A in the Same Cohort of Paediatric Outpatients; Increased Influenza Severity and Potential Insights. J Paediatr Child Health 2025; 61:734-740. [PMID: 39935033 DOI: 10.1111/jpc.70014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/31/2025] [Accepted: 02/02/2025] [Indexed: 02/13/2025]
Abstract
INTRODUCTION The variation in the clinical severity of viral infections remains a matter of scientific debate. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and Influenza A virus (IAV) are both RNA viruses, exhibiting a range of clinical presentations, ranging from asymptomatic cases to fatalities, influenced by both viral and host factors. A descriptive real-life study was conducted, aiming to compare the clinical manifestations of recent COVID-19 with those of IAV infection in the same group of non-hospitalised, unvaccinated children. MATERIALS AND METHOD Between September 2021 and February 2023, we evaluated 115 individuals (male 48.7%, mean age 101.8 ± 50.8 months) who had not received vaccination for both SARS-CoV-2 and the quadrivalent influenza vaccine and were documented to be infected with both viruses within a 12-month time interval. The visual analogue scale (VAS-5) was used to assess parents'/patients' evaluation comparing the two infections. RESULTS In cases of IAV infection, a significantly higher prevalence of prolonged high fever, upper and lower respiratory symptoms, and secondary infections was observed. The majority (74.8%) of patients and/or parents assessed IAV as a more severe clinical syndrome compared with SARS-CoV-2. CONCLUSIONS The clinical burden of IAV was found to be greater than that of SARS-CoV-2 infection in the same unvaccinated children. This variation in clinical severity may provide a reference for effective vaccination policies; however, underlying mechanisms that could explain this difference require further evaluation.
Collapse
Affiliation(s)
| | | | | | | | - Kanariou G Maria
- Pediatric Immunology Unit, IASO Children's Hospital, Athens, Greece
- Department of Immunology-Histocompatibility, Agia Sophia Children Hospital Athens, Greece
| |
Collapse
|
3
|
Balla J, Rathore APS, St. John AL. Maternal IgE Influence on Fetal and Infant Health. Immunol Rev 2025; 331:e70029. [PMID: 40281548 PMCID: PMC12032057 DOI: 10.1111/imr.70029] [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: 03/24/2025] [Accepted: 04/07/2025] [Indexed: 04/29/2025]
Abstract
Immunoglobulin E (IgE) is the most recently discovered and evolved mammalian antibody type, best known for interacting with mast cells (MCs) as immune effectors. IgE-mediated antigen sensing by MC provides protection from parasites, venomous animals, bacteria, and other insults to barrier tissues exposed to the environment. IgE and MCs act as inflammation amplifiers and immune response adjuvants. Thus, IgE production and memory formation are greatly constrained and require specific licensing. Failure of regulation gives rise to allergic disease, one of the top causes of chronic illness. Increasing evidence suggests allergy development often starts early in life, including prenatally, with maternal influence being central in shaping the offspring's immune system. Although IgE often exists before birth, an endogenous source of IgE-producing B cells has not been identified. This review discusses the mechanisms of maternal IgE transfer into the offspring, its interactions with offspring MCs and antigen-presenting cells, and the consequences for allergic inflammation and allergen sensitization development. We discuss the multifaceted effects of pre-existing IgG, IgE, and their glycosylation on maternal IgE transfer and functionality in the progeny. Understanding the IgE-mediated mechanisms predisposing for early life allergy development may allow their targeting with existing therapeutics and guide the development of new ones.
Collapse
Affiliation(s)
- Jozef Balla
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
| | - Abhay P. S. Rathore
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
| | - Ashley L. St. John
- Programme in Emerging Infectious DiseasesDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Department of PathologyDuke University Medical CenterDurhamNorth CarolinaUSA
- Department of Microbiology and Immunology, Yong Loo Lin School of MedicineNational University of SingaporeSingaporeSingapore
- SingHealth Duke‐NUS Global Health InstituteSingaporeSingapore
| |
Collapse
|
4
|
Day-Lewis ME, Berbert L, DeGrazia M, Yee C, Fried AJ, Nguyen AA, Hale JE, Counihan A, Comeau AM, Treffeisen ER, Reed MP, Platt CD, Chou J. Incidence and risk factors for CMV and EBV infection in infants with low T-cell receptor excision circles on newborn screen. Clin Immunol 2025; 277:110510. [PMID: 40316158 DOI: 10.1016/j.clim.2025.110510] [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: 02/13/2025] [Revised: 04/22/2025] [Accepted: 04/25/2025] [Indexed: 05/04/2025]
Abstract
BACKGROUND Newborn screening for severe combined immunodeficiency (SCID) using T cell receptor excision circles (TRECs) identifies patients with other causes of lymphopenia. The risk of opportunistic infection in patients with non-SCID lymphopenia is poorly understood. We aim to describe incidence and risk factors associated with cytomegalovirus (CMV) and Epstein-Barr virus (EBV) infection in patients with low TRECs. METHODS This retrospective study analyzed 289 patients with ≥1 abnormal TREC result. RESULTS Nineteen patients had CMV or EBV detected by PCR. Most had resolution of infection (n = 13). Two have chronic viremia, and four expired due to disseminated CMV. Risk factors included undetectable TRECs, consanguinity, family history, low NK, naïve CD4, naïve CD8 cells, and phytohemagglutinin. CONCLUSION Infection with CMV and EBV in patients with low TRECs is rare, however some may benefit from preventative measures. Consideration of risk factors may aid in decision-making and improve outcomes.
Collapse
Affiliation(s)
- Megan E Day-Lewis
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA.
| | - Laura Berbert
- Institutional Centers for Clinical and Translational Research, Boston Children's Hospital, Boston, MA, USA
| | - Michele DeGrazia
- Cardiovascular and Critical Care Services, Boston Children's Hospital, Boston, MA, USA; Division of Neonatology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Christina Yee
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Ari J Fried
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Jaime E Hale
- New England Newborn Screening Program, ForHealth Consulting, UMass Chan Medical School, Worcester, MA, USA
| | - Anne Counihan
- New England Newborn Screening Program, ForHealth Consulting, UMass Chan Medical School, Worcester, MA, USA
| | - Anne Marie Comeau
- New England Newborn Screening Program, ForHealth Consulting, UMass Chan Medical School, Worcester, MA, USA; Department of Pediatrics, T.H. Chan School of Medicine, Worcester, MA, USA
| | - Elsa R Treffeisen
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Mary Poyner Reed
- Medicine Patient Services, Boston Children's Hospital, Boston, MA, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Ethridge AD, Yagi K, Martínez LA, Rasky AJ, Morris SB, Falkowski NR, Huffnagle GB, Lukacs NW. RSV infection in neonatal mice and gastrointestinal microbiome alteration contribute to allergic predisposition. Mucosal Immunol 2025:S1933-0219(25)00045-5. [PMID: 40280490 DOI: 10.1016/j.mucimm.2025.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/18/2025] [Accepted: 04/22/2025] [Indexed: 04/29/2025]
Abstract
Severe respiratory syncytial virus (RSV) infection during infancy is associated with a 2 to 4-fold increased risk for the development of wheezing and asthma. Recent studies have implicated microbiome changes, either within the lung or gut, during early life can also affect the development of pulmonary disease. Our studies demonstrate long-term gastrointestinal and lung microbiome changes following early life (EL) RSV infection. To determine the respective roles of ELRSV infection and the gut microbiome, we performed germ-free neonatal infection and microbiome colonization using a microbiome from an uninfected animal followed by cockroach allergen (CRA)-induced asthma 4 weeks later. Germ-free animals with ELRSV infection displayed increased airway disease that was diminished by microbiome colonization, including airway hyperreactivity (AHR), mucus, and eosinophil infiltration. To address the role of virus induced gastrointestinal microbiome alterations, we utilized GF mice conventionalized with RSV-associated or naive microbiomes followed by CRA-induced disease. Transfer of neonatal microbiome taken during acute RSV infection did not alter the allergic response to CRA. However, the transfer of a naive adult microbiome conferred protection from enhanced AHR in response to CRA whereas an RSV associated microbiome did not. Taken together, our data indicate that microbiome alteration and early life RSV infection both contribute to allergic predisposition.
Collapse
Affiliation(s)
- Alexander D Ethridge
- Immunology Graduate Program, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Kazuma Yagi
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA.
| | | | - Andrew J Rasky
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Susan B Morris
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Nicole R Falkowski
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| | - Gary B Huffnagle
- Immunology Graduate Program, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA; Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA
| | - Nicholas W Lukacs
- Immunology Graduate Program, Rackham Graduate School, University of Michigan, Ann Arbor, MI, USA; Department of Pathology, University of Michigan, Ann Arbor, MI, USA; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, USA; Mary H. Weiser Food Allergy Center, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
6
|
Gardner PN, Hangoma J, Sialubanje C, Chipoya M, Lamba L, Mwenechanya M, Chilyabanyama R, Kasonde M, Simwaba D, Kapina M, Young S, Mwangilwa K, Chilengi R, Fwemba I. Factors associated with measles vaccine immunogenicity in children at University Teaching Hospitals, Lusaka, Zambia. PLOS GLOBAL PUBLIC HEALTH 2025; 5:e0003954. [PMID: 40267112 PMCID: PMC12017566 DOI: 10.1371/journal.pgph.0003954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 03/12/2025] [Indexed: 04/25/2025]
Abstract
Measles poses a significant global public health challenge, particularly in low-resource settings where vaccination coverage is limited. This study examined factors associated with measles vaccine immunogenicity in children aged 2-15 years at the University Teaching Children's Hospital and the HIV Pediatric Centre of Excellence in Lusaka, Zambia. This comparative analytical cross-sectional study was conducted from April to July 2024, enrolling 200 children, including 100 HIV-positive and 100 HIV-negative participants. All children had received at least two doses of a measles-containing vaccine and had no history of measles infection in the past six months. Blood samples were analyzed for measles immunity, while data on age, HIV status, breastfeeding history, and socio-demographic factors were collected. Among HIV-negative children, 75% retained immunity, whereas only 38% of HIV-positive children retained immunity. Multivariate logistic regression showed that children aged 10-15 years were less likely to retain immunity compared to those aged 2-4 years (AOR = 0.270, 95% CI [0.114-0.618], p = 0.002). HIV-positive children had lower odds of retaining immunity compared to HIV-negative children (AOR = 0.290, 95% CI [0.137-0.594], p < 0.001). Breastfed children had higher immunity retention (AOR = 0.336, 95% CI [0.147-0.738], p = 0.007) compared to non-breastfed children. Residing in Lusaka was associated with lower immunity retention (AOR = 0.250, 95% CI [0.066-0.859], p = 0.031). These findings highlight the protective role of breastfeeding and suggest that older and HIV-infected children may benefit from booster doses to sustain measles immunity.
Collapse
Affiliation(s)
- Priscilla Nkonde Gardner
- University of Zambia, Institute of Distance Education Lusaka, Zambia
- Zambia National Public Health Institute, Lusaka, Zambia
| | - Jimmy Hangoma
- University of Zambia, Institute of Distance Education Lusaka, Zambia
- School of Health Sciences, Levy Mwanawasa Medical University, Lusaka, Zambia
| | - Cephas Sialubanje
- Zambia National Public Health Institute, Lusaka, Zambia
- School of Public Health, Levy Mwanawasa Medical University, Lusaka, Zambia
| | | | - Lillian Lamba
- Zambia National Public Health Institute, Lusaka, Zambia
| | - Musaku Mwenechanya
- Department of Child Health, University Teaching Children’s Hospital, Lusaka, Zambia
| | | | | | - Davie Simwaba
- University of Zambia, Institute of Distance Education Lusaka, Zambia
| | - Muzala Kapina
- Zambia National Public Health Institute, Lusaka, Zambia
| | - Soo Young
- Health Programs, Churches Health Association of Zambia, Lusaka, Zambia
| | | | - Roma Chilengi
- Zambia National Public Health Institute, Lusaka, Zambia
| | - Isaac Fwemba
- University of Zambia, Institute of Distance Education Lusaka, Zambia
| |
Collapse
|
7
|
Soriano-Arandes A, Andrés C, Perramon-Malavez A, Creus-Costa A, Gatell A, Martín-Martín R, Solà-Segura E, Riera-Bosch MT, Fernández E, Biosca M, Capdevila R, Sánchez A, Soler I, Chiné M, Sanz L, Quezada G, Pérez S, Canadell D, Salvadó O, Ridao M, Sau I, Rifà MÀ, Macià E, Burgaya-Subirana S, Vila M, Vila J, Mejías A, Antón A, Soler-Palacin P, Prats C. Implementing Symptom-Based Predictive Models for Early Diagnosis of Pediatric Respiratory Viral Infections. Viruses 2025; 17:546. [PMID: 40284989 PMCID: PMC12031125 DOI: 10.3390/v17040546] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
(1) Background: Respiratory viral infections, including those caused by SARS-CoV-2, respiratory syncytial virus (RSV), influenza viruses, rhinovirus, and adenovirus, are major causes of acute respiratory infections (ARIs) in children. Symptom-based predictive models are valuable tools for expediting diagnoses, particularly in primary care settings. This study assessed the effectiveness of machine learning-based models in estimating infection probabilities for these common pediatric respiratory viruses, using symptom data. (2) Methods: Data were collected from 868 children with ARI symptoms evaluated across 14 primary care centers, members of COPEDICAT (Coronavirus Pediatria Catalunya), from October 2021 to October 2023. Random forest and boosting models with 10-fold cross-validation were used, applying SMOTE-NC to address class imbalance. Model performance was evaluated via area under the curve (AUC), sensitivity, specificity, and Shapley additive explanations (SHAP) values for feature importance. (3) Results: The model performed better for RSV (AUC: 0.81, sensitivity: 0.64, specificity: 0.77) and influenza viruses (AUC: 0.71, sensitivity: 0.70, specificity: 0.59) and effectively ruled out SARS-CoV-2 based on symptom absence, such as crackles and wheezing. Predictive performance was lower for non-enveloped viruses like rhinovirus and adenovirus, due to their nonspecific symptom profiles. SHAP analysis identified key symptoms patterns for each virus. (4) Conclusions: The study demonstrated that symptom-based predictive models effectively identify pediatric respiratory infections, with notable accuracy for those caused by RSV, SARS-CoV-2, and influenza viruses.
Collapse
Affiliation(s)
- Antoni Soriano-Arandes
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Children’s Hospital, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain;
- Infection and Immunity in Pediatric Patients, Vall d’Hebron Research Institute, 08035 Barcelona, Spain;
| | - Cristina Andrés
- Respiratory Virus Unit, Microbiology Department, Vall d′Hebron Research Institute, Vall d′Hebron University Hospital, Vall d′Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (A.A.)
| | - Aida Perramon-Malavez
- Computational Biology and Complex Systems (BIOCOM-SC) Group, Department of Physics, Barcelona School of Agri-Food and Biosystems Engineering, Universitat Politècnica de Catalunya, 08860 Castelldefels, Spain; (A.P.-M.); (C.P.)
| | - Anna Creus-Costa
- Infection and Immunity in Pediatric Patients, Vall d’Hebron Research Institute, 08035 Barcelona, Spain;
- Department of Pediatrics, Children’s Hospital, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (A.C.-C.); (J.V.)
| | - Anna Gatell
- Equip Pediatria Territorial Garraf, 08812 Barcelona, Spain;
| | | | | | | | - Eduard Fernández
- EAP Vic Nord, Vic, 08500 Barcelona, Spain; (E.S.-S.); (M.T.R.-B.); (E.F.)
| | - Mireia Biosca
- EAP Les Borges Blanques, 25400 Lleida, Spain; (M.B.); (R.C.)
| | - Ramon Capdevila
- EAP Les Borges Blanques, 25400 Lleida, Spain; (M.B.); (R.C.)
| | | | | | | | | | - Gabriela Quezada
- CAP Marià Fortuny, Reus, 43205 Tarragona, Spain; (R.M.-M.); (G.Q.)
| | - Sandra Pérez
- CAP Barberà del Vallès, 08210 Barcelona, Spain; (S.P.); (D.C.)
| | - Dolors Canadell
- CAP Barberà del Vallès, 08210 Barcelona, Spain; (S.P.); (D.C.)
| | | | - Marisa Ridao
- EAP Sant Vicenç dels Horts, 08620 Barcelona, Spain;
| | - Imma Sau
- EAP Santa Coloma de Farners, 17430 Girona, Spain;
| | | | | | | | | | - Jorgina Vila
- Infection and Immunity in Pediatric Patients, Vall d’Hebron Research Institute, 08035 Barcelona, Spain;
- Department of Pediatrics, Children’s Hospital, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (A.C.-C.); (J.V.)
| | - Asunción Mejías
- Department of Infectious Diseases, Diseases, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA;
| | - Andrés Antón
- Respiratory Virus Unit, Microbiology Department, Vall d′Hebron Research Institute, Vall d′Hebron University Hospital, Vall d′Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain; (C.A.); (A.A.)
| | - Pere Soler-Palacin
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Children’s Hospital, Vall d’Hebron Barcelona Hospital Campus, 08035 Barcelona, Spain;
- Infection and Immunity in Pediatric Patients, Vall d’Hebron Research Institute, 08035 Barcelona, Spain;
| | - Clara Prats
- Computational Biology and Complex Systems (BIOCOM-SC) Group, Department of Physics, Barcelona School of Agri-Food and Biosystems Engineering, Universitat Politècnica de Catalunya, 08860 Castelldefels, Spain; (A.P.-M.); (C.P.)
| |
Collapse
|
8
|
Lukyanov DK, Kriukova VV, Ladell K, Shagina IA, Staroverov DB, Minasian BE, Fedosova AS, Shelyakin P, Suchalko ON, Komkov AY, Blagodatskikh KA, Miners KL, Britanova OV, Franke A, Price DA, Chudakov DM. Repertoire-based mapping and time-tracking of T helper cell subsets in scRNA-Seq. Front Immunol 2025; 16:1536302. [PMID: 40255395 PMCID: PMC12006041 DOI: 10.3389/fimmu.2025.1536302] [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: 11/28/2024] [Accepted: 02/21/2025] [Indexed: 04/22/2025] Open
Abstract
Introduction The functional programs of CD4+ T helper (Th) cell clones play a central role in shaping immune responses to different challenges. While advances in single-cell RNA sequencing (scRNA-Seq) have significantly improved our understanding of the diversity of Th cells, the relationship between scRNA-Seq clusters and the traditionally characterized Th subsets remains ambiguous. Methods In this study, we introduce TCR-Track, a method leveraging immune repertoire data to map phenotypically sorted Th subsets onto scRNA-Seq profiles. Results and discussion This approach accurately positions the Th1, Th1-17, Th17, Th22, Th2a, Th2, T follicular helper (Tfh), and regulatory T-cell (Treg) subsets, outperforming mapping based on CITE-Seq. Remarkably, the mapping is tightly focused on specific scRNA-Seq clusters, despite 4-year interval between subset sorting and the effector CD4+ scRNA-Seq experiment. These findings highlight the intrinsic program stability of Th clones circulating in peripheral blood. Repertoire overlap analysis at the scRNA-Seq level confirms that the circulating Th1, Th2, Th2a, Th17, Th22, and Treg subsets are clonally independent. However, a significant clonal overlap between the Th1 and cytotoxic CD4+ T-cell clusters suggests that cytotoxic CD4+ T cells differentiate from Th1 clones. In addition, this study resolves a longstanding ambiguity: we demonstrate that, while CCR10+ Th cells align with a specific Th22 scRNA-Seq cluster, CCR10-CCR6+CXCR3-CCR4+ cells, typically classified as Th17, represent a mixture of bona fide Th17 cells and clonally unrelated CCR10low Th22 cells. The clear distinction between the Th17 and Th22 subsets should influence the development of vaccine- and T-cell-based therapies. Furthermore, we show that severe acute SARS-CoV-2 infection induces systemic type 1 interferon (IFN) activation of naive Th cells. An increased proportion of effector IFN-induced Th cells is associated with a moderate course of the disease but remains low in critical COVID-19 cases. Using integrated scRNA-Seq, TCR-Track, and CITE-Seq data from 122 donors, we provide a comprehensive Th scRNA-Seq reference that should facilitate further investigation of Th subsets in fundamental and clinical studies.
Collapse
Affiliation(s)
- Daniil K. Lukyanov
- Center for Molecular and Cellular Biology, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
| | | | - Kristin Ladell
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Irina A. Shagina
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Dmitry B. Staroverov
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | | | | | - Pavel Shelyakin
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
| | | | | | | | - Kelly L. Miners
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Olga V. Britanova
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
| | - Andre Franke
- Institute of Clinical Molecular Biology, Kiel University, Kiel, Germany
| | - David A. Price
- Division of Infection and Immunity, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
- Systems Immunity Research Institute, Cardiff University School of Medicine, University Hospital of Wales, Cardiff, United Kingdom
| | - Dmitry M. Chudakov
- Center for Molecular and Cellular Biology, Moscow, Russia
- Genomics of Adaptive Immunity Department, Institute of Bioorganic Chemistry, Moscow, Russia
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Abu Dhabi Stem Cell Center, Al Muntazah, United Arab Emirates
- Department of Molecular Medicine, Central European Institute of Technology, Brno, Czechia
| |
Collapse
|
9
|
Lewinsohn DA, Kain D, Awad W, McElfresh GW, Cansler M, Swarbrick G, Poa K, McNeice C, Boggy G, Rott K, Null M, Lewinsohn D, Rossjohn J, Bimber B. Human Neonatal MR1T Cells Have Diverse TCR Usage, are Less Cytotoxic and are Unable to Respond to Many Common Childhood Pathogens. RESEARCH SQUARE 2025:rs.3.rs-6265058. [PMID: 40235492 PMCID: PMC11998791 DOI: 10.21203/rs.3.rs-6265058/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
Neonatal sepsis is a leading cause of childhood mortality. Understanding immune cell development can inform strategies to combat this. MR1-restricted T (MR1T) cells can be defined by their recognition of small molecules derived from microbes, self, and drug and drug-like molecules, presented by the MHC class 1-related molecule (MR1). In healthy adults, the majority of MR1T cells express an invariant α-chain; TRAV1-2/TRAJ33/12/20 and are referred to as mucosal-associated invariant T (MAIT) cells. Neonatal MR1T cells isolated from cord blood (CB) demonstrate more diversity in MR1T TCR usage, with the majority of MR1-5-OP-RU-tetramer(+) cells being TRAV1-2(-). To better understand this diversity, we performed single-cell-RNA-seq/TCR-seq (scRNA-seq/scTCR-seq) on MR1-5-OP-RU-tetramer(+) cells from CB (n=5) and adult participants (n=5). CB-derived MR1T cells demonstrate a less cytotoxic/pro-inflammatory phenotype, and a more diverse TCR repertoire. A panel of CB and adult MAIT and TRAV1-2(-) MR1T cell clones were generated, and CB-derived clones were unable to recognize several common riboflavin-producing childhood pathogens (S. aureus, S. pneumoniae, M. tuberculosis). Biochemical and structural investigation of one CB MAIT TCR (CB964 A2; TRAV1-2/TRBV6-2) showed a reduction in binding affinity toward the canonical MR1-antigen, 5-OP-RU, compared to adult MAIT TCRs that correlated with differences in β-chain contribution in the TCR-MR1 interface. Overall, this data shows that CB MAIT and TRAV1-2(-) MR1T cells, express a diverse TCR repertoire, a more restricted childhood pathogen recognition profile and diminished cytotoxic and pro-inflammatory capacity. Understanding this diversity, along with the functional ability of TRAV1-2(-) MR1T cells, could provide insight into increased neonatal susceptibility to infections.
Collapse
|
10
|
Kain D, Awad W, McElfresh GW, Cansler M, Swarbrick GM, Poa KCY, McNeice C, Boggy G, Rott K, Null MD, Lewinsohn DM, Rossjohn J, Bimber BN, Lewinsohn DA. Human Neonatal MR1T Cells Have Diverse TCR Usage, are Less Cytotoxic and are Unable to Respond to Many Common Childhood Pathogens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.17.643805. [PMID: 40166301 PMCID: PMC11956999 DOI: 10.1101/2025.03.17.643805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Neonatal sepsis is a leading cause of childhood mortality. Understanding immune cell development can inform strategies to combat this. MR1-restricted T (MR1T) cells can be defined by their recognition of small molecules derived from microbes, self, and drug and drug-like molecules, presented by the MHC class 1-related molecule (MR1). In healthy adults, the majority of MR1T cells express an invariant α-chain; TRAV1-2/TRAJ33/12/20 and are referred to as mucosal-associated invariant T (MAIT) cells. Neonatal MR1T cells isolated from cord blood (CB) demonstrate more diversity in MR1T TCR usage, with the majority of MR1-5-OP-RU-tetramer(+) cells being TRAV1-2(-). To better understand this diversity, we performed single-cell-RNA-seq/TCR-seq (scRNA-seq/scTCR-seq) on MR1-5-OP-RU-tetramer(+) cells from CB (n=5) and adult participants (n=5). CB-derived MR1T cells demonstrate a less cytotoxic/pro-inflammatory phenotype, and a more diverse TCR repertoire. A panel of CB and adult MAIT and TRAV1-2(-) MR1T cell clones were generated, and CB-derived clones were unable to recognize several common riboflavin-producing childhood pathogens (S. aureus, S. pneumoniae, M. tuberculosis). Biochemical and structural investigation of one CB MAIT TCR (CB964 A2; TRAV1-2/TRBV6-2) showed a reduction in binding affinity toward the canonical MR1-antigen, 5-OP-RU, compared to adult MAIT TCRs that correlated with differences in β-chain contribution in the TCR-MR1 interface. Overall, this data shows that CB MAIT and TRAV1-2(-) MR1T cells, express a diverse TCR repertoire, a more restricted childhood pathogen recognition profile and diminished cytotoxic and pro-inflammatory capacity. Understanding this diversity, along with the functional ability of TRAV1-2(-) MR1T cells, could provide insight into increased neonatal susceptibility to infections.
Collapse
Affiliation(s)
- Dylan Kain
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
- Division of Infectious Diseases,Department of Medicine, University of Toronto, Toronto, ON, Canada
| | - Wael Awad
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - G W McElfresh
- Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
| | - Meghan Cansler
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Gwendolyn M Swarbrick
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Kean Chan Yew Poa
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Conor McNeice
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Gregory Boggy
- Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
| | - Katherine Rott
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - Megan D Null
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| | - David M Lewinsohn
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jamie Rossjohn
- Infection and Immunity Program and Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
- Institute of Infection and Immunity, Cardiff University, School of Medicine, Heath Park, Cardiff, UK
| | - Benjamin N Bimber
- Oregon National Primate Research Center, OHSU, Beaverton, OR, USA
- Vaccine and Gene Therapy Institute, OHSU, Beaverton, OR, USA
| | - Deborah A Lewinsohn
- Division of Infectious Diseases, Department of Pediatrics, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
11
|
Silinskaite U, Valciukiene J, Jakubauskas M, Poskus T. The Immune Environment in Colorectal Adenoma: A Systematic Review. Biomedicines 2025; 13:699. [PMID: 40149674 PMCID: PMC11940254 DOI: 10.3390/biomedicines13030699] [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: 02/10/2025] [Accepted: 02/28/2025] [Indexed: 03/29/2025] Open
Abstract
Background/Objectives: Research on colorectal adenoma is significantly less comprehensive compared to studies on colorectal carcinoma. Although colorectal adenoma is a precursor of the majority of sporadic colorectal cancers, not all adenomas develop into carcinomas. The complex interaction of immune responses in the premalignant tumor microenvironment might be a factor for that. Methods: In this systematic review, we aim to provide a thorough analysis of the current research examining the immune infiltration patterns in sporadic colorectal adenoma tissues in the context of immune cell-based, cytokine-based, and other immunological factor-related changes along the conventional adenoma-carcinoma sequence. The articles included in the review extend up to December 2024 in PubMed and Web of Science databases. Results: Most included studies have shown significant differences in immune cell counts, densities, and cytokine expression levels associated with premalignant colorectal lesions (and/or colorectal cancer). No consensus on the immune-related tendencies concerning CD4+T cells and CD8+T cells was reached. Decreasing expression of mDCs and plasma and naïve B cells were detected along the ACS. The increased density of tissue eosinophils in the adenoma tissue dramatically diminishes after the transition to carcinoma. As the adenoma progresses, the increasing expression of IL-1α, IL-4, IL-6, IL-8, IL-10, IL-17A, IL-21, IL-23, IL-33, and TGF-β and decreasing levels of IL-12A, IL-18, IFN-γ, and TNFα cytokines in the invasive carcinoma stage is being detected. The over-expression of COX-2, PD-1/PD-L1, CTLA-4, and ICOS/ICOSLG in the colorectal adenomatous and cancerous tissues was also observed. Conclusions: Further studies are needed for a better understanding of the whole picture of colorectal adenoma-associated immunity and its impact on precancerous lesion's potential to progress.
Collapse
|
12
|
Zhou X, Xue J, Zhang Y, Xia R, Shan Z, Zhang L, Gui M, Liu G, Chen Z. Evolution of Biological Hydroxyapatite Modification Strategy: Anti-Inflammation Approach Rescues the Calcium-NOD-Like Receptor-Inflammation Axis-Mediated Periodontal Redevelopment Failure. Biomater Res 2025; 29:0131. [PMID: 40012607 PMCID: PMC11862812 DOI: 10.34133/bmr.0131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Revised: 11/02/2024] [Accepted: 12/14/2024] [Indexed: 02/28/2025] Open
Abstract
Periodontal regenerative medicine is currently undergoing a paradigm shift from dissecting the healing process toward utilization of the developmental program. Biological hydroxyapatite (BHA), a major component of guided tissue regeneration, has long been optimized for inducing multidirectional differentiation of periodontal ligament cells (PDLCs). However, this approach runs counter to the redevelopment strategy. Thus, the conventional BHA should evolve to induce the redevelopment process of periodontal tissue. In this study, histopathological changes and immune microenvironment characteristics of the periodontal developmental process, natural healing process (Blank group), and BHA-mediated healing process (BHA group) were compared to evaluate the main manifestations of BHA-mediated periodontal "developmental engineering" outcome. Our results suggested that neither the Blank nor BHA group could recur key events in periodontal development. The implantation of BHA led to pro-inflammatory immune microenvironment and an unstable blood fibrin clot structure, which facilitated the invasion of outer gingival fibroblasts, consequently disrupting redevelopmental events. High-throughput chip technology was further used to explore the underlying mechanism of immune activation, revealing that the calcium-NOD-like receptor-inflammation axis signaling axis promoted the activation of pro-inflammatory immune response that contributed to redevelopment failure. An immunomodulatory cytokine interleukin 4 (IL4)-modified BHA was used to further validate the efficacy of developmental engineering strategy. IL4 could partially rescued the redevelopment failure through immunosuppression. This study presented an innovative strategy for the development of advanced periodontal regenerative materials and offered a potential approach for the application of development-inspired periodontal tissue engineering strategies. It represented a marked advancement in the development of regenerative medicine and propelled the clinical organ restoration forward.
Collapse
Affiliation(s)
- Xuan Zhou
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Junlong Xue
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Yanshu Zhang
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Ruidi Xia
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Zhengjie Shan
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Linjun Zhang
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Mixiao Gui
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Guanqi Liu
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| | - Zetao Chen
- Hospital of Stomatology,
Guanghua School of Stomatology, Sun Yat-sen University, and Guangdong Provincial Key Laboratory of Stomatology, Guangzhou 510055, China
- Guangdong Research Center for Dental and Cranial Rehabilitation and Material Engineering, Guangzhou 510055, China
| |
Collapse
|
13
|
Mai KL, Pan WQ, Lin ZS, Wang Y, Yang ZF. Pathogenesis of influenza and SARS-CoV-2 co-infection at the extremes of age: decipher the ominous tales of immune vulnerability. ADVANCED BIOTECHNOLOGY 2025; 3:5. [PMID: 39883362 DOI: 10.1007/s44307-025-00057-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/18/2024] [Accepted: 01/09/2025] [Indexed: 01/31/2025]
Abstract
The co-circulation of influenza and SARS-CoV-2 has led to co-infection events, primarily affecting children and older adults, who are at higher risk for severe disease. Although co-infection prevalence is relatively low, it is associated with worse outcomes compared to mono-infections. Previous studies have shown that the outcomes of co-infection depend on multiple factors, including viral interference, virus-host interaction and host response. Children and the elderly exhibit distinct patterns of antiviral response, which involve airway epithelium, mucociliary clearance, innate and adaptive immune cells, and inflammatory mediators. This review explores the pathogeneses of SARS-CoV-2 and influenza co-infection, focusing on the antiviral responses in children and the elderly. By comparing immature immunity in children and immune senescence in older adults, we aim to provide insights for the clinical management of severe co-infection cases.
Collapse
Affiliation(s)
- Kai-Lin Mai
- Henan University College of Medicine, Kaifeng, 475004, China
- School of Life Sciences, Henan University, Kaifeng, 475004, China
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Wei-Qi Pan
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Zheng-Shi Lin
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China
| | - Yang Wang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| | - Zi-Feng Yang
- National Clinical Research Center for Respiratory Disease, State Key Laboratory of Respiratory Disease, Guangzhou Institute of Respiratory Health, the First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, China.
- Guangzhou National Laboratory, Guangzhou, 510005, China.
| |
Collapse
|
14
|
Tucker MH, Kalamvoki M, Tilak K, Raje N, Sampath V. The immunogenetic basis of severe herpes simplex infections in neonates and children: a review. Pediatr Res 2025:10.1038/s41390-025-03830-7. [PMID: 39827257 DOI: 10.1038/s41390-025-03830-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 12/05/2024] [Accepted: 12/09/2024] [Indexed: 01/22/2025]
Abstract
Human herpes simplex virus (HSV) is a double stranded DNA virus with two distinct types, HSV-1 and HSV-2. The global burden of HSV is high with an estimated 2/3 of the adult population seropositive for at least one of these types of HSV. HSV rarely causes life-threatening disease in immunocompetent children and adults. However, in neonates and children with a developmentally immature immune system it can cause disseminated disease and herpes simplex encephalitis (HSE). Recent studies in children and neonates suggest that genetic risk-factors can contribute to severe HSV phenotypes in neonates and children. In particular, genetic defects in the Toll Like Receptor 3 (TLR3) signaling pathway that attenuate the type I interferon response to HSV are being increasingly recognized in children with severe phenotypes of HSV. In this review, we discuss the epidemiology and immunological aspects of HSV disease in neonates and children and provide an in-depth review of the studies characterizing the role of inborn errors in the TLR3 pathway and other immune genes in HSV. We highlight the need for future research to identify the immunogenetic basis of severe or recurrent HSV disease in neonates and children. IMPACT: Review the epidemiology and phenotypes of herpes simplex virus (HSV) infection in neonates and children. Discuss the mechanisms of immunity against HSV highlighting the developmental vulnerability of neonates and infants to severe HSV disease. Explore in depth the genes and immune pathways that underlie genetic predisposition to severe HSV disease in neonates and children, and outline strategies for multi-disciplinary clinical evaluation of severe disease.
Collapse
Affiliation(s)
- Megan H Tucker
- Department of Neonatology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA.
| | - Maria Kalamvoki
- Department of Microbiology, Molecular Genetics and Immunology, University of Kansas Medical Center, Kansas City, KS, USA
| | - Kedar Tilak
- Department of Neonatology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Nikita Raje
- Department of Allergy and Immunology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Venkatesh Sampath
- Department of Neonatology, Children's Mercy Kansas City, University of Missouri-Kansas City, Kansas City, MO, USA
| |
Collapse
|
15
|
Kang N, Subramanian VS, Agrawal A. Influence of Aging and Immune Alterations on Susceptibility to Pneumococcal Pneumonia in the Elderly. Pathogens 2025; 14:41. [PMID: 39861002 PMCID: PMC11768109 DOI: 10.3390/pathogens14010041] [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: 12/11/2024] [Revised: 01/04/2025] [Accepted: 01/06/2025] [Indexed: 01/27/2025] Open
Abstract
Pneumonia is a common respiratory infection affecting individuals of all ages, with a significantly higher incidence among the elderly. As the aging population grows, pneumonia is expected to become an increasingly critical health concern. In non-institutionalized elderly individuals, the annual incidence ranges from 25 to 44 per 1000, approximately four times higher than in those under 65. Streptococcus pneumoniae, a Gram-positive diplococcus, is the leading cause of pneumonia-related deaths in older adults. Management of S. pneumoniae infections in the elderly is challenging due to impaired antibody responses to polysaccharides and surface proteins, compounded by rising antibiotic resistance. The underlying mechanisms for increased susceptibility remain unclear, but age-related changes in the immune system, particularly in dendritic cells and T cells, are implicated. This review explores how aging-related immune alterations contribute to the heightened vulnerability of the elderly to S. pneumoniae infections.
Collapse
Affiliation(s)
- Nathan Kang
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA 92697, USA
| | - Veedamali S. Subramanian
- Division of Gastroenterology, Department of Medicine, University of California Irvine, Irvine, CA 92697, USA;
| | - Anshu Agrawal
- Division of Basic and Clinical Immunology, Department of Medicine, University of California Irvine, Irvine, CA 92697, USA
| |
Collapse
|
16
|
Mella C, Tsarouhas P, Brockwell M, Ball HC. The Role of Chronic Inflammation in Pediatric Cancer. Cancers (Basel) 2025; 17:154. [PMID: 39796780 PMCID: PMC11719864 DOI: 10.3390/cancers17010154] [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: 12/04/2024] [Revised: 12/31/2024] [Accepted: 01/01/2025] [Indexed: 01/13/2025] Open
Abstract
Inflammation plays a crucial role in wound healing and the host immune response following pathogenic invasion. However, unresolved chronic inflammation can result in tissue fibrosis and genetic alterations that contribute to the pathogenesis of human diseases such as cancer. Recent scientific advancements exploring the underlying mechanisms of malignant cellular transformations and cancer progression have exposed significant disparities between pediatric and adult-onset cancers. For instance, pediatric cancers tend to have lower mutational burdens and arise in actively developing tissues, where cell-cycle dysregulation leads to gene, chromosomal, and fusion gene development not seen in adult-onset counterparts. As such, scientific findings in adult cancers cannot be directly applied to pediatric cancers, where unique mutations and inherent etiologies remain poorly understood. Here, we review the role of chronic inflammation in processes of genetic and chromosomal instability, the tumor microenvironment, and immune response that result in pediatric tumorigenesis transformation and explore current and developing therapeutic interventions to maintain and/or restore inflammatory homeostasis.
Collapse
Affiliation(s)
- Christine Mella
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
| | - Panogiotis Tsarouhas
- Department of Biology, The University of Akron, 302 Buchtel Common, Akron, OH 44325, USA;
| | - Maximillian Brockwell
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
| | - Hope C. Ball
- Division of Hematology Oncology, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA;
- College of Medicine, Northeast Ohio Medical University, 4029 State Route 44, Rootstown, OH 44272, USA;
- Rebecca D. Considine Research Institute, Akron Children’s Hospital, One Perkins Square, Akron, OH 44308, USA
| |
Collapse
|
17
|
Choe JY, Jones HP. Methods for Modeling Early Life Stress in Rodents. Methods Mol Biol 2025; 2868:205-219. [PMID: 39546232 DOI: 10.1007/978-1-0716-4200-9_11] [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: 11/17/2024]
Abstract
Animal models of early life stress/adversity (ELS) have provided a foundation from which our understanding of the psychoneuroimmunology of childhood trauma has expanded over recent decades. Rodent models are a cornerstone of the ELS literature with many studies utilizing paradigms based on early life separation/deprivation protocols and manipulating the cage environment. However, no animal model is perfect. In particular, the lack of standardization across ELS models has led to inconsistent results and raised questions regarding the translational value of common preclinical models. In this chapter, we present an overview of the history of ELS rodent models and discuss considerations relevant to the ongoing efforts to both improve existing models and generate novel paradigms to meet the evolving needs of molecular- and mechanism-based ELS research.
Collapse
Affiliation(s)
- Jamie Y Choe
- Texas College of Osteopathic Medicine, University of North Texas Health Science Center, Fort Worth, TX, USA
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Harlan P Jones
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA.
- Institute for Health Disparities, University of North Texas Health Science Center, Fort Worth, TX, USA.
| |
Collapse
|
18
|
Anticoli S, Capanna S, Volpin A, Melis P, Ortona E, Vonesch N, Tomao P, Scapellato ML, Ruggieri A. Sex differences in response to HBV vaccination in a cohort of health care workers. Vaccine X 2025; 22:100605. [PMID: 39811673 PMCID: PMC11731748 DOI: 10.1016/j.jvacx.2024.100605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 10/15/2024] [Accepted: 12/16/2024] [Indexed: 01/16/2025] Open
Abstract
Globally, healthcare workers (HCWs) are at greater risk of contracting Hepatitis B virus (HBV) infection than the general population, due to their frequent contact with blood or body fluids. For this reason, WHO underlined the importance of HBV immunization for all HCWs. Although sex is now considered one of the key factors influencing the intensity and duration of the immune response to vaccines, sex-specific analysis of vaccine-induced anti-HBs antibodies is rarely conducted. Our study aimed to evaluate the anti-HBs antibodies' long-lasting protective titers in male and female HCWs to identify sex differences in the humoral response to the HBV vaccine. The study was conducted on 2106 HCWs (1636 females and 470 males) working at the Padova University Hospital, Italy. Our results report significantly higher anti-HBs levels in female HCWs than in males, particularly when considering HCWs for whom the elapsed time since vaccination is greater than 10 years, suggesting a greater persistence of anti-HBs in females. In conclusion, our findings could contribute to understanding differences in the antibody response to the HBV vaccine between male and female subjects and improving vaccination strategies for HCWs.
Collapse
Affiliation(s)
- Simona Anticoli
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Silvia Capanna
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, (Rome) Italy
| | - Anna Volpin
- Occupational Medicine Unit, University Hospital of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Paola Melis
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, (Rome) Italy
| | - Elena Ortona
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| | - Nicoletta Vonesch
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, (Rome) Italy
| | - Paola Tomao
- Department of Occupational and Environmental Medicine, Epidemiology and Hygiene, Italian Workers' Compensation Authority (INAIL), Monte Porzio Catone, (Rome) Italy
| | - Maria Luisa Scapellato
- Occupational Medicine Unit, University Hospital of Padova, Via Giustiniani 2, 35128 Padova, Italy
- Department of Cardiac Thoracic Vascular Sciences and Public Health, University of Padova, Via Giustiniani 2, 35128 Padova, Italy
| | - Anna Ruggieri
- Reference Center for Gender-specific Medicine, Istituto Superiore di Sanità [Italian National Institute of Health], Rome, Italy
| |
Collapse
|
19
|
Vissers M, van de Garde MDB, He SWJ, Brandsen M, Hendriksen R, Nicolaie MA, van der Maas L, Meiring HD, van Els CACM, van Beek J, Rots NY. Quantity and Quality of Naturally Acquired Antibody Immunity to the Pneumococcal Proteome Throughout Life. J Infect Dis 2024; 230:1466-1475. [PMID: 38888894 DOI: 10.1093/infdis/jiae255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 05/07/2024] [Indexed: 06/20/2024] Open
Abstract
BACKGROUND Young children and older adults are susceptible for invasive pneumococcal disease (IPD) caused by Streptococcus pneumoniae. Pneumococcal protein-specific antibodies play a protective role against IPD; however, not much is known about the pace of acquisition, maturation, and maintenance of these antibodies throughout life. METHODS Immunoglobulin G (IgG) and IgA levels, avidity, and/or specificity to the pneumococcal proteome in serum and saliva from healthy young children, adults, and older adults, with known carriage status, were measured by enzyme-linked immunosorbent assay (ELISA) and 2-dimensional western blotting against ΔcpsTIGR4. RESULTS Eleven-month-old children, the youngest age group tested, had the lowest pneumococcal proteome-specific IgG and IgA levels and avidity in serum and saliva, followed by 24-month-old children and were further elevated in adult groups. Among adult groups, the parents had the highest serum and saliva IgG and IgA antibody levels. In children, antibody levels and avidity correlated with daycare attendance and presence of siblings, posing as proxy for exposure and immunization. Immunodominance patterns slightly varied throughout life. CONCLUSIONS Humoral immunity against the pneumococcal proteome is acquired through multiple episodes of pneumococcal exposure. Low-level and low-avidity antiproteome antibody profiles in young children may contribute to their IPD susceptibility, while in overall antiproteome antibody-proficient older adults other factors likely play a role.
Collapse
Affiliation(s)
- Marloes Vissers
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Martijn D B van de Garde
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Samantha W J He
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Milou Brandsen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Rosanne Hendriksen
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Mioara Alina Nicolaie
- Expertise Centre for Methodology and Information Services, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Larissa van der Maas
- Product Characterization and Formulation, Institute for Translational Vaccinology, Bilthoven, The Netherlands
| | - Hugo D Meiring
- Product Characterization and Formulation, Institute for Translational Vaccinology, Bilthoven, The Netherlands
| | - Cecile A C M van Els
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Infectious Diseases and Immunology, Department of Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands
| | - Josine van Beek
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Nynke Y Rots
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
20
|
Gu W, Xie D, Li Q, Feng H, Xue Y, Chen Y, Tang J, Zhou Y, Wang D, Tong S, Liu S. Association of humidity and precipitation with asthma: a systematic review and meta-analysis. FRONTIERS IN ALLERGY 2024; 5:1483430. [PMID: 39713043 PMCID: PMC11659254 DOI: 10.3389/falgy.2024.1483430] [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: 08/21/2024] [Accepted: 11/25/2024] [Indexed: 12/24/2024] Open
Abstract
Introduction The relationship of asthma with humidity and precipitation remains controversial. The objective of this study was to investigate the association of increased humidity and precipitation with asthma risk. Methods A comprehensive systematic search was conducted across various databases, including PubMed, Embase, Cochrane Library, Web of Science, Chinese Wanfang, CQVIP, and CNKI. A total of 21 studies with 1,052,960 participants from 9 countries or regions were included. The fixed and random effect model were used to analyze the data. Results The study revealed a pooled odds ratio (OR) of 1.0489 [95% confidence interval (CI): 1.0061, 1.0935] for the association between humidity and asthma risk. Specifically, among individuals under 18 years of age, the OR (95% CI) was 1.0898 (1.0290, 1.1541). Furthermore, the OR (95% CI) for developing countries or regions was 1.0927 (1.0220, 1.1684), while it was 1.1298 (0.9502, 1.3433) for regions with a high latitude (41°-51°). The pooled OR for precipitation and asthma risk was 0.9991 (0.9987, 0.9995). The OR (95%CI) values were 0.9991 (0.9987, 0.9995), 0.9991 (0.9987, 0.9995) and 0.9990 (0.9986, 0.9994) in people above the age of 18, developing countries or regions, and middle latitudes (31°-41°), respectively. Discussion The impact of humidity on asthma risk is particularly pronounced among individuals below 18 years of age, people living in developing countries or regions and in regions located in high latitudes. And the influence of precipitation on asthma persons over the age of 18, developing countries or regions, and middle latitudes significantly. Increased humidity appears to elevate asthma risk, and increased precipitation may reduce the risk. In addition, there appears to be a combined effect of humidity and precipitation on asthma. Systematic Review Registration PROSPERO, identifier, CRD42023482446.
Collapse
Affiliation(s)
- Wangyang Gu
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Dan Xie
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Qinpeng Li
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Huike Feng
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Yihao Xue
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Yang Chen
- Department of Epidemiology and Statistics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jingyi Tang
- Department of Epidemiology and Statistics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Yushi Zhou
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Dan Wang
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| | - Shilu Tong
- Department of Epidemiology and Statistics, School of Public Health and Social Work, Queensland University of Technology, Brisbane, QLD, Australia
- Chinese Centers for Disease Control and Prevention, National Institute of Environmental Health, Beijing, China
| | - Shijian Liu
- Department of Big Data Center, Sanya Women and Children's Hospital Affiliated to Hainan Medical University, Sanya, China
- Hainan Branch, Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
- Department of Epidemiology and Statistics, School of Public Health, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Children’s Medical Center, School of Medicine, Shanghai Jiao Tong University, Sanya, China
| |
Collapse
|
21
|
Zhang G, Wang K, Ba L, Dong S, Gao J. Dynamic changes in the circulation of respiratory pathogens in children during and after the containment of the 2019 coronavirus disease pandemic in Kunming, China. Eur J Clin Microbiol Infect Dis 2024; 43:2259-2268. [PMID: 39292354 DOI: 10.1007/s10096-024-04945-1] [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: 05/24/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
PURPOSE We aimed to determine the changes in the frequency of respiratory pathogens and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) during containment of the 2019 coronavirus disease pandemic and elucidate the epidemiological interference that may have occurred after lifting pandemic measures. METHODS A total of 4,770 Nasopharyngeal swab samples were collected from children with ARTIs from the First People's Hospital of Yunnan Province between January 2022 and December 2023 and subjected to nucleic acid testing for 13 types of respiratory pathogens and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). RESULTS The frequency of pathogens among children from 2020 to 2022 was in the following order: HRV > Mp > HADV > H3N2 > HMPV and HRV > HRSV > HPIV > H1N1 > H3N2. In weeks 1 to 3 of 2023, the frequency of pathogens significantly declined, and then H1N1 rebounded significantly in 2023. HRV, HRSV, and H3N2 showed a shift in the season of high frequency. Patterns of multi-pathogen infections were more complex in 2023 than in 2022, with HRV having a higher frequency and co-infection rate than other pathogens. These changes may have been associated with interference caused by the resurgence of SARS-CoV-2 prevalence, in addition to being influenced by changes in pandemic containment and lifting measures. CONCLUSIONS The frequency rate of common respiratory pathogens among children was not significantly different and remained high. The study findings help elucidate the aforementioned unique historical period and effectively control respiratory tract infections to reduce the harm to pediatric health caused by respiratory pathogens.
Collapse
Affiliation(s)
- Guiqian Zhang
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
- Institute of Basic and Clinical Medicine, Yunnan Provincial Key Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Kunming, 650100, China
| | - Kaimei Wang
- Department of Medical Technology, Yunnan University of Business Management, Kunming, Yunnan, China
| | - Limei Ba
- Department of Clinical Laboratory, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, China
| | - Shumei Dong
- Department of Medical Technology, Yunnan University of Business Management, Kunming, Yunnan, China
| | - Jianmei Gao
- Institute of Basic and Clinical Medicine, Yunnan Provincial Key Laboratory of Clinical Virology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, 157 Jinbi Road, Kunming, 650100, China.
| |
Collapse
|
22
|
Fernandez M, Pezier T, Papadopoulos S, Laurent F, Werts C, Lacroix-Lamandé S. Deleterious intestinal inflammation in neonatal mice treated with TLR2/TLR6 agonists. J Leukoc Biol 2024; 116:1142-1156. [PMID: 38872374 DOI: 10.1093/jleuko/qiae140] [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: 09/19/2023] [Revised: 05/16/2024] [Accepted: 06/13/2024] [Indexed: 06/15/2024] Open
Abstract
By providing innate immune modulatory stimuli, the early-life immune system can be enhanced to increase resistance to infections. Activation of innate cell surface receptors called pattern recognition receptors by Toll-like receptor (TLR) ligands is one promising approach that can help to control infections as described for listeriosis and cryptosporidiosis. In this study, the effect of TLR2/TLR1 and TLR2/TLR6 agonists was compared when injected into neonatal mice. Surprisingly, the stimulation of TLR2/TLR6 led to the death of the neonatal mice, which was not observed in adult mice. The TLR2/TLR6 agonist administration induced higher systemic and intestinal inflammation in both adult and neonatal mice when compared with TLR2/TLR1 agonist. The mortality of neonatal mice was interferon γ dependent and involved the intestinal production of interleukin-22 and interleukin-17A. This study clearly demonstrates that targeting TLRs as new control strategy of neonatal infections has to be used with caution. Depending on its heterodimeric form, TLR2 stimulation can induce more or less severe adverse effects relying on the age-related immune functions of the host.
Collapse
Affiliation(s)
- Mégane Fernandez
- Infectiologie et Santé Publique, Université de Tours, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, F-37380 Nouzilly, France
| | - Tiffany Pezier
- Infectiologie et Santé Publique, Université de Tours, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, F-37380 Nouzilly, France
| | - Stylianos Papadopoulos
- Centre National de la Recherche Scientifique UMR6047, Institut National de la Santé et de la Recherche Médicale U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Université Paris Cité, Paris, France
| | - Fabrice Laurent
- Infectiologie et Santé Publique, Université de Tours, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, F-37380 Nouzilly, France
| | - Catherine Werts
- Centre National de la Recherche Scientifique UMR6047, Institut National de la Santé et de la Recherche Médicale U1306, Unité de Biologie et Génétique de la Paroi Bactérienne, Institut Pasteur, Université Paris Cité, Paris, France
| | - Sonia Lacroix-Lamandé
- Infectiologie et Santé Publique, Université de Tours, Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement, F-37380 Nouzilly, France
| |
Collapse
|
23
|
Groen J, van der Kuip M, Budding D, Bos MP, Benninga MA, Niemarkt HJ, de Meij TGJ. Assessing Diagnostic Performance of Molecular Culture for Neonatal Sepsis: Protocol of the CHAMPIONS Study. Diagnostics (Basel) 2024; 14:1930. [PMID: 39272715 PMCID: PMC11394283 DOI: 10.3390/diagnostics14171930] [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: 08/05/2024] [Revised: 08/26/2024] [Accepted: 08/29/2024] [Indexed: 09/15/2024] Open
Abstract
Managing neonatal sepsis is challenging due to nonspecific clinical signs, hematological markers with poor accuracy, and a lengthy turnaround time for the identification of microorganisms. Delaying the initiation of antibiotics in truly infected infants can lead to severe morbidity and mortality. Therefore, decisions regarding empiric antibiotic treatment are risk stratified, which exposes many uninfected infants to antibiotics. This causes gut microbiota perturbation, unnecessary hospital admissions, and the generation of multi-resistant organisms. High-speed diagnostic assays could expedite discontinuation or avert the initiation of antibiotics in uninfected infants. This study will evaluate the diagnostic performance of molecular culture (MC), a rapid broad-range PCR-based bacterial profiling technique, for diagnosing neonatal sepsis in infants below 90 days old. A multi-center prospective observational cohort study will include infants evaluated for early and late-onset sepsis. Routine evaluation for suspected sepsis includes microbiological cultures of blood. Additionally, blood for MC will be collected. For early-onset sepsis, umbilical cord blood may be used alternatively. Primary outcome is the agreement between MC and conventional blood culture results. Secondary outcome is the agreement of both assays with clinical sepsis using four different, commonly used definitions. Faster diagnostic pathways for sepsis may reduce antibiotic exposure time. Broad-range molecular assays may identify pathogens undetectable by conventional methods. Employment of umbilical cord blood samples for early-onset sepsis diagnosis can resolve challenges in collecting adequate blood volume and could further expedite treatment decisions.
Collapse
Affiliation(s)
- Jip Groen
- Department of Pediatric Gastroenterology, Amsterdam University Medical Center,1105 AZ Amsterdam, The Netherlands
- Amsterdam Reproduction and Development Research Institute, 1105 AZ Amsterdam, The Netherlands
- Gastroenterology and Hepatology, Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Center, University of Amsterdam, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Martijn van der Kuip
- Department of Pediatric Infectious Diseases, Rheumatology and Immunology, Amsterdam University Medical Center, 1105 AZ Amsterdam, The Netherlands
| | | | | | - Marc A Benninga
- Department of Pediatric Gastroenterology, Amsterdam University Medical Center,1105 AZ Amsterdam, The Netherlands
| | - Hendrik J Niemarkt
- Maxima Medical Center, Department of Neonatology, 5504 DB Veldhoven, The Netherlands
| | - Tim G J de Meij
- Department of Pediatric Gastroenterology, Amsterdam University Medical Center,1105 AZ Amsterdam, The Netherlands
| |
Collapse
|
24
|
Gal DB, Cleveland JD, Vergales JE, Kipps AK. Immunisation deferral practices surrounding congenital heart surgery. Cardiol Young 2024; 34:1550-1553. [PMID: 38557603 DOI: 10.1017/s1047951124000507] [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] [Indexed: 04/04/2024]
Abstract
BACKGROUND Perioperative immunisation administration surrounding congenital heart surgery is controversial. Delayed immunisation administration results in children being at risk of vaccine-preventable illnesses and is associated with failure to complete immunisation schedules. Among children with CHD, many of whom are medically fragile, vaccine-preventable illnesses can be devastating. Limited research shows perioperative immunisation may be safe and effective. METHODS We surveyed Pediatric Acute Care Cardiology Collaborative member centres and explored perioperative immunisation practices. We analysed responses using descriptive statistics. RESULTS Complete responses were submitted by 35/46 (76%) centres. Immunisations were deferred for any period prior to surgery by 23 (66%) centres and after surgery by 31 (89%) centres. Among those who deferred post-operative immunisation, 20 (65%) required deferral only for patients whose operations required cardiopulmonary bypass. Duration of deferral in the pre- and post-operative periods was variable. Many centres included exceptions to their policy for specific vaccine-preventable illnesses. Almost all (34, 97%) centres administer routine childhood immunisation to patients who remain admitted for prolonged periods. CONCLUSIONS Most centres defer routine childhood immunisation for some period before and after congenital heart surgery. Centre specific practices vary. Immunisation deferral confers risk to patients and may not be warranted in this population. Further research would be necessary to understand the immunologic impact of these practices.
Collapse
Affiliation(s)
- Dana B Gal
- Department of Pediatrics, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
- Division of Pediatric Cardiology, Children's Hospital of Los Angeles, Los Angeles, CA, USA
| | - John D Cleveland
- Division of Pediatric Cardiology, Children's Hospital of Los Angeles, Los Angeles, CA, USA
- Department of Cardiothoracic Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Jeffrey E Vergales
- Division of Pediatric Cardiology, University of Virginia, Charlottesville, VA, USA
| | - Alaina K Kipps
- Division of Pediatric Cardiology, Department of Pediatrics, Stanford University School of Medicine, Palo Alto, CA, USA
| |
Collapse
|
25
|
Zhu S, Wei W. Progress in research on the role of fluoride in immune damage. Front Immunol 2024; 15:1394161. [PMID: 38807586 PMCID: PMC11130356 DOI: 10.3389/fimmu.2024.1394161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024] Open
Abstract
Excessive fluoride intake from residential environments may affect multiple tissues and organs; however, the specific pathogenic mechanisms are unclear. Researchers have recently focused on the damaging effects of fluoride on the immune system. Damage to immune function seriously affects the quality of life of fluoride-exposed populations and increases the incidence of infections and malignant tumors. Probing the mechanism of damage to immune function caused by fluoride helps identify effective drugs and methods to prevent and treat fluorosis and improve people's living standards in fluorosis-affected areas. Here, the recent literature on the effects of fluoride on the immune system is reviewed, and research on fluoride damage to the immune system is summarized in terms of three perspectives: immune organs, immune cells, and immune-active substances. We reviewed that excessive fluoride can damage immune organs, lead to immune cells dysfunction and interfere with the expression of immune-active substances. This review aimed to provide a potential direction for future fluorosis research from the perspective of fluoride-induced immune function impairment. In order to seek the key regulatory indicators of fluoride on immune homeostasis in the future.
Collapse
Affiliation(s)
- Siqi Zhu
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
| | - Wei Wei
- Center for Endemic Disease Control, Chinese Center for Disease Control and Prevention, Harbin Medical University, Harbin, Heilongjiang, China
- Key Lab of Etiology and Epidemiology, Education Bureau of Heilongjiang Province & Ministry of Health, Harbin Medical University, Harbin, China
- Heilongjiang Provincial Key Lab of Trace Elements and Human Health Harbin Medical University, Harbin, China
| |
Collapse
|
26
|
Alugupalli KR. A TLR4 ligand-based adjuvant for promoting the immunogenicity of typhoid subunit vaccines. Front Immunol 2024; 15:1383476. [PMID: 38799439 PMCID: PMC11116679 DOI: 10.3389/fimmu.2024.1383476] [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: 02/07/2024] [Accepted: 04/19/2024] [Indexed: 05/29/2024] Open
Abstract
None of the typhoid Vi Polysaccharide (ViPS) subunit vaccines incorporate adjuvants, and the immunogenicity of ViPS vaccines (e.g. Typbar TCV® and Typhim Vi®) is in part due to associated TLR4 ligands such as endotoxin present in these vaccines. Since endotoxin content in vaccines is variable and kept very low due to inherent toxicity, it was hypothesized that incorporating a defined amount of a non-toxic TLR4-ligand such as monophosphoryl lipid A in ViPS vaccines would improve their immunogenicity. To test this hypothesis, a monophosphoryl lipid A-based adjuvant formulation named Turbo was developed. Admixing Turbo with Typbar TCV® (ViPS-conjugated to tetanus toxoid) increased the levels of anti-ViPS IgM, IgG1, IgG2b, IgG2a/c, and IgG3 in inbred and outbred mice. In infant mice, a single immunization with Turbo adjuvanted Typbar TCV® resulted in a significantly increased and durable IgG response and improved the control of bacterial burden compared to mice immunized without Turbo. Similarly, when adjuvanted with Turbo, the antibody response and control of bacteremia were also improved in mice immunized with Typhim Vi®, an unconjugated vaccine. The immunogenicity of unconjugated ViPS is inefficient in young mice and is lost in adult mice when immunostimulatory ligands in ViPS are removed. Nevertheless, when adjuvanted with Turbo, poorly immunogenic ViPS induced a robust IgG response in young and adult mice, and this was observed even under antigen-limiting conditions. These data suggest that incorporation of Turbo as an adjuvant will make typhoid vaccines more immunogenic regardless of their intrinsic immunogenicity or conjugation status and maximize the efficacy across all ages.
Collapse
Affiliation(s)
- Kishore R. Alugupalli
- Department of Microbiology and Immunology, Thomas Jefferson University, Philadelphia, PA, United States
- TurboVax Inc, Philadelphia, PA, United States
| |
Collapse
|
27
|
Uhl C, Klevebro S, Sverremark-Ekström E, Tedner SG, Brandström J, Papageorgiou C, Melén E, Konradsen JR, Nilsson C, Asarnoj A. High Degree of Desensitization After 1 Year of Early-Life Peanut Oral Immunotherapy: Small Children Oral Immunotherapy (SmaChO) Randomized Controlled Trial. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2024; 12:1297-1305. [PMID: 38428524 DOI: 10.1016/j.jaip.2024.02.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 02/02/2024] [Accepted: 02/21/2024] [Indexed: 03/03/2024]
Abstract
BACKGROUND The prevalence of peanut allergy is about 2% and mostly lifelong. Studies of oral immunotherapy (OIT) with peanut (the daily oral intake of an initially low and then increasing dose of peanut) often show problematic side effects, but there are indications of better safety and effect in younger children compared with older children and adults. OBJECTIVE To determine the safety and effectiveness of peanut OIT with a slow up-dosing strategy and low maintenance dose in children aged 1 to 3 years who were allergic to peanut, through a 1-year interim analysis. METHOD In a randomized controlled trial (2:1 ratio), 75 children, median age 31 months (interquartile range [IQR], 23-40 months) were assigned to receive peanut OIT (n = 50) or peanut avoidance (n = 25). RESULTS In the OIT and avoidance groups, 43 of 50 and 20 of 25 children, respectively, performed the 1-year open oral peanut challenge. A cumulative dose of 750 mg peanut protein after 1 year was tolerated by 72% (36 of 50 children) in the OIT group compared with 4% (1 of 25) in the avoidance group (P < .001). Median tolerated cumulative dose was 2,750 mg (IQR, 275-5,000 mg) peanut protein in the OIT group compared with 2.8 mg (IQR, 0.3-27.8 mg) in the avoidance group (P < .001). Of the doses administered at home during the first year of OIT, 1.4% resulted in adverse events and 79% were mild, and three doses of epinephrine were given at home to two individuals. CONCLUSION In children aged 1 to 3 years, peanut OIT with the combination of slow up-dosing and low maintenance dose seems safe and effective after 1 year.
Collapse
Affiliation(s)
- Carina Uhl
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Susanna Klevebro
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden.
| | - Eva Sverremark-Ekström
- Department of Molecular Biosciences, Wenner-Gren Institute, Stockholm University, Stockholm, Sweden
| | - Sandra G Tedner
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden
| | - Josef Brandström
- Department of Neonatology, Astrid Lindgrens Childreńs Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Chrystalleni Papageorgiou
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Erik Melén
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Jon R Konradsen
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Allergy and Pulmonology Unit, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| | - Caroline Nilsson
- Department of Clinical Science and Education, Södersjukhuset, Karolinska Institutet, Stockholm, Sweden; Sachs' Children and Youth Hospital, Södersjukhuset, Stockholm, Sweden
| | - Anna Asarnoj
- Department of Women's and Children's Health, Karolinska Institutet, Stockholm, Sweden; Pediatric Allergy and Pulmonology Unit, Astrid Lindgren Children's Hospital, Karolinska University Hospital, Stockholm, Sweden
| |
Collapse
|
28
|
Tosca MA, Varricchio A, Schiavetti I, Naso M, Damiani V, Ciprandi G. Managing children with frequent respiratory infections and associated wheezing: a preliminary randomized study with a new multicomponent nasal spray. Allergol Immunopathol (Madr) 2024; 52:22-30. [PMID: 38721952 DOI: 10.15586/aei.v52i3.1040] [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: 11/15/2023] [Accepted: 02/05/2024] [Indexed: 06/09/2024]
Abstract
BACKGROUND Preschoolers frequently have respiratory infections (RIs), which may cause wheezing in some subjects. Type 2 polarization may favor increased susceptibility to RIs and associated wheezing. Non-pharmacological remedies are garnering increasing interest as possible add-on therapies. The present preliminary study investigated the efficacy and safety of a new multi-component nasal spray in preschoolers with frequent RIs and associated wheezing. METHODS Some preschoolers with these characteristics randomly took this product, containing lactoferrin, dipotassium glycyrrhizinate, carboxymethyl-beta-glucan, and vitamins C and D3 (Saflovir), two sprays per nostril twice daily for 3 months. Other children were randomly treated only with standard therapy. Outcomes included the number of RIs and wheezing episodes, use of medications, and severity of clinical manifestations. RESULTS Preschoolers treated add-on with this multicomponent product experienced fewer RIs and used fewer beta-2 agonists than untreated children (P = 0.01 and 0.029, respectively). CONCLUSIONS This preliminary study demonstrated that a multicomponent product, administered add-on as a nasal spray, could reduce the incidence of RIs and use of symptomatic drugs for relieving wheezing in children.
Collapse
Affiliation(s)
| | | | | | - Matteo Naso
- Allergy Center, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | | | | |
Collapse
|
29
|
Boll EJ, Lopez DV, Terne M, Hessing S, Parschat K, Jensen SR. Human milk oligosaccharides differentially support gut barrier integrity and enhance Th1 and Th17 cell effector responses in vitro. Front Immunol 2024; 15:1359499. [PMID: 38510254 PMCID: PMC10950922 DOI: 10.3389/fimmu.2024.1359499] [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: 12/21/2023] [Accepted: 02/15/2024] [Indexed: 03/22/2024] Open
Abstract
Human milk oligosaccharides (HMOs) can modulate the intestinal barrier and regulate immune cells to favor the maturation of the infant intestinal tract and immune system, but the precise functions of individual HMOs are unclear. To determine the structure-dependent effects of individual HMOs (representing different structural classes) on the intestinal epithelium as well as innate and adaptive immune cells, we assessed fucosylated (2'FL and 3FL), sialylated (3'SL and 6'SL) and neutral non-fucosylated (LNT and LNT2) HMOs for their ability to support intestinal barrier integrity, to stimulate the secretion of chemokines from intestinal epithelial cells, and to modulate cytokine release from LPS-activated dendritic cells (DCs), M1 macrophages (MØs), and co-cultures with naïve CD4+ T cells. The fucosylated and neutral non-fucosylated HMOs increased barrier integrity and protected the barrier following an inflammatory insult but exerted minimal immunomodulatory activity. The sialylated HMOs enhanced the secretion of CXCL10, CCL20 and CXCL8 from intestinal epithelial cells, promoted the secretion of several cytokines (including IL-10, IL-12p70 and IL-23) from LPS-activated DCs and M1 MØs, and increased the secretion of IFN-γ and IL-17A from CD4+ T cells primed by LPS-activated DCs and MØs while reducing the secretion of IL-13. Thus, 3'SL and 6'SL supported Th1 and Th17 responses while reducing Th2 responses. Collectively, our data show that HMOs exert structure-dependent effects on the intestinal epithelium and possess immunomodulatory properties that confer benefits to infants and possibly also later in life.
Collapse
Affiliation(s)
| | | | - Mandy Terne
- Chr. Hansen A/S, Applied HMOs, Hoersholm, Denmark
| | - Sara Hessing
- Chr. Hansen A/S, Applied HMOs, Hoersholm, Denmark
| | | | | |
Collapse
|
30
|
Froń A, Orczyk-Pawiłowicz M. Understanding the Immunological Quality of Breast Milk in Maternal Overweight and Obesity. Nutrients 2023; 15:5016. [PMID: 38140275 PMCID: PMC10746120 DOI: 10.3390/nu15245016] [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: 11/14/2023] [Revised: 11/30/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Maternal obesity, affecting many pregnant women globally, not only poses immediate health risks but also modulates breast milk composition. Obesity is linked to inflammation and oxidative stress, impacting breast milk's immune properties. This paper explores the intricate relationship between maternal metabolic disorders, such as obesity, and breast milk's immunological components. We conducted a thorough search for original and review articles published until 17 October 2023 in the PUBMED/Scopus database. This search included several terms related to human breast milk, immunological properties, and obesity. Articles were selected with the consensus of all authors. Maternal metabolic disorders have discernible effects on the composition of immune-related components in breast milk, such as immunoglobulins, lactoferrin, leptin, ghrelin, adiponectin, C-reactive protein, growth factors, extracellular vesicles, and lymphocytes. These changes in breast milk composition can significantly impact the newborn's immune system, with potential long-term health implications beyond the immediate postnatal period. Maternal metabolic health is a critical factor in shaping the health trajectory of the neonate through breastfeeding, although the full advantages of breastfeeding for children of mothers with obesity remain uncertain. Ongoing research aims to understand and unravel these links.
Collapse
Affiliation(s)
- Anita Froń
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| | - Magdalena Orczyk-Pawiłowicz
- Division of Chemistry and Immunochemistry, Department of Biochemistry and Immunochemistry, Wroclaw Medical University, M. Skłodowskiej-Curie 48/50, 50-369 Wroclaw, Poland
| |
Collapse
|
31
|
Joshi D, Nyhoff LE, Zarnitsyna VI, Moreno A, Manning K, Linderman S, Burrell AR, Stephens K, Norwood C, Mantus G, Ahmed R, Anderson EJ, Staat MA, Suthar MS, Wrammert J. Infants and young children generate more durable antibody responses to SARS-CoV-2 infection than adults. iScience 2023; 26:107967. [PMID: 37822504 PMCID: PMC10562792 DOI: 10.1016/j.isci.2023.107967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 08/25/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023] Open
Abstract
As SARS-CoV-2 becomes endemic, it is critical to understand immunity following early-life infection. We evaluated humoral responses to SARS-CoV-2 in 23 infants/young children. Antibody responses to SARS-CoV-2 spike antigens peaked approximately 30 days after infection and were maintained up to 500 days with little apparent decay. While the magnitude of humoral responses was similar to an adult cohort recovered from mild/moderate COVID-19, both binding and neutralization titers to WT SARS-CoV-2 were more durable in infants/young children, with spike and RBD IgG antibody half-life nearly 4X as long as in adults. IgG subtype analysis revealed that while IgG1 formed the majority of the response in both groups, IgG3 was more common in adults and IgG2 in infants/young children. These findings raise important questions regarding differential regulation of humoral immunity in infants/young children and adults and could have broad implications for the timing of vaccination and booster strategies in this age group.
Collapse
Affiliation(s)
- Devyani Joshi
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Lindsay E. Nyhoff
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | | | - Alberto Moreno
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
- Department of Medicine, Emory University, School of Medicine, Atlanta, GA, USA
| | - Kelly Manning
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Susanne Linderman
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Allison R. Burrell
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Environmental and Public Health Sciences, Division of Epidemiology, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Kathy Stephens
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Carson Norwood
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Grace Mantus
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| | - Rafi Ahmed
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
| | - Evan J. Anderson
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
- Department of Medicine, Emory University, School of Medicine, Atlanta, GA, USA
| | - Mary A. Staat
- Division of Infectious Diseases, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, USA
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, USA
| | - Mehul S. Suthar
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
- Department of Microbiology and Immunology, Emory University, Atlanta, GA, USA
- Emory National Primate Research Center, Atlanta, GA, USA
| | - Jens Wrammert
- Division of Infectious Diseases, Department of Pediatrics, School of Medicine, Emory University, Atlanta, GA, USA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA, USA
- Centers for Childhood Infections and Vaccines, Children’s Healthcare of Atlanta, Emory University Department of Pediatrics Department of Medicine, Atlanta, GA, USA
| |
Collapse
|
32
|
Joshi D, Nyhoff LE, Zarnitsyna VI, Moreno A, Manning K, Linderman S, Burrell AR, Stephens K, Norwood C, Mantus G, Ahmed R, Anderson EJ, Staat MA, Suthar MS, Wrammert J. Infants and young children generate more durable antibody responses to SARS-CoV-2 infection than adults. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.04.10.23288360. [PMID: 37090559 PMCID: PMC10120804 DOI: 10.1101/2023.04.10.23288360] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/25/2023]
Abstract
Since the emergence of SARS-CoV-2, research has shown that adult patients mount broad and durable immune responses to infection. However, response to infection remains poorly studied in infants/young children. In this study, we evaluated humoral responses to SARS-CoV-2 in 23 infants/young children before and after infection. We found that antibody responses to SARS-CoV-2 spike antigens peaked approximately 30 days after infection and were maintained up to 500 days with little apparent decay. While the magnitude of humoral responses was similar to an adult cohort recovered from mild/moderate COVID-19, both binding and neutralization titers to WT SARS-CoV-2 were more durable in infants/young children, with Spike and RBD IgG antibody half-life nearly 4X as long as in adults. The functional breadth of adult and infant/young children SARS-CoV-2 responses were comparable, with similar reactivity against panel of recent and previously circulating viral variants. Notably, IgG subtype analysis revealed that while IgG1 formed the majority of both adults' and infants/young children's response, IgG3 was more common in adults and IgG2 in infants/young children. These findings raise important questions regarding differential regulation of humoral immunity in infants/young children and adults and could have broad implications for the timing of vaccination and booster strategies in this age group.
Collapse
|
33
|
Zhou SH, Zhang RY, You ZW, Zou YK, Wen Y, Wang J, Ding D, Bian MM, Zhang ZM, Yuan H, Yang GF, Guo J. pH-Sensitive and Biodegradable Mn 3(PO 4) 2·3H 2O Nanoparticles as an Adjuvant of Protein-Based Bivalent COVID-19 Vaccine to Induce Potent and Broad-Spectrum Immunity. ACS APPLIED MATERIALS & INTERFACES 2023; 15:acsami.2c19736. [PMID: 36748861 PMCID: PMC9924082 DOI: 10.1021/acsami.2c19736] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Developing a novel and potent adjuvant with great biocompatibility for immune response augmentation is of great significance to enhance vaccine efficacy. In this work, we prepared a long-term stable, pH-sensitive, and biodegradable Mn3(PO4)2·3H2O nanoparticle (nano-MnP) by simply mixing MnCl2/NaH2PO4/Na2HPO4 solution for the first time and employed it as an immune stimulant in the bivalent COVID-19 protein vaccine comprised of wild-type S1 (S1-WT) and Omicron S1 (S1-Omicron) proteins as antigens to elicit a broad-spectrum immunity. The biological experiments indicated that the nano-MnP could effectively activate antigen-presenting cells through the cGAS-STING pathway. Compared with the conventional Alum-adjuvanted group, the nano-MnP-adjuvanted bivalent vaccine elicited approximately 7- and 8-fold increases in IgG antibody titers and antigen-specific IFN-γ secreting T cells, respectively. Importantly, antisera of the nano-MnP-adjuvanted group could effectively cross-neutralize the SARS-CoV-2 and its five variants of concern (VOCs) including Alpha, Beta, Gamma, Delta, and Omicron, demonstrating that this bivalent vaccine based on S1-WT and S1-Omicron proteins is an effective vaccine design strategy to induce broad-spectrum immune responses. Collectively, this nano-MnP material may provide a novel and efficient adjuvant platform for various prophylactic and therapeutic vaccines and provide insights for the development of the next-generation manganese adjuvant.
Collapse
Affiliation(s)
| | | | - Zi-Wei You
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Yong-Ke Zou
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Yu Wen
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Jian Wang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Dong Ding
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Miao-Miao Bian
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Zhi-Ming Zhang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Hong Yuan
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| | - Jun Guo
- Key Laboratory of Pesticide & Chemical Biology of Ministry of Education,
International Joint Research Center for Intelligent Biosensing Technology and Health,
Hubei International Scientific and Technological Cooperation Base of Pesticide and Green
Synthesis, College of Chemistry, Central China Normal
University, Wuhan 430079, China
| |
Collapse
|