1
|
van der Staak M, ten Hulscher HI, Nicolaie AM, Smits GP, de Swart RL, de Wit J, Rots NY, van Binnendijk RS. Long-term Dynamics of Measles Virus-Specific Neutralizing Antibodies in Children Vaccinated Before 12 Months of Age. Clin Infect Dis 2025; 80:904-910. [PMID: 39492687 PMCID: PMC12043057 DOI: 10.1093/cid/ciae537] [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: 09/05/2024] [Revised: 10/07/2024] [Accepted: 10/31/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Measles is a highly contagious disease, presenting a significant risk for unvaccinated infants and adults. Measles vaccination under the age of 12 months provides early protection but has also been associated with blunting of antibody responses to subsequent measles vaccinations and assumed to have lower vaccine effectiveness. METHODS Our study included children who received an early measles, mumps, and rubella (MMR) vaccination between 6 and 12 months of age (n = 79, given in addition to the regular MMR vaccination schedule at 14 months and 9 years) and a group without additional early vaccination (n = 44). We evaluated measles virus (MeV)-specific neutralizing antibodies before vaccination at 14 months and up to 6 years thereafter using a plaque reduction neutralization test according to the standard set by the World Health Organization. RESULTS We found a significant association between age of first MMR and MeV-specific neutralizing antibody levels later in life. Although most children who received early vaccination seroconverted after the first dose, children vaccinated before 8.5 months of age exhibited a markedly faster antibody decay and lost their protective neutralizing antibody levels over 6 years. CONCLUSIONS Routine vaccination of infants under 8.5 months of age may lead to blunted MeV-specific antibody responses to subsequent MMR vaccination. Early MMR vaccination should only be considered during measles outbreaks or in other situations of increased risk of MeV infection. Clinical Trials Registration. EudraCT 2013-003078-28.
Collapse
Affiliation(s)
- Maaike van der Staak
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Hinke I ten Hulscher
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Alina M Nicolaie
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Gaby P Smits
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Rik L de Swart
- Department of Viroscience, Erasmus MC, Rotterdam, The Netherlands
| | - Jelle de Wit
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Nynke Y Rots
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| | - Robert S van Binnendijk
- Center for Infectious Disease Control, National Institute for Public Health and the Environment, Bilthoven, The Netherlands
| |
Collapse
|
2
|
Young Graf Z, Kolagani R, Stetter C, Hausman BL, Cruz AT, Lambert SJ. Association of Vaccination in Pregnancy With Newborn Hepatitis B Vaccine Receipt. Perm J 2025; 29:84-88. [PMID: 40040392 PMCID: PMC11907668 DOI: 10.7812/tpp/24.061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 09/28/2024] [Accepted: 01/08/2025] [Indexed: 03/06/2025]
Abstract
INTRODUCTION The authors evaluated factors associated with neonatal hepatitis B vaccination (HepB), including prenatal vaccinations, race, ethnicity, neonatal disposition, parity, and maternal age to identify potential areas of engagement to improve maternal and child health. METHODS The authors conducted a retrospective cohort study of patients who received prenatal care and delivered at an academic tertiary care hospital in central Pennsylvania from 2015-2020. A multiple logistic regression model was used to assess factors associated with newborn receipt of HepB. RESULTS Prenatal vaccination was significantly (P < .0001) associated with subsequent neonatal HepB vaccination in the hospital following birth. Race, Hispanic ethnicity, age at delivery, neonatal disposition, and parity were not shown to be associated with HepB vaccine uptake. CONCLUSION Prenatal vaccination was significantly associated with neonatal in-hospital HepB vaccine uptake.
Collapse
Affiliation(s)
- Zeva Young Graf
- Pennsylvania State University College of Medicine, Hershey, PA, USA
- Now with Department of Pediatrics, Children's Hospital of Colorado, Aurora, CO, USA
| | - Ramya Kolagani
- Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Christina Stetter
- Department of Obstetrics and Gynecology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| | - Bernice L Hausman
- Department of Humanities, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Amy T Cruz
- Department of Obstetrics and Gynecology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
- Now with Department of Clinical Obstetrics, Gynecology, and Reproductive Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, USA
| | - Stephanie J Lambert
- Department of Obstetrics and Gynecology, Penn State Health Milton S Hershey Medical Center, Hershey, PA, USA
| |
Collapse
|
3
|
Wang Y, Li R, Tong R, Chen T, Sun M, Luo L, Li Z, Chen Y, Zhao Y, Zhang C, Wei L, Lin W, Chen H, Qian K, Chen AF, Liu J, Chen L, Li B, Wang F, Wang L, Su B, Pu J. Integrating single-cell RNA and T cell/B cell receptor sequencing with mass cytometry reveals dynamic trajectories of human peripheral immune cells from birth to old age. Nat Immunol 2025; 26:308-322. [PMID: 39881000 PMCID: PMC11785523 DOI: 10.1038/s41590-024-02059-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 12/09/2024] [Indexed: 01/31/2025]
Abstract
A comprehensive understanding of the evolution of the immune landscape in humans across the entire lifespan at single-cell transcriptional and protein levels, during development, maturation and senescence is currently lacking. We recruited a total of 220 healthy volunteers from the Shanghai Pudong Cohort (NCT05206643), spanning 13 age groups from 0 to over 90 years, and profiled their peripheral immune cells through single-cell RNA-sequencing coupled with single T cell and B cell receptor sequencing, high-throughput mass cytometry, bulk RNA-sequencing and flow cytometry validation experiments. We revealed that T cells were the most strongly affected by age and experienced the most intensive rewiring in cell-cell interactions during specific age. Different T cell subsets displayed different aging patterns in both transcriptomes and immune repertoires; examples included GNLY+CD8+ effector memory T cells, which exhibited the highest clonal expansion among all T cell subsets and displayed distinct functional signatures in children and the elderly; and CD8+ MAIT cells, which reached their peaks of relative abundance, clonal diversity and antibacterial capability in adolescents and then gradually tapered off. Interestingly, we identified and experimentally verified a previously unrecognized 'cytotoxic' B cell subset that was enriched in children. Finally, an immune age prediction model was developed based on lifecycle-wide single-cell data that can evaluate the immune status of healthy individuals and identify those with disturbed immune functions. Our work provides both valuable insights and resources for further understanding the aging of the immune system across the whole human lifespan.
Collapse
MESH Headings
- Humans
- Aged
- Single-Cell Analysis/methods
- Adult
- Aged, 80 and over
- Middle Aged
- Infant
- Child
- Child, Preschool
- Male
- Female
- Adolescent
- Aging/immunology
- Aging/genetics
- Infant, Newborn
- Receptors, Antigen, B-Cell/genetics
- Receptors, Antigen, B-Cell/immunology
- Flow Cytometry
- Young Adult
- Receptors, Antigen, T-Cell/genetics
- Sequence Analysis, RNA
- T-Lymphocyte Subsets/immunology
- B-Lymphocytes/immunology
Collapse
Affiliation(s)
- Yufei Wang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Ronghong Li
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Renyang Tong
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Taiwei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Mingze Sun
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lingjie Luo
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Li
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yifan Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Yichao Zhao
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Chensheng Zhang
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Lin Wei
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Wei Lin
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Haoyan Chen
- Department of Gastroenterology and Hepatology, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kun Qian
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
- School of Biomedical Engineering, Institute of Medical Robotics and Shanghai Academy of Experimental Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Alex F Chen
- Institute for Developmental and Regenerative Cardiovascular Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junling Liu
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lei Chen
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China
| | - Bin Li
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Wang
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bing Su
- Center for Immune-Related Diseases at Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Pu
- Department of Cardiology, Renji Hospital, School of Medicine, State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Shanghai Jiao Tong University, Shanghai, China.
- Aging Biomarker Consortium, Beijing, China.
| |
Collapse
|
4
|
Schumer A, Bonney EA, Harby E, Majumdar D. Neonatal SARS-CoV-2 mRNA Vaccination Efficacy Is Influenced by Maternal Antibodies. Am J Reprod Immunol 2024; 92:e70001. [PMID: 39436146 DOI: 10.1111/aji.70001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/20/2024] [Accepted: 09/30/2024] [Indexed: 10/23/2024] Open
Abstract
PROBLEM Vaccination in pregnancy guards against infection. Maternal antibodies, however, can inhibit antibody production in neonates. We sought to determine the effects of maternal vaccination on neonatal immune response to a SARS-CoV-2 mRNA vaccine. METHOD OF STUDY We hypothesized that mRNA-lipid nanoparticles (LNP) vaccination allows for a de novo neonatal antibody response even in the presence of vertically transmitted maternal antibodies. Female mice were vaccinated with SARS-CoV-2 spike receptor binding domain (RBD) mRNA-LNPs. Mice were then bred, and 21-day-old pups were inoculated with the same mRNA-LNPs. Spike-specific IgG ELISAs were performed using mouse serum. A SARS-CoV-2 spike protein peptide library to perform peptide ELISAs characterized high affinity binding domains within the spike protein. Results were analyzed with one-way ANOVAs with Tukey's multiple comparisons tests. RESULTS Compared to pups of unvaccinated dams, there were high levels of spike-specific IgG detected in the pups of vaccinated dams at 3 weeks of life (p < 0.0001). After neonatal vaccination, pups of unvaccinated dams had higher spike-specific serum IgG than pups of vaccinated dams at 12 weeks of life (p < 0.001). Antibody specificity to peptide moieties within spike RBD were similar when comparing a vaccinated dam to her pup at Week 3 of life, with different binding affinities observed in the pups by Week 15 of life. CONCLUSIONS Pre-existing maternal antibodies may partially blunt the initial neonatal antibody response to mRNA-LNPs vaccination. This vaccine strategy, however, does not prohibit the subsequent development of a broad range of RBD antibody specificities that may be protective.
Collapse
Affiliation(s)
- Amy Schumer
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Medical Center, Burlington, Vermont, USA
| | - Elizabeth A Bonney
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Vermont Medical Center, Burlington, Vermont, USA
| | - Ethan Harby
- Biological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Devdoot Majumdar
- Department of Surgery, University of Vermont Medical Center, Burlington, Vermont, USA
- Department of Electrical and Biomedical Engineering, University of Vermont Medical Center, Burlington, Vermont, USA
- UVM Cancer Center, University of Vermont Medical Center, Burlington, Vermont, USA
| |
Collapse
|
5
|
Damtie D, Gelaw A, Wondimeneh Y, Aleka Y, Kick MK, Tigabu Z, Sack U, Mekuria ZH, Vlasova AN, Tessema B. Rotavirus A Infection Prevalence and Spatio-Temporal Genotype Shift among Under-Five Children in Amhara National Regional State, Ethiopia: A Multi-Center Cross-Sectional Study. Vaccines (Basel) 2024; 12:866. [PMID: 39203992 PMCID: PMC11360187 DOI: 10.3390/vaccines12080866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 09/03/2024] Open
Abstract
Background: Globally, rotavirus (RV) A (RVA) is the most common cause of severe and sometimes fatal diarrhea in young children. It is also the major cause of acute gastroenteritis among children in Ethiopia. Currently, the WHO has prequalified four RVA vaccines for universal childhood immunization. Ethiopia introduced the monovalent Rotarix vaccine into its national immunization program in 2013. Since then, only a few studies on the burden and genotype distribution of RVA infection post-vaccine introduction have been conducted (mostly at sentinel surveillance sites). Therefore, this study aimed to assess RVA prevalence and genotype distribution among children under five years in Ethiopia (February 2021-December 2022). Methods: This multi-center hospital-based cross-sectional study involved 537 diarrheic children under-five years old. Rotavirus A detection was conducted using a one-step reverse-transcriptase polymerase chain reaction (RT-PCR). Genotyping was conducted by Sanger sequencing of the VP7 (complete) and VP4 (partial) genes. Descriptive analysis and Pearson's chi-squared test were carried out using SPSS version 29. Phylogenetic analysis with 1000 bootstrap replicates was performed using MEGA version 11 software. Statistical significance was set at p < 0.05 for all analyses. Results: The prevalence of RVA infection among diarrheic children was 17.5%. The most prevalent G-types identified were G3 (37%), the previously uncommon G12 (28%), and G1 (20%). The predominant P-types were P[8] (51%), P[6] (29%), and P[4] (14%). The three major G/P combinations observed were G3P[8] (32.8%), G12P[6] (28.4%), and G1P[8] (19.4%). Phylogenetic analysis revealed clustering of Ethiopian strains with the globally reported strains. Many strains exhibited amino acid differences in the VP4 (VP8* domain) and VP7 proteins compared to vaccine strains, potentially affecting virus neutralization. Conclusions: Despite the high RVA vaccination rate, the prevalence of RVA infection remains significant among diarrheic children in Ethiopia. There is an observable shift in circulating RVA genotypes from G1 to G3, alongside the emergence of unusual G/P genotype combinations such as G9P[4]. Many of these circulating RVA strains have shown amino acid substitutions that may allow for neutralization escape. Therefore, further studies are warranted to comprehend the emergence of these unusual RVA strains and the diverse factors influencing the vaccine's diminished effectiveness in developing countries.
Collapse
Affiliation(s)
- Debasu Damtie
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia; (A.G.); (Y.W.); (B.T.)
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia;
- Ohio State University Global One Health Initiative LLC, Eastern Africa Regional Office, Bole Road, Noah Plaza, 2nd Floor, Addis Ababa, Ethiopia
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Aschalew Gelaw
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia; (A.G.); (Y.W.); (B.T.)
| | - Yitayih Wondimeneh
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia; (A.G.); (Y.W.); (B.T.)
| | - Yetemwork Aleka
- Department of Immunology and Molecular Biology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia;
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Maryssa K. Kick
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Zemene Tigabu
- Department of Pediatrics and Child Health, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia;
| | - Ulrich Sack
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany;
| | - Zelalem H. Mekuria
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA;
- Global One Health initiative (GOHi), The Ohio State University, Columbus, OH 43210, USA
| | - Anastasia N. Vlasova
- Center for Food Animal Health, Department of Animal Sciences, College of Food Agricultural and Environmental Sciences, The Ohio State University, Wooster, OH 44691, USA;
- Department of Veterinary Preventive Medicine, College of Veterinary Medicine, The Ohio State University, Columbus, OH 43210, USA;
| | - Belay Tessema
- Department of Medical Microbiology, School of Biomedical and Laboratory Sciences, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia; (A.G.); (Y.W.); (B.T.)
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany;
| |
Collapse
|
6
|
Lababidi G, Lababidi H, Bitar F, Arabi M. COVID-19 Vaccines in the Pediatric Population: A Focus on Cardiac Patients. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:2667033. [PMID: 38779616 PMCID: PMC11111306 DOI: 10.1155/2024/2667033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 04/04/2024] [Accepted: 04/27/2024] [Indexed: 05/25/2024]
Abstract
Due to the deleterious global impact of the COVID-19 pandemic, tremendous effort has been invested in the development of vaccines against the virus. Vaccine candidates are first tested in adult populations, a number of which have been approved for EUL by the WHO, and are in use across the USA and MENA region. The question remains whether these (or other) vaccines should be recommended to a neonatal, pediatric, and/or adolescent cohort. Incidence and severity of COVID-19 infection are low in pediatric, neonatal, and adolescent patients. Since both overall incidence and severity are lower in children than in adults, safety is an important consideration in vaccine approval for these age groups, in addition to efficacy and a decreased risk of transmission. The following review discusses vaccine immunology in children aged 0-18 years, with emphasis on the negative impact of the COVID-19 pandemic on the lives of children, considerations for pediatric vaccine approval, and available vaccines for pediatric cohorts along with a breakdown of the efficacy, advantages, and disadvantages for each. This review also contains current and future perspectives, as well as a section on the cardiovascular implications and related dynamics of pediatric COVID-19 vaccination.
Collapse
Affiliation(s)
- Ghena Lababidi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Hossam Lababidi
- Faculty of Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Fadi Bitar
- Children's Heart Center, Division of Pediatric Cardiology, Pediatric Department, American University of Beirut Medical Center, Beirut, Lebanon
| | - Mariam Arabi
- Children's Heart Center, Division of Pediatric Cardiology, Pediatric Department, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
7
|
Li C, Zhao M, Liu X, Li Y, Xu B, Zhou L, Sun X, Sun W, Kang N, Ji Z, Li T, An H, Wang F, Wu C, Ye JY, Zhang JR, Wang Q, Zhao X, Li Z, Liu W. Ion channel TRPV2 is critical in enhancing B cell activation and function. J Exp Med 2024; 221:e20221042. [PMID: 38353705 PMCID: PMC10866685 DOI: 10.1084/jem.20221042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 11/28/2023] [Accepted: 01/22/2024] [Indexed: 02/16/2024] Open
Abstract
The function of transient receptor potential vanilloid (TRPV) cation channels governing B cell activation remains to be explored. We present evidence that TRPV2 is highly expressed in B cells and plays a crucial role in the formation of the B cell immunological synapse and B cell activation. Physiologically, TRPV2 expression level is positively correlated to influenza-specific antibody production and is low in newborns and seniors. Pathologically, a positive correlation is established between TRPV2 expression and the clinical manifestations of systemic lupus erythematosus (SLE) in adult and child SLE patients. Correspondingly, mice with deficient TRPV2 in B cells display impaired antibody responses following immunization. Mechanistically, the pore and N-terminal domains of TRPV2 are crucial for gating cation permeation and executing mechanosensation in B cells upon antigen stimulation. These processes synergistically contribute to membrane potential depolarization and cytoskeleton remodeling within the B cell immunological synapse, fostering efficient B cell activation. Thus, TRPV2 is critical in augmenting B cell activation and function.
Collapse
Affiliation(s)
- Cuifeng Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| | - Meng Zhao
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Xiaohang Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Yuxin Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Bihua Xu
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Lina Zhou
- Department of Pediatric Research Institute, Children’s Hospital of Chongqing Medical University, Chongqing, China
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, PR China
| | - Xiaolin Sun
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Wenbo Sun
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Na Kang
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Zhenglin Ji
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Tong Li
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Haoran An
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Fei Wang
- Chengdu Institute of Biology, Chinese Academy of Sciences, Chengdu, China
| | - Chuan Wu
- Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jing-Ying Ye
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Jing-Ren Zhang
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Center for Infectious Disease Research, School of Medicine, Tsinghua University, Beijing, China
| | - Qingwen Wang
- Department of Rheumatism and Immunology, Peking University Shenzhen Hospital, Shenzhen, China
- Shenzhen Key Laboratory of Inflammatory and Immunology Diseases, Shenzhen, China
| | - Xiaodong Zhao
- Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders (Chongqing), China International Science and Technology Cooperation Base of Child Development and Critical Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, PR China
- Department of Rheumatology and Immunology, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Zhanguo Li
- Tsinghua-Peking Center for Life Sciences, Beijing, China
- Department of Rheumatology and Immunology, Peking University People’s Hospital, Beijing, China
- Beijing Key Laboratory for Rheumatism Mechanism and Immune Diagnosis (BZ0135), Beijing, China
| | - Wanli Liu
- State Key Laboratory of Membrane Biology, School of Life Sciences, Institute for Immunology, China Ministry of Education Key Laboratory of Protein Sciences, Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
- Tsinghua-Peking Center for Life Sciences, Beijing, China
| |
Collapse
|
8
|
Gao C, Chen Q, Hao X, Wang Q. Immunomodulation of Antibody Glycosylation through the Placental Transfer. Int J Mol Sci 2023; 24:16772. [PMID: 38069094 PMCID: PMC10705935 DOI: 10.3390/ijms242316772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/18/2023] Open
Abstract
Establishing an immune balance between the mother and fetus during gestation is crucial, with the placenta acting as the epicenter of immune tolerance. The placental transfer of antibodies, mainly immunoglobulin G (IgG), is critical in protecting the developing fetus from infections. This review looks at how immunomodulation of antibody glycosylation occurs during placental transfer and how it affects fetal health. The passage of maternal IgG antibodies through the placental layers, including the syncytiotrophoblast, stroma, and fetal endothelium, is discussed. The effect of IgG subclass, glycosylation, concentration, maternal infections, and antigen specificity on antibody transfer efficiency is investigated. FcRn-mediated IgG transport, influenced by pH-dependent binding, is essential for placental transfer. Additionally, this review delves into the impact of glycosylation patterns on antibody functionality, considering both protective and pathological effects. Factors affecting the transfer of protective antibodies, such as maternal vaccination, are discussed along with reducing harmful antibodies. This in-depth examination of placental antibody transfer and glycosylation provides insights into improving neonatal immunity and mitigating the effects of maternal autoimmune and alloimmune conditions.
Collapse
Affiliation(s)
| | | | | | - Qiushi Wang
- Department of Blood Transfusion, Shengjing Hospital of China Medical University, Shenyang 110004, China
| |
Collapse
|
9
|
Sears KT, Nasrin S, Baliban SM, Council DN, Pasetti MF, Tennant SM. Evaluation of Three Candidate Live-Attenuated Salmonella enterica Serovar Typhimurium Vaccines to Prevent Non-Typhoidal Salmonella Infection in an Infant Mouse Model. Vaccines (Basel) 2023; 11:1562. [PMID: 37896965 PMCID: PMC10610874 DOI: 10.3390/vaccines11101562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 09/22/2023] [Accepted: 09/26/2023] [Indexed: 10/29/2023] Open
Abstract
Nontyphoidal Salmonella enterica (NTS) is a leading cause of foodborne illness worldwide, including in the United States, where infants show the highest incidence amongst all age groups. S. enterica serovar Typhimurium is one of the most frequently isolated serovars from NTS infections. We have developed several candidate live-attenuated S. Typhimurium vaccines to prevent NTS infection. The goal of the current study was to assess three live S. Typhimurium vaccine strains (CVD 1921, CVD 1921 ∆htrA and CVD 1926, which have two, three and four gene deletions, respectively) with various levels of reactogenicity and immunogenicity in infant BALB/c mice to predict how they would perform following peroral immunization of infants. We first tested intranasal immunization of 14-day-old mice with three doses delivered at 1-week intervals and evaluated antibody responses and protection against lethal infection with wild-type S. Typhimurium. The vaccines were administered to 14-day-old mice via the peroral route at 1- or 2-week intervals and to 28-day-old mice at 2-week intervals. The three vaccine strains were immunogenic following intranasal immunization of infant mice with vaccine efficacies of 80% (CVD 1921), 63% (CVD 1921 ∆htrA) and 31% (CVD 1926). In contrast, peroral immunization of 14-day-old mice yielded much poorer protection against lethal infection and only immunization of 28-day-old mice at 2-week intervals showed similar protective capacity as intranasal administration (CVD 1921: 83%, CVD 1921 ∆htrA: 43% and CVD 1926: 58%). CVD 1921 was consistently more protective than both CVD 1921 ∆htrA and CVD 1926, regardless of the route of vaccination, immunization schedule and age of mice. Anti-LPS serum IgG responses were similar between the three strains and did not correlate with protection. Due to previously observed reactogenicity of CVD 1921, CVD 1921 ∆htrA and CVD 1926 are our preferred vaccines, but these data show that further improvements would need to be made to achieve suitable protection in young infants when using peroral immunization.
Collapse
Affiliation(s)
- Khandra T. Sears
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.S.); (S.M.B.); (M.F.P.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Shamima Nasrin
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.S.); (S.M.B.); (M.F.P.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Scott M. Baliban
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.S.); (S.M.B.); (M.F.P.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Danielle N. Council
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.S.); (S.M.B.); (M.F.P.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Marcela F. Pasetti
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.S.); (S.M.B.); (M.F.P.)
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sharon M. Tennant
- Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD 21201, USA; (K.T.S.); (S.M.B.); (M.F.P.)
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
10
|
Zhang Y, Xu JC, Hu ZD, Fan XY. Advances in protein subunit vaccines against tuberculosis. Front Immunol 2023; 14:1238586. [PMID: 37654500 PMCID: PMC10465801 DOI: 10.3389/fimmu.2023.1238586] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 07/25/2023] [Indexed: 09/02/2023] Open
Abstract
Tuberculosis (TB), also known as the "White Plague", is caused by Mycobacterium tuberculosis (Mtb). Before the COVID-19 epidemic, TB had the highest mortality rate of any single infectious disease. Vaccination is considered one of the most effective strategies for controlling TB. Despite the limitations of the Bacille Calmette-Guérin (BCG) vaccine in terms of protection against TB among adults, it is currently the only licensed TB vaccine. Recently, with the evolution of bioinformatics and structural biology techniques to screen and optimize protective antigens of Mtb, the tremendous potential of protein subunit vaccines is being exploited. Multistage subunit vaccines obtained by fusing immunodominant antigens from different stages of TB infection are being used both to prevent and to treat TB. Additionally, the development of novel adjuvants is compensating for weaknesses of immunogenicity, which is conducive to the flourishing of subunit vaccines. With advances in the development of animal models, preclinical vaccine protection assessments are becoming increasingly accurate. This review summarizes progress in the research of protein subunit TB vaccines during the past decades to facilitate the further optimization of protein subunit vaccines that may eradicate TB.
Collapse
Affiliation(s)
- Ying Zhang
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Jin-chuan Xu
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
| | - Zhi-dong Hu
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai, China
| | - Xiao-yong Fan
- Shanghai Public Health Clinical Center, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai, China
- TB Center, Shanghai Emerging and Re-emerging Infectious Disease Institute, Fudan University, Shanghai, China
| |
Collapse
|
11
|
Stoddard CI, Sung K, Yaffe ZA, Weight H, Beaudoin-Bussières G, Galloway J, Gantt S, Adhiambo J, Begnel ER, Ojee E, Slyker J, Wamalwa D, Kinuthia J, Finzi A, Matsen FA, Lehman DA, Overbaugh J. Elevated binding and functional antibody responses to SARS-CoV-2 in infants versus mothers. Nat Commun 2023; 14:4864. [PMID: 37567924 PMCID: PMC10421871 DOI: 10.1038/s41467-023-40554-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 08/01/2023] [Indexed: 08/13/2023] Open
Abstract
Infant antibody responses to viral infection can differ from those in adults. However, data on the specificity and function of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies in infants, and direct comparisons between infants and adults are limited. Here, we characterize antibody binding and functionality against Wuhan-Hu-1 (B lineage) strain SARS-CoV-2 in convalescent plasma from 36 postpartum women and 14 of their infants infected with SARS-CoV-2 from a vaccine-naïve prospective cohort in Nairobi, Kenya. We find significantly higher antibody titers against SARS-CoV-2 Spike, receptor binding domain and N-terminal domain, and Spike-expressing cell-surface staining levels in infants versus mothers. Plasma antibodies from mothers and infants bind to similar regions of the Spike S2 subunit, including the fusion peptide (FP) and stem helix-heptad repeat 2. However, infants display higher antibody levels and more consistent antibody escape pathways in the FP region compared to mothers. Finally, infants have significantly higher levels of antibody-dependent cellular cytotoxicity (ADCC), though, surprisingly, Spike pseudovirus neutralization titers between infants and mothers are similar. These results suggest infants develop distinct SARS-CoV-2 binding and functional antibody activities and reveal age-related differences in humoral immunity to SARS-CoV-2 infection that could be relevant to protection and COVID-19 disease outcomes.
Collapse
Affiliation(s)
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Zak A Yaffe
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Medical Scientist Training Program, University of Washington, Seattle, WA, USA
| | - Haidyn Weight
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Jared Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Soren Gantt
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
- Centre de Recherche du CHU Sainte-Justine, Université de Montréal, Montreal, QC, Canada
| | - Judith Adhiambo
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Emily R Begnel
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Ednah Ojee
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - Jennifer Slyker
- Department of Global Health, University of Washington, Seattle, WA, USA
| | - Dalton Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi, Nairobi, Kenya
| | - John Kinuthia
- Department of Global Health, University of Washington, Seattle, WA, USA
- Department of Research and Programs, Kenyatta National Hospital, Nairobi, Kenya
| | - Andrés Finzi
- Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal, Montreal, QC, Canada
| | - Frederick A Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Dara A Lehman
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Department of Global Health, University of Washington, Seattle, WA, USA.
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
- Public Health Sciences Division, Fred Hutchinson Cancer Center, Seattle, WA, USA.
| |
Collapse
|
12
|
López-Serrano S, Mahmmod YS, Christensen D, Ebensen T, Guzmán CA, Rodríguez F, Segalés J, Aragón V. Immune responses following neonatal vaccination with conserved F4 fragment of VtaA proteins from virulent Glaesserella parasuis adjuvanted with CAF®01 or CDA. Vaccine X 2023; 14:100330. [PMID: 37361051 PMCID: PMC10285277 DOI: 10.1016/j.jvacx.2023.100330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/07/2023] [Accepted: 06/07/2023] [Indexed: 06/28/2023] Open
Abstract
Glaesserella parasuis is a Gram-negative bacterium that colonizes the upper airways of swine, capable of causing a systemic infection called Glässer's disease. This disease is more frequent in young post-weaning piglets. Current treatments against G. parasuis infection are based on the use of antimicrobials or inactivated vaccines, which promote limited cross-protection against different serovars. For this reason, there is an interest in developing novel subunit vaccines with the capacity to confer effective protection against different virulent strains. Herein, we characterize the immunogenicity and the potential benefits of neonatal immunization with two different vaccine formulations based on the F4 polypeptide, a conserved immunogenic protein fragment from the virulence-associated trimeric autotransporters of virulent G. parasuis strains. With this purpose, we immunized two groups of piglets with F4 combined with cationic adjuvant CAF®01 or cyclic dinucleotide CDA. Piglets immunized with a commercial bacterin and non-immunized animals served as control groups. The vaccinated piglets received two doses of vaccine, at 14 days old and 21 days later. The immune response induced against the F4 polypeptide varied depending on the adjuvant used. Piglets vaccinated with the F4+CDA vaccine developed specific anti-F4 IgGs, biased towards the induction of IgG1 responses, whereas no anti-F4 IgGs were de novo induced after immunization with the CAF®01 vaccine. Piglets immunized with both formulations displayed balanced memory T-cell responses, evidenced upon in vitro re-stimulation of peripheral blood mononuclear cells with F4. Interestingly, pigs immunized with F4+CAF®01 controlled more efficiently a natural nasal colonization by a virulent serovar 4 G. parasuis that spontaneously occurred during the experimental procedure. According to the results, the immunogenicity and the protection afforded by F4 depend on the adjuvant used. F4 may represent a candidate to consider for a Glässer's disease vaccine and could contribute to a better understanding of the mechanisms involved in protection against virulent G. parasuis colonization.
Collapse
Affiliation(s)
- Sergi López-Serrano
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Catalonia, Spain
| | - Yasser S. Mahmmod
- Department of Animal Medicine, Faculty of Veterinary Medicine, Zagazig University, Zagazig 44511, Egypt
- Section of Veterinary Sciences, Health Sciences Division, Al Ain Men’s College, Higher Colleges of Technology, Al Ain 17155, United Arab Emirates
| | - Dennis Christensen
- Department of Infectious Disease Immunology, Statens Serum Institut, Artillerivej 5, 2300 Copenhagen, Denmark
| | - Thomas Ebensen
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Carlos A. Guzmán
- Department of Vaccinology and Applied Microbiology, Helmholtz Centre for Infection Research, Inhoffenstraße 7, 38124 Braunschweig, Germany
| | - Fernando Rodríguez
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Catalonia, Spain
| | - Joaquim Segalés
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Departament de Sanitat i Anatomia animals. Facultat de Veterinària. Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
| | - Virginia Aragón
- Unitat mixta d’Investigació IRTA-UAB en Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- Institut de Recerca i Tecnologia Agroalimentàries, Programa de Sanitat Animal, Centre de Recerca en Sanitat Animal (CReSA), Campus de la Universitat Autònoma de Barcelona (UAB), Bellaterra 08193, Catalonia, Spain
- WOAH Collaborating Centre for the Research and Control of Emerging and Re-Emerging Swine Diseases in Europe (IRTA-CReSA), 08193 Bellaterra, Catalonia, Spain
| |
Collapse
|
13
|
Lodi L, Barbati F, Amicizia D, Baldo V, Barbui AM, Bondi A, Costantino C, Da Dalt L, Ferrara L, Fortunato F, Guarnieri V, Icardi G, Indolfi G, Martinelli D, Martini M, Moriondo M, Nieddu F, Peroni DG, Prato R, Ricci S, Russo F, Tirelli F, Vitale F, Ladhani SN, Azzari C. Four-Component Recombinant Protein-Based Vaccine Effectiveness Against Serogroup B Meningococcal Disease in Italy. JAMA Netw Open 2023; 6:e2329678. [PMID: 37594762 PMCID: PMC10439479 DOI: 10.1001/jamanetworkopen.2023.29678] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 06/21/2023] [Indexed: 08/19/2023] Open
Abstract
Importance Population-based data on the 4-component recombinant protein-based (4CMenB) vaccine effectiveness and reduction in incidence rate ratios (IRRs) are continuously needed to assess vaccine performance in the prevention of serogroup B invasive meningococcal disease (IMD). Objective To assess the effectiveness and reduction in IRRs associated with the 4CMenB vaccine in the pediatric population in 6 regions in Italy. Design, Setting, and Participants This retrospective cohort screening study and case-control study included data from children aged younger than 6 years in 6 highly populated Italian regions from January 1, 2006, to January 1, 2020. Participants included children younger than 6 years diagnosed with serogroup B IMD without predisposing factors. Data were collected from regional surveillance and vaccination registries and were analyzed from September 2021 to January 2022. Exposures Routine 4CMenB vaccination, per regional vaccination programs. Main Outcomes and Measures The main outcome was the effectiveness of the 4CMenB vaccine in the prevention of serogroup B IMD in the population of children aged younger than 6 years in 6 Italian regions. The percentages of vaccine effectiveness (VE) were obtained through the concomitant use of a screening method and a case-control study. Secondary outcomes were the comparison of effectiveness results obtained using the 2 different computational methods, the description of serogroup B IMD incidence rates, and reduction in IRRs before and after 4CMenB introduction, as a proxy for vaccine impact. Results The cohort screening study included a resident population of 587 561 children younger than 6 years in 3 regions with similar surveillance protocols, and the matched-case controls study assessed a resident population of 1 080 620 children younger than 6 years in 6 regions. Analyses found that 4CMenB VE in fully immunized children was 94.9% (95% CI, 83.1%-98.4%) using the screening method and 91.7% (95% CI, 24.4%-98.6%) using the case-control method. Overall reduction in IRR was 50%, reaching 70% in regions with early-start vaccination schedules. The case-control method involving 6 highly-populated Italian regions included 26 cases and 52 controls and found an estimated VE of 92.4% (95% CI, 67.6%-97.9%) in children old enough for the first vaccine dose and 95.6% (95% CI, 71.7%-99.1%) in fully immunized children. VE was more than 90% for partially immunized children. Even in regions where the first dose was administered at age 2 months, almost 20% of unvaccinated cases were among infants too young to receive the first 4CMenB dose. Conclusions and Relevance This screening cohort study and matched case-controls study found high effectiveness of 4CMenB vaccination and greater reduction in IRR for early-start vaccination schedules in preventing invasive serogroup B meningococcal disease. The high proportion of children too young to be vaccinated among unvaccinated cases suggests that starting the vaccination even earlier may prevent more cases. Screening and case-control methods provided similar estimates of VE: either method may be used in different study settings, but concomitant use can provide more robust estimates.
Collapse
Affiliation(s)
- Lorenzo Lodi
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Federica Barbati
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Daniela Amicizia
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Vincenzo Baldo
- Department of Cardiac, Thoracic, Vascular Sciences and Public Health University of Padua, Padua, Italy
| | - Anna Maria Barbui
- S.C. Microbiology and Virology Laboratory, City of Health and Science, Turin, Italy
| | - Alessandro Bondi
- S.C. Microbiology and Virology Laboratory, City of Health and Science, Turin, Italy
| | - Claudio Costantino
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Excellence Specialties “G. D’Alessandro,” University of Palermo, Palermo, Italy
| | - Liviana Da Dalt
- Department of Woman’s and Child’s Health, Padua University Hospital, Padua, Italy
| | - Lorenza Ferrara
- Regional Epidemiology Reference Service for the Surveillance, Prevention and Control of Infectious Diseases, Local Health Unit of Alessandria, Alessandria, Italy
| | - Francesca Fortunato
- Hygiene Unit, Department of Medical and Surgical Sciences, Policlinico Foggia Hospital, University of Foggia, Foggia, Italy
| | - Valentina Guarnieri
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Giancarlo Icardi
- Department of Health Sciences, University of Genoa, Genoa, Italy
| | - Giuseppe Indolfi
- Paediatric and Liver Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Neurofarba, University of Florence, Florence, Italy
| | - Domenico Martinelli
- Hygiene Unit, Department of Medical and Surgical Sciences, Policlinico Foggia Hospital, University of Foggia, Foggia, Italy
| | | | - Maria Moriondo
- Laboratory of Immunology and Molecular Microbiology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Francesco Nieddu
- Laboratory of Immunology and Molecular Microbiology, Meyer Children’s Hospital IRCCS, Florence, Italy
| | - Diego G. Peroni
- Pediatric Clinic, Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Rosa Prato
- Hygiene Unit, Department of Medical and Surgical Sciences, Policlinico Foggia Hospital, University of Foggia, Foggia, Italy
| | - Silvia Ricci
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| | - Francesca Russo
- Veneto Regional Directorate of Prevention, Food Safety, Veterinary, Public Health, Venice, Italy
| | - Francesca Tirelli
- Department of Woman’s and Child’s Health, Padua University Hospital, Padua, Italy
| | - Francesco Vitale
- Department of Health Promotion Sciences, Maternal and Infant Care, Internal Medicine and Excellence Specialties “G. D’Alessandro,” University of Palermo, Palermo, Italy
| | - Shamez N. Ladhani
- National Infection Service, Public Health England, London, United Kingdom
- Paediatric Infectious Diseases Research Group, St George’s University of London, London, United Kingdom
| | - Chiara Azzari
- Immunology Unit, Meyer Children’s Hospital IRCCS, Florence, Italy
- Department of Health Sciences, University of Florence, Florence, Italy
| |
Collapse
|
14
|
Mataramvura H, Bunders MJ, Duri K. Human immunodeficiency virus and antiretroviral therapy-mediated immune cell metabolic dysregulation in children born to HIV-infected women: potential clinical implications. Front Immunol 2023; 14:1182217. [PMID: 37350953 PMCID: PMC10282157 DOI: 10.3389/fimmu.2023.1182217] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 05/25/2023] [Indexed: 06/24/2023] Open
Abstract
Commencing lifelong antiretroviral therapy (ART) immediately following HIV diagnosis (Option B+) has dramatically improved the health of HIV-infected women and their children, with the majority being of HIV-exposed children born uninfected (HEU). This success has led to an increasing population of HIV-infected women receiving ART during pregnancy and children exposed to ART in utero. Nonetheless, a small proportion of children are still infected with HIV (HEI) each year. HEI children suffer from reduced immunocompetence and host-defence, due to CD4+ T lymphocyte depletion, but also dysregulation of other immune cells including CD8+ T lymphocytes, natural killer (NK) cells, macrophages including B lymphocytes. Furthermore, although HEU children are uninfected, altered immune responses are observed and associated with increased vulnerability to infections. The mechanisms underlying immune dysregulation in HEU children remain poorly described. Building on early studies, emerging data suggests that HIV/ART exposure early in life affects cell metabolic function of HEU children. Prenatal HIV/ART exposure has been associated with dysregulation of mitochondria, including impaired DNA polymerase activity. Furthermore, dysregulation of oxidative phosphorylation (OXPHOS) causes a decreased generation of adenosine triphosphate (ATP) and increased production of reactive oxygen species (ROS), resulting in oxidative stress. These altered metabolic processes can affect immune cell viability and immune responses. Recent studies have indicated that immune-metabolic dysregulation may contribute to HIV-associated pathogenesis and clinical observations associated with HIV and ART exposure in HEU/HEI children. Given the critical role metabolic processes in immune cell functioning, immune-metabolic dysregulation in HEU and HEI children may have implications in effective host-defence responses against pathogens, as well as efficacy of standard ART regimens and future novel HIV cure approaches in HEI children. At the same time, targeting metabolic pathways of immune cells may provide safer and novel approaches for HIV cure strategies. Here, we review the current literature investigating immune-metabolic dysregulation in paediatric HIV pathogenesis.
Collapse
Affiliation(s)
- Hope Mataramvura
- Immunology Unit, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| | - Madeleine J. Bunders
- III. Medical Department, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- Department of Virus Immunology, Leibniz Institute of Virology, Hamburg, Germany
| | - Kerina Duri
- Immunology Unit, University of Zimbabwe Faculty of Medicine and Health Sciences (UZ-FMHS), Harare, Zimbabwe
| |
Collapse
|
15
|
Gurbuz F, Canbulat Sahiner N, Unal E, Gurbuz AS, Baysal T. Breastfeeding does not protect against the development of carditis in children with acute rheumatic fever. Cardiol Young 2023; 33:899-903. [PMID: 35718952 DOI: 10.1017/s1047951122001834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Acute rheumatic fever is an autoimmune disease that develops due to streptococcal infection. The positive effect of breastfeeding on the development of the child's immune system is well documented. In this study, we aimed to investigate the effect of breast milk intake period on the development of carditis. MATERIALS AND METHODS Patients (n: 182) who were diagnosed with acute rheumatic fever between 2010 and 2019 were enrolled in the study. The patients were divided into groups according to carditis development. The demographic, socio-economic, and breastfeeding data were compared between groups. RESULTS The mean age of the patients was 10.5 ± 3.4, and 43.4 % (n: 79) of them were female. Independent predictors of the development of carditis in the first acute rheumatic fever episode were the number of children at home (OR: 1.773, CI 95%: 1.105, 2.845; p: 0.018) and breast milk intake less than 6 months (OR: 0.404, CI 95%: 0.174, 0.934; p: 0.034). Independent predictors of the development of carditis in any of the acute rheumatic fever episodes were the number of children at home (OR: 1.858, CI 95%: 1.100, 3.137; p: 0.021) and female gender (OR: 3.504, CI 95%: 1.227, 10.008; p: 0.019). The only independently predictor of the development of chorea during acute rheumatic fever was female gender (OR: 3.801, CI 95%: 1.463, 9.874; p: 0.006). CONCLUSION Although the occurrence of carditis is less common during the first acute rheumatic fever attack in patients with breast milk intake less than six months, this advantage is lost in recurrent attacks. This study showed that breast milk does not have a negative effect on acute rheumatic fever carditis.
Collapse
Affiliation(s)
- Funda Gurbuz
- Department of Nursing, Karamanoglu Mehmetbey University Health Sciences Institute, Karaman, Turkey
| | - Nejla Canbulat Sahiner
- Department of Nursing, Karamanoglu Mehmetbey University Health Sciences Institute, Karaman, Turkey
| | - Emin Unal
- Department of Pediatric Cardiology, Necmettin Erbakan University, Meram Medicine Faculty, Konya, Turkey
| | - Ahmet Seyfeddin Gurbuz
- Department of Cardiology, Necmettin Erbakan University, Meram Medicine Faculty, Konya, Turkey
| | - Tamer Baysal
- Department of Pediatric Cardiology, Necmettin Erbakan University, Meram Medicine Faculty, Konya, Turkey
| |
Collapse
|
16
|
Burger S, Stenger T, Pierson M, Sridhar A, Huggins MA, Kucaba TA, Griffith TS, Hamilton SE, Schuldt NJ. Natural Microbial Exposure from the Earliest Natural Time Point Enhances Immune Development by Expanding Immune Cell Progenitors and Mature Immune Cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 210:1740-1751. [PMID: 37074206 PMCID: PMC10192123 DOI: 10.4049/jimmunol.2300061] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 03/24/2023] [Indexed: 04/20/2023]
Abstract
Microbial experience fundamentally shapes immunity, particularly during the perinatal period when the immune system is underdeveloped, and novel microbial encounters are common. Most animal models are raised in specific pathogen-free (SPF) conditions with relatively uniform microbial communities. How SPF housing conditions alter early-life immune development relative to natural microbial exposure (NME) has not been thoroughly investigated. In this article, we compare immune development in SPF-raised mice with mice born from immunologically experienced mothers in microbially diverse environments. NME induced broad immune cell expansion, including naive cells, suggesting mechanisms besides activation-induced proliferation contribute to the increase in immune cell numbers. We found NME conditions also expanded immune cell progenitor cell populations in the bone marrow, suggesting microbial experience enhances immune development at the earliest stages of immune cell differentiation. Multiple immune functions characteristically impaired in infants were also enhanced by NME, including T cell memory and Th1 polarization, B cell class switching and Ab production, proinflammatory cytokine expression, and bacterial clearance after Listeria monocytogenes challenge. Collectively, our studies reveal numerous impairments in immune development in SPF conditions relative to natural immune development.
Collapse
Affiliation(s)
- Sarah Burger
- Center for Immunology, Minneapolis, MN, USA
- Department of Pediatrics, Minneapolis, MN, USA
| | - Terran Stenger
- Center for Immunology, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, Minneapolis, MN, USA
| | - Mark Pierson
- Center for Immunology, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, Minneapolis, MN, USA
| | - Adhvaith Sridhar
- Center for Immunology, Minneapolis, MN, USA
- Department of Pediatrics, Minneapolis, MN, USA
| | - Matthew A. Huggins
- Center for Immunology, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, Minneapolis, MN, USA
| | | | - Thomas S. Griffith
- Center for Immunology, Minneapolis, MN, USA
- Department of Urology, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology PhD Program, Minneapolis, MN, USA
- Masonic Cancer Center, Minneapolis, MN, USA
- Minneapolis VA Health Care System, Minneapolis, MN, USA
| | - Sara E. Hamilton
- Center for Immunology, Minneapolis, MN, USA
- Department of Laboratory Medicine and Pathology, Minneapolis, MN, USA
- Microbiology, Immunology, and Cancer Biology PhD Program, Minneapolis, MN, USA
- Masonic Cancer Center, Minneapolis, MN, USA
| | - Nathaniel J. Schuldt
- Center for Immunology, Minneapolis, MN, USA
- Department of Pediatrics, Minneapolis, MN, USA
- Masonic Cancer Center, Minneapolis, MN, USA
- Lead Contact
| |
Collapse
|
17
|
Baloh CH, Venturi GM, Fischer BM, Sadder LS, Kim-Chang JJ, Chan C, De Paris K, Yin L, Aldrovandi GM, Goodenow MM, Sleasman JW. Biomarkers detected in cord blood predict vaccine responses in young infants. Front Immunol 2023; 14:1152538. [PMID: 37251388 PMCID: PMC10213698 DOI: 10.3389/fimmu.2023.1152538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Introduction Factors influencing vaccine immune priming in the first year of life involve both innate and adaptive immunity but there are gaps in understanding how these factors sustain vaccine antibody levels in healthy infants. The hypothesis was that bioprofiles associated with B cell survival best predict sustained vaccine IgG levels at one year. Methods Longitudinal study of plasma bioprofiles in 82 term, healthy infants, who received standard recommended immunizations in the United States, with changes in 15 plasma biomarker concentrations and B cell subsets associated with germinal center development monitored at birth, soon after completion of the initial vaccine series at 6 months, and prior to the 12-month vaccinations. Post vaccination antibody IgG levels to Bordetella pertussis, tetanus toxoid, and conjugated Haemophilus influenzae type B (HiB) were outcome measures. Results Using a least absolute shrinkage and selection operator (lasso) regression model, cord blood (CB) plasma IL-2, IL-17A, IL-31, and soluble CD14 (sCD14) were positively associated with pertussis IgG levels at 12 months, while CB plasma concentrations of APRIL and IL-33 were negatively associated. In contrast, CB concentrations of sCD14 and APRIL were positively associated with sustained tetanus IgG levels. A separate cross-sectional analysis of 18 mother/newborn pairs indicated that CB biomarkers were not due to transplacental transfer, but rather due to immune activation at the fetal/maternal interface. Elevated percentages of cord blood switched memory B cells were positively associated with 12-month HiB IgG levels. BAFF concentrations at 6 and 12 months were positively associated with pertussis and HiB IgG levels respectively. Discussion Sustained B cell immunity is highly influenced by early life immune dynamics beginning prior to birth. The findings provide important insights into how germinal center development shapes vaccine responses in healthy infants and provide a foundation for studies of conditions that impair infant immune development.
Collapse
Affiliation(s)
- Carolyn H. Baloh
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Guglielmo M. Venturi
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Bernard M. Fischer
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Liane S. Sadder
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Julie J. Kim-Chang
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| | - Cliburn Chan
- Department of Biostatistics and Bioinformatics, Duke University School of Medicine, Durham, NC, United States
| | - Kristina De Paris
- Institute of Global Health and Infectious Diseases, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
- Department of Microbiology and Immunology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, NC, United States
| | - Li Yin
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - Grace M. Aldrovandi
- Division of Infectious Diseases, Department of Pediatrics, University of California, Los Angeles, CA, United States
| | - Maureen M. Goodenow
- Molecular HIV Host Interactions Section, National Institute of Allergy and Infectious Diseases, Bethesda, MD, United States
| | - John W. Sleasman
- Division of Allergy and Immunology, Department of Pediatrics, Duke University School of Medicine, Durham, NC, United States
| |
Collapse
|
18
|
Diethelm-Varela B, Soto JA, Riedel CA, Bueno SM, Kalergis AM. New Developments and Challenges in Antibody-Based Therapies for the Respiratory Syncytial Virus. Infect Drug Resist 2023; 16:2061-2074. [PMID: 37063935 PMCID: PMC10094422 DOI: 10.2147/idr.s379660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2023] [Accepted: 03/29/2023] [Indexed: 04/18/2023] Open
Abstract
Since the discovery of the human respiratory syncytial virus (hRSV), multiple research efforts have been conducted to develop vaccines and treatments capable of reducing the risk of severe disease, hospitalization, long-term sequelae, and death from this pathogen in susceptible populations. In this sense, therapies specifically directed against hRSV are mainly based on monoclonal and polyclonal antibodies such as intravenous IgG (IVIG)-RSV and the monoclonal antibody palivizumab. However, these therapies are associated with significant limitations, including the need for the recruitment of a high number of convalescent volunteers who donate blood to procure IVIG-RSV and the costs associated with the need for repeated administrations of palivizumab. These limitations render this product not cost-effective for populations other than high-risk patients. These problems have underscored that it is still necessary to identify new safe and effective therapies for human use. However, these new therapies must benefit from a comparatively cheap production cost and the opportunity to be available to the high-risk population and anyone who requires treatment. Here, we review the different antibodies used to prevent the pathology caused by hRSV infection, highlighting therapies currently approved for human use and their clinical value. Also, the new, most promising candidates based on preclinical studies and clinical trial results are revised.
Collapse
Affiliation(s)
- Benjamín Diethelm-Varela
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Jorge A Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Claudia A Riedel
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago, Chile
| | - Susan M Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Alexis M Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
19
|
Zhu J, Naughton S, Bowman N, LeRoith T, Luo X, Leeth C. Maternal antibody repertoire restriction modulates the development of lupus-like disease in BXSB offspring. Int Immunol 2023; 35:95-104. [PMID: 36190342 DOI: 10.1093/intimm/dxac049] [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: 05/28/2022] [Accepted: 10/01/2022] [Indexed: 11/13/2022] Open
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disease that has a strong preference for women of child-bearing age. Maternal factors play an essential role in shaping the immune system of the newborn, yet it is unknown whether maternal factors could modulate the development of SLE in the offspring. Activation-induced cytidine deaminase (AID) is an enzyme required for somatic hypermutation and class switch recombination. Given that IgG and IgA isotypes account for the vast majority of passive immunity in rodents, our previously established AID-deficient BXSB mice provide a model in which maternal antibodies that can be transferred to the offspring are greatly diminished and have restricted repertoire. In this study, we compared genotypically identical mice born to either AID-sufficient dams or AID-deficient dams and evaluated the effects of maternal antibodies in disease progression. Offspring from knockout dams developed disease at a faster rate, as shown by more severe nephritis and elevated pathogenic autoantibodies compared to their counterparts born to wild-type dams. When immune competent pups were cross fostered onto AID-deficient dams, these mice exhibited more severe disease characteristics, including exacerbated lupus nephritis, increased levels of circulating antinuclear antibodies, and more activated T cells. These results suggest that a protective antibody effect contributes to the modulation of SLE progression in postnatal period. Overall, these findings highlight the importance of maternal antibodies in programming the immune system and altering SLE development in offspring.
Collapse
Affiliation(s)
- Jing Zhu
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, 3280 Litton Reaves Hall, Blacksburg, VA 24061, USA.,Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Samantha Naughton
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, 3280 Litton Reaves Hall, Blacksburg, VA 24061, USA
| | - Nina Bowman
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, 3280 Litton Reaves Hall, Blacksburg, VA 24061, USA
| | - Tanya LeRoith
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Xin Luo
- Department of Biomedical Sciences and Pathobiology, Virginia Polytechnic Institute and State University, Blacksburg, VA, USA
| | - Caroline Leeth
- Department of Animal and Poultry Sciences, Virginia Tech, 175 West Campus Drive, 3280 Litton Reaves Hall, Blacksburg, VA 24061, USA
| |
Collapse
|
20
|
Stoddard CI, Sung K, Yaffe ZA, Weight H, Beaudoin-Bussières G, Galloway J, Gantt S, Adhiambo J, Begnel ER, Ojee E, Slyker J, Wamalwa D, Kinuthia J, Finzi A, Matsen FA, Lehman DA, Overbaugh J. Elevated binding and functional antibody responses to SARS-CoV-2 in infants versus mothers. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.06.527330. [PMID: 36798400 PMCID: PMC9934573 DOI: 10.1101/2023.02.06.527330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
Abstract
Infant antibody responses to viral infection can differ from those in adults. However, data on the specificity and function of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antibodies in infants, and direct comparisons between infants and adults are limited. We characterized antibody binding and functionality in convalescent plasma from postpartum women and their infants infected with SARS-CoV-2 from a vaccine-naïve prospective cohort in Nairobi, Kenya. Antibody titers against SARS-CoV-2 Spike, receptor binding domain and N-terminal domain, and Spike-expressing cell-surface staining levels were significantly higher in infants than in mothers. Plasma antibodies from mothers and infants bound to similar regions of the Spike S2 subunit, including the fusion peptide (FP) and stem helix-heptad repeat 2. However, infants displayed higher antibody levels and more consistent antibody escape pathways in the FP region compared to mothers. Finally, infants had significantly higher levels of antibody-dependent cellular cytotoxicity (ADCC), though, surprisingly, neutralization titers between infants and mothers were similar. These results suggest infants develop distinct SARS-CoV-2 binding and functional antibody repertoires and reveal age-related differences in humoral immunity to SARS-CoV-2 infection that could be relevant to protection and COVID-19 disease outcomes.
Collapse
Affiliation(s)
| | - Kevin Sung
- Public Health Sciences Division, Fred Hutchinson Cancer Center
| | - Zak A Yaffe
- Human Biology Division, Fred Hutchinson Cancer Center
- Medical Scientist Training Program, University of Washington
| | - Haidyn Weight
- Human Biology Division, Fred Hutchinson Cancer Center
| | - Guillaume Beaudoin-Bussières
- Centre de Recherche du CHUM, Université de Montréal
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal
| | - Jared Galloway
- Public Health Sciences Division, Fred Hutchinson Cancer Center
| | - Soren Gantt
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal
- Centre de Recherche du CHU Sainte-Justine, Université de Montréal
| | - Judith Adhiambo
- Department of Pediatrics and Child Health, University of Nairobi
| | | | - Ednah Ojee
- Department of Pediatrics and Child Health, University of Nairobi
| | | | - Dalton Wamalwa
- Department of Pediatrics and Child Health, University of Nairobi
| | - John Kinuthia
- Department of Global Health, University of Washington
- Department of Research and Programs, Kenyatta National Hospital
| | - Andrés Finzi
- Centre de Recherche du CHUM, Université de Montréal
- Département de Microbiologie, Infectiologie et Immunologie, Université de Montréal
| | - Frederick A Matsen
- Public Health Sciences Division, Fred Hutchinson Cancer Center
- Howard Hughes Medical Institute
| | - Dara A Lehman
- Human Biology Division, Fred Hutchinson Cancer Center
- Department of Global Health, University of Washington
| | - Julie Overbaugh
- Human Biology Division, Fred Hutchinson Cancer Center
- Public Health Sciences Division, Fred Hutchinson Cancer Center
- Lead contact
| |
Collapse
|
21
|
Presa J, Serra L, Weil-Olivier C, York L. Preventing invasive meningococcal disease in early infancy. Hum Vaccin Immunother 2022; 18:1979846. [PMID: 35482946 PMCID: PMC9196819 DOI: 10.1080/21645515.2021.1979846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
This review considers the pathogenesis, diagnosis, and epidemiology of invasive meningococcal disease in infants, to examine and critique meningococcal disease prevention in this population through vaccination. High rates of meningococcal disease and poor outcomes, particularly for very young infants, highlight the importance of meningococcal vaccination in early infancy. Although effective and safe meningococcal vaccines are available for use from 6 weeks of age, they are not recommended globally. Emerging real-world data from the increased incorporation of these vaccines within immunization programs inform recommendations regarding effectiveness, appropriate vaccination schedule, possible long-term safety effects, and persistence of antibody responses. Importantly, to protect infants from IMD, national vaccination recommendations should be consistent with available data regarding vaccine safety, effectiveness, and disease risk.
Collapse
Affiliation(s)
- Jessica Presa
- Vaccine Medical, Development, Scientific, and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | - Lidia Serra
- Global Vaccines Medical Development and Scientific, and Clinical Affairs, Pfizer Inc, Collegeville, PA, USA
| | | | - Laura York
- York Biologics Consulting LLC, Wayne, PA, USA
| |
Collapse
|
22
|
Pieren DKJ, Boer MC, de Wit J. The adaptive immune system in early life: The shift makes it count. Front Immunol 2022; 13:1031924. [PMID: 36466865 PMCID: PMC9712958 DOI: 10.3389/fimmu.2022.1031924] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 10/31/2022] [Indexed: 10/13/2023] Open
Abstract
Respiratory infectious diseases encountered early in life may result in life-threatening disease in neonates, which is primarily explained by the relatively naive neonatal immune system. Whereas vaccines are not readily available for all infectious diseases, vaccinations have greatly reduced childhood mortality. However, repeated vaccinations are required to reach protective immunity in infants and not all vaccinations are effective at young age. Moreover, protective adaptive immunity elicited by vaccination wanes more rapidly at young age compared to adulthood. The infant adaptive immune system has previously been considered immature but this paradigm has changed during the past years. Recent evidence shows that the early life adaptive immune system is equipped with a strong innate-like effector function to eliminate acute pathogenic threats. These strong innate-like effector capacities are in turn kept in check by a tolerogenic counterpart of the adaptive system that may have evolved to maintain balance and to reduce collateral damage. In this review, we provide insight into these aspects of the early life's adaptive immune system by addressing recent literature. Moreover, we speculate that this shift from innate-like and tolerogenic adaptive immune features towards formation of immune memory may underlie different efficacy of infant vaccination in these different phases of immune development. Therefore, presence of innate-like and tolerogenic features of the adaptive immune system may be used as a biomarker to improve vaccination strategies against respiratory and other infections in early life.
Collapse
Affiliation(s)
| | | | - Jelle de Wit
- Centre for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
23
|
Strandmark J, Darboe A, Diray-Arce J, Ben-Othman R, Vignolo SM, Rao S, Smolen KK, Leroux-Roels G, Idoko OT, Sanchez-Schmitz G, Ozonoff A, Levy O, Kollmann TR, Marchant A, Kampmann B. A single birth dose of Hepatitis B vaccine induces polyfunctional CD4 + T helper cells. Front Immunol 2022; 13:1043375. [PMID: 36426360 PMCID: PMC9681035 DOI: 10.3389/fimmu.2022.1043375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 10/21/2022] [Indexed: 04/13/2024] Open
Abstract
A single birth-dose of Hepatitis B vaccine (HepB) can protect newborns from acquiring Hepatitis B infection through vertical transmission, though several follow-up doses are required to induce long-lived protection. In addition to stimulating antibodies, a birth-dose of HepB might also induce polyfunctional CD4+ T-cells, which may contribute to initial protection. We investigated whether vaccination with HepB in the first week of life induced detectable antigen-specific CD4+ T-cells after only a single dose and following completion of the entire HepB vaccine schedule (3 doses). Using HBsAg- stimulated peripheral blood mononuclear cells from 344 infants, we detected increased populations of antigen-specific polyfunctional CD154+IL-2+TNFα+ CD4+ T-cells following a single birth-dose of HepB in a proportion of infants. Frequencies of polyfunctional T-cells increased following the completion of the HepB schedule but increases in the proportion of responders as compared to following only one dose was marginal. Polyfunctional T-cells correlated positively with serum antibody titres following the birth dose (day30) and completion of the 3-dose primary HepB vaccine series (day 128). These data indicate that a single birth dose of HepB provides immune priming for both antigen-specific B- and T cells.
Collapse
Affiliation(s)
- Julia Strandmark
- Vaccines & Immunity Theme, Medical Research Council (MRC) Unit The Gambia at London School of Hygiene & Tropical Medicine (LSHTM), Fajara, Gambia
| | - Alansana Darboe
- Vaccines & Immunity Theme, Medical Research Council (MRC) Unit The Gambia at London School of Hygiene & Tropical Medicine (LSHTM), Fajara, Gambia
| | - Joann Diray-Arce
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Rym Ben-Othman
- Department of Paediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Sofia M. Vignolo
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
| | - Shun Rao
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
| | - Kinga K. Smolen
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | | | - Olubukola T. Idoko
- Vaccines & Immunity Theme, Medical Research Council (MRC) Unit The Gambia at London School of Hygiene & Tropical Medicine (LSHTM), Fajara, Gambia
| | - Guzmán Sanchez-Schmitz
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
| | - Al Ozonoff
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Klarman Cell Observatory & Global Health Initiative, Broad Institute of the Massachusetts Institute of Technology (MIT) & Harvard, Cambridge, MA, United States
| | - Ofer Levy
- Precision Vaccines Program, Boston Children’s Hospital, Boston, MA, United States
- Department of Pediatrics, Harvard Medical School, Boston, MA, United States
- Klarman Cell Observatory & Global Health Initiative, Broad Institute of the Massachusetts Institute of Technology (MIT) & Harvard, Cambridge, MA, United States
| | - Tobias R. Kollmann
- Department of Paediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Brussels, Belgium
| | - Beate Kampmann
- Vaccines & Immunity Theme, Medical Research Council (MRC) Unit The Gambia at London School of Hygiene & Tropical Medicine (LSHTM), Fajara, Gambia
- The Vaccine Centre, London School of Hygiene and Tropical Medicine, London, United Kingdom
| |
Collapse
|
24
|
Adherence to Recommendations for Βacillus Calmette-Guérin Vaccination of High-risk Neonates in Greece. Pediatr Infect Dis J 2022; 41:857-859. [PMID: 35763676 DOI: 10.1097/inf.0000000000003623] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
In 2016 a Βacillus Calmette-Guérin vaccination policy targeting high-risk neonates for tuberculosis before discharge from maternity hospital was adopted in Greece. Vaccination rates were 38.2% in 2019 and 24.7% in 2020. Vaccination coverage varied by risk group (higher for neonates in close contact with an active noncompliant or multidrug-resistant tuberculosis case and lower for Roma and immigrant neonates).
Collapse
|
25
|
Breastfeeding: The Basics, the History, and Barriers in the Modern Day. Obstet Gynecol Surv 2022; 77:423-432. [DOI: 10.1097/ogx.0000000000001041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
26
|
Mutanga JN, Whitaker BI, Forshee RA. Regulatory considerations for study of infant protection through maternal immunization. Vaccine 2022; 40:3556-3565. [PMID: 35570075 DOI: 10.1016/j.vaccine.2022.04.087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 03/29/2022] [Accepted: 04/27/2022] [Indexed: 11/17/2022]
Abstract
Childhood Immunization is one of the critical strategies to decrease infant morbidity and mortality due to infectious diseases, but primary immunization schedules for infants in most countries start at 2 months of age. Childhood vaccines therefore begin providing adequate protection later in life, leaving infants vulnerable to infectious diseases and creating an immunity gap that results in higher morbidity and mortality among younger infants. Maternal immunization, the practice of vaccinating individuals during pregnancy, reduces the risk of infant infection primarily through the transfer of protective maternal antibodies to the fetus during late pregnancy. Although much progress has been made in public health policies to support maternal immunization research, inclusion of pregnant individuals and children in clinical trials remains challenging. This has resulted in paucity of evidence regarding safety and effectiveness of vaccines to support licensure of products intended for use during pregnancy and lactation to prevent disease in the infant. In addition, although safeguards for clinical research in pregnancy are supportive, experimental vaccines, e.g., Respiratory Syncytial Virus, are more complicated to study because data on safety, efficacy, and dosing are limited. This requires randomized controlled trials with safety monitoring for the mother, the fetus, and the infant with follow-up for at least 1 year or longer to assess long-term health outcomes that may be associated with peripartum vaccine exposure. The goal of this paper is to discuss the general regulatory considerations for clinical research to evaluate safety and effectiveness of vaccines administered during pregnancy to protect infants from disease. This could be useful to inform future vaccine trials. This discussion is not intended to provide agency guidance nor to articulate agency policy.
Collapse
Affiliation(s)
- Jane Namangolwa Mutanga
- US Food and Drug Administration, Center for Biologics Evaluation and Research, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA.
| | - Barbee I Whitaker
- US Food and Drug Administration, Center for Biologics Evaluation and Research, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| | - Richard A Forshee
- US Food and Drug Administration, Center for Biologics Evaluation and Research, 10903 New Hampshire Ave, Silver Spring, MD 20993, USA
| |
Collapse
|
27
|
Soto JA, Díaz FE, Retamal-Díaz A, Gálvez NMS, Melo-González F, Piña-Iturbe A, Ramírez MA, Bohmwald K, González PA, Bueno SM, Kalergis AM. BCG-Based Vaccines Elicit Antigen-Specific Adaptive and Trained Immunity against SARS-CoV-2 and Andes orthohantavirus. Vaccines (Basel) 2022; 10:vaccines10050721. [PMID: 35632475 PMCID: PMC9143576 DOI: 10.3390/vaccines10050721] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Revised: 04/24/2022] [Accepted: 04/25/2022] [Indexed: 01/27/2023] Open
Abstract
Background:Mycobacterium bovis Bacillus Calmette-Guérin (BCG) is a live attenuated vaccine mainly administered to newborns and used for over 100 years to prevent the disease caused by Mycobacterium tuberculosis (M. tb). This vaccine can induce immune response polarization towards a Th1 profile, which is desired for counteracting M. tb, other mycobacteria, and unrelated intracellular pathogens. The vaccine BCG has been used as a vector to express recombinant proteins and has been shown to protect against several diseases, particularly respiratory viruses. Methods: BCG was used to develop recombinant vaccines expressing either the Nucleoprotein from SARS-CoV-2 or Andes orthohantavirus. Mice were immunized with these vaccines with the aim of evaluating the safety and immunogenicity parameters. Results: Immunization with two doses of 1 × 108 CFU or one dose of 1 × 105 CFU of these BCGs was safe in mice. A statistically significant cellular immune response was induced by both formulations, characterized as the activation of CD4+ and CD8+ T cells. Stimulation with unrelated antigens resulted in increased expression of activation markers by T cells and secretion of IL-2 and IFN-γ, while increased secretion of IL-6 was found for both recombinant vaccines; all of these parameters related to a trained immunity profile. The humoral immune response elicited by both vaccines was modest, but further exposure to antigens could increase this response. Conclusions: The BCG vaccine is a promising platform for developing vaccines against different pathogens, inducing a marked antigen-specific immune response.
Collapse
Affiliation(s)
- Jorge A. Soto
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 7550196, Chile
| | - Fabián E. Díaz
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Angello Retamal-Díaz
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Nicolás M. S. Gálvez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Felipe Melo-González
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
- Millennium Institute on Immunology and Immunotherapy, Departamento de Ciencias Biológicas, Facultad de Ciencias de la Vida, Universidad Andrés Bello, Santiago 7550196, Chile
| | - Alejandro Piña-Iturbe
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Mario A. Ramírez
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Karen Bohmwald
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Pablo A. González
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Susan M. Bueno
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
| | - Alexis M. Kalergis
- Millennium Institute of Immunology and Immunotherapy, Departamento de Genética Molecular y Microbiología, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Av. Portugal 49, Santiago 8320000, Chile; (J.A.S.); (F.E.D.); (A.R.-D.); (N.M.S.G.); (F.M.-G.); (A.P.-I.); (M.A.R.); (K.B.); (P.A.G.); (S.M.B.)
- Departamento de Endocrinología, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago 8320000, Chile
- Correspondence: or ; Tel.: +56-2-686-2842
| |
Collapse
|
28
|
Xie D, Niu Y, Mu R, Campos de Souza S, Yin X, Dong L, Wang C. A Toll-like Receptor-Activating, Self-Adjuvant Glycan Nanocarrier. Front Chem 2022; 10:864206. [PMID: 35592309 PMCID: PMC9110926 DOI: 10.3389/fchem.2022.864206] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/31/2022] [Indexed: 11/13/2022] Open
Abstract
The global pandemic of COVID-19 highlights the importance of vaccination, which remains the most efficient measure against many diseases. Despite the progress in vaccine design, concerns with suboptimal antigen immunogenicity and delivery efficiency prevail. Self-adjuvant carriers–vehicles that can simultaneously deliver antigens and act as adjuvants–may improve efficacies in these aspects. Here, we developed a self-adjuvant carrier based on an acetyl glucomannan (acGM), which can activate toll-like receptor 2 (TLR2) and encapsulate the model antigen ovalbumin (OVA) via a double-emulsion process. In vitro tests showed that these OVA@acGM-8k nanoparticles (NPs) enhanced cellular uptake and activated TLR2 on the surface of dendritic cells (DCs), with increased expression of co-stimulatory molecules (e.g. CD80 and CD86) and pro-inflammatory cytokines (e.g. TNF-α and IL12p70). In vivo experiments in mice demonstrated that OVA@acGM-8k NPs accumulated in the lymph nodes and promoted DCs’ maturation. The immunization also boosted the humoral and cellular immune responses. Our findings suggest that this self-adjuvant polysaccharide carrier could be a promising approach for vaccine development.
Collapse
Affiliation(s)
- Daping Xie
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Yiming Niu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Ruoyu Mu
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Senio Campos de Souza
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
| | - Xiaoyu Yin
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Lei Dong
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- *Correspondence: Chunming Wang, ; Lei Dong,
| | - Chunming Wang
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Taipa, Macao SAR, China
- *Correspondence: Chunming Wang, ; Lei Dong,
| |
Collapse
|
29
|
Impact of maternal and pre-existing antibodies on immunogenicity of inactivated rotavirus vaccines. Vaccine 2022; 40:3843-3850. [DOI: 10.1016/j.vaccine.2022.05.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 04/26/2022] [Accepted: 05/11/2022] [Indexed: 11/20/2022]
|
30
|
Benedicto-Matambo P, Bines JE, Malamba-Banda C, Shawa IT, Barnes K, Kamng’ona AW, Hungerford D, Jambo KC, Iturriza-Gomara M, Cunliffe NA, Flanagan KL, Jere KC. Leveraging Beneficial Off-Target Effects of Live-Attenuated Rotavirus Vaccines. Vaccines (Basel) 2022; 10:418. [PMID: 35335050 PMCID: PMC8948921 DOI: 10.3390/vaccines10030418] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Revised: 03/03/2022] [Accepted: 03/04/2022] [Indexed: 12/14/2022] Open
Abstract
Following the introduction of live-attenuated rotavirus vaccines in many countries, a notable reduction in deaths and hospitalisations associated with diarrhoea in children <5 years of age has been reported. There is growing evidence to suggest that live-attenuated vaccines also provide protection against other infections beyond the vaccine-targeted pathogens. These so called off-target effects of vaccination have been associated with the tuberculosis vaccine Bacille Calmette Guérin (BCG), measles, oral polio and recently salmonella vaccines, and are thought to be mediated by modified innate and possibly adaptive immunity. Indeed, rotavirus vaccines have been reported to provide greater than expected reductions in acute gastroenteritis caused by other enteropathogens, that have mostly been attributed to herd protection and prior underestimation of rotavirus disease. Whether rotavirus vaccines also alter the immune system to reduce non targeted gastrointestinal infections has not been studied directly. Here we review the current understanding of the mechanisms underlying off-target effects of vaccines and propose a mechanism by which the live-attenuated neonatal rotavirus vaccine, RV3-BB, could promote protection beyond the targeted pathogen. Finally, we consider how vaccine developers may leverage these properties to improve health outcomes in children, particularly those in low-income countries where disease burden and mortality is disproportionately high relative to developed countries.
Collapse
Affiliation(s)
- Prisca Benedicto-Matambo
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, Faculty of Biomedical Sciences and Health Professions, College of Medicine, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Julie E. Bines
- Enteric Diseases Group, Murdoch Children’s Research Institute, Department of Gastroenterology and Clinical Nutrition, Royal Children’s Hospital and Department of Paediatrics, The University of Melbourne, Parkville, VIC 3052, Australia;
| | - Chikondi Malamba-Banda
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, Faculty of Biomedical Sciences and Health Professions, College of Medicine, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- Department of Biological Sciences, Academy of Medical Sciences, Malawi University of Science and Technology, Blantyre 312225, Malawi
| | - Isaac T. Shawa
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Department of Medical Laboratory Sciences, Faculty of Biomedical Sciences and Health Professions, College of Medicine, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Kayla Barnes
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Harvard School of Public Health, Boston, MA 02115, USA
| | - Arox W. Kamng’ona
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Department of Biomedical Sciences, Faculty of Biomedical Sciences and Health Profession, College of Medicine, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
| | - Daniel Hungerford
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Kondwani C. Jambo
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Department of Clinical Sciences, Liverpool School of Tropical Medicine, Liverpool L3 5QA, UK
| | - Miren Iturriza-Gomara
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
- Centre for Vaccine Innovation and Access, Program for Appropriate Technology in Health (PATH), 1218 Geneva, Switzerland
| | - Nigel A. Cunliffe
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| | - Katie L. Flanagan
- School of Medicine, University of Tasmania, Hobart, TAS 7005, Australia;
- School of Health and Biomedical Science, Royal Melbourne Institute of Technology (RMIT), Bundoora, VIC 3083, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, VIC 3004, Australia
| | - Khuzwayo C. Jere
- Virology Research Group, Malawi-Liverpool-Wellcome Trust Clinical Research Programme, Blantyre 312225, Malawi; (P.B.-M.); (C.M.-B.); (I.T.S.); (K.B.); (A.W.K.); (K.C.J.)
- Centre for Global Vaccine Research, Institute of Infection, Veterinary and Ecological Sciences, University of Liverpool, Liverpool L69 7BE, UK; (D.H.); (M.I.-G.); (N.A.C.)
- Department of Medical Laboratory Sciences, Faculty of Biomedical Sciences and Health Professions, College of Medicine, Kamuzu University of Health Sciences, Blantyre 312225, Malawi
- NIHR Health Protection Research Unit in Gastrointestinal Infections, University of Liverpool, Liverpool L69 7BE, UK
| |
Collapse
|
31
|
Toothaker JM, Olaloye O, McCourt BT, McCourt CC, Silva TN, Case RM, Liu P, Yimlamai D, Tseng G, Konnikova L. Immune landscape of human placental villi using single-cell analysis. Development 2022; 149:274057. [PMID: 35050308 PMCID: PMC8935213 DOI: 10.1242/dev.200013] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2021] [Accepted: 12/30/2021] [Indexed: 12/19/2022]
Abstract
Maintenance of a healthy pregnancy is reliant on a successful balance between the fetal and maternal immune systems. Although the maternal mechanisms responsible have been well studied, those used by the fetal immune system remain poorly understood. Using suspension mass cytometry and various imaging modalities, we report a complex immune system within the mid-gestation (17-23 weeks) human placental villi (PV). Consistent with recent reports in other fetal organs, T cells with memory phenotypes, although rare in abundance, were detected within the PV tissue and vasculature. Moreover, we determined that T cells isolated from PV samples may be more proliferative after T cell receptor stimulation than adult T cells at baseline. Collectively, we identified multiple subtypes of fetal immune cells within the PV and specifically highlight the enhanced proliferative capacity of fetal PV T cells.
Collapse
Affiliation(s)
- Jessica M. Toothaker
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA,Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | | | - Blake T. McCourt
- Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | - Collin C. McCourt
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15219, USA
| | - Tatiana N. Silva
- Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | - Rebecca M. Case
- Department of Pediatrics, Children's Hospital of Pittsburgh of UPMC, Pittsburgh, PA 15219, USA
| | - Peng Liu
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Dean Yimlamai
- Department of Pediatrics, Yale University, New Haven, CT 06510, USA
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Liza Konnikova
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA,Department of Pediatrics, Yale University, New Haven, CT 06510, USA,Department of Obstetrics, Gynecology and Reproductive Sciences, Yale University, New Haven, CT 06519, USA,Program in Human and Translational Immunology, Yale University, New Haven, CT 06519, USA,Author for correspondence ()
| |
Collapse
|
32
|
Walker K, Green J, Petty J, Whiting L, Staff L, Bromley P, Fowler C, Jones LK. Breastfeeding in the context of the COVID-19 pandemic: A discussion paper. JOURNAL OF NEONATAL NURSING : JNN 2022; 28:9-15. [PMID: 34366687 PMCID: PMC8332735 DOI: 10.1016/j.jnn.2021.08.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/09/2021] [Revised: 06/24/2021] [Accepted: 08/02/2021] [Indexed: 01/03/2023]
Abstract
Breastfeeding offers one of the most fundamental global health benefits for babies. Breastmilk is lifesaving, providing not only nutrition but immunologic benefits and as such is strongly supported by the World Health Organization and leading healthcare associations worldwide. When the COVID-19 pandemic started in 2020, the impact of the restrictions to prevent the spread of the disease created challenges and questions about provision of safe, quality care, including breastfeeding practices, in a new 'normal' environment. Mothers were temporarily separated from their babies where infection was present or suspected, parents were prevented from being present on neonatal units and vital breastfeeding support was prevented. This discussion paper provides an overview of essential areas of knowledge related to practice for neonatal nurses and midwives who care for breastfeeding mothers and babies, in the context of the COVID-19 pandemic and the latest global guidance. Three areas will be discussed; the protective benefits of breastfeeding, keeping breastfeeding mothers and babies together and supporting mothers to breastfeed their babies. Finally, care recommendations are presented to serve as a summary of key points for application to practice for neonatal nurses and midwives.
Collapse
Affiliation(s)
| | - Janet Green
- School of Nursing, College of Health and Medicine, University of Tasmania, Australia
| | - Julia Petty
- School of Health and Social Work, The University of Hertfordshire, Hatfield, UK
| | - Lisa Whiting
- School of Health and Social Work, The University of Hertfordshire, Hatfield, UK
| | - Lynette Staff
- School of Nursing, College of Health and Medicine, University of Tasmania, Australia
| | - Patricia Bromley
- School of Nursing, College of Health and Medicine, University of Tasmania, Australia
| | - Cathrine Fowler
- Faculty of Health University of Technology, Sydney, Australia
| | - Linda K Jones
- School of Nursing, College of Health and Medicine, University of Tasmania, Australia
| |
Collapse
|
33
|
Shen CJ, Fu YC, Lin YP, Shen CF, Sun DJ, Chen HY, Cheng CM. Evaluation of Transplacental Antibody Transfer in SARS-CoV-2-Immunized Pregnant Women. Vaccines (Basel) 2022; 10:101. [PMID: 35062762 PMCID: PMC8778956 DOI: 10.3390/vaccines10010101] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection during pregnancy could result in adverse perinatal outcome. Clinical data on the assessment of the immune response in vaccinated pregnant women and subsequent transplacental antibody transfer are quite limited. OBJECTIVE To assess maternal and neonatal neutralizing antibody levels against both wildtype and Delta (B.1.617.2) variants after maternal mRNA vaccination. STUDY DESIGN This cohort study was conducted 29 pregnant women who were vaccinated at least one dose of Moderna (mRNA-1273) vaccine. Both neutralizing antibody (wildtype and Delta variant) and S1 receptor binding domain IgG antibody levels were evaluated in maternal and cord blood on the day of delivery. RESULTS Superiority of antibody level was significant in fully vaccinated women compared with the one-dose group (maternal sera, median, 97.46%; cord sera, median, 97.37% versus maternal sera, median, 4.01%; cord sera, median, 1.44%). No difference in antibody level was noted in relation to interval of second immunization to delivery in the two-dose group (95.99% in 0-2 weeks, 97.45% in 2-4 weeks, 97.48% in 4-8 weeks, 97.72% in 8-10 weeks). The most pronounced reduction was observed for the Delta variant. The wildtype neutralizing antibody level of full-vaccinated women was not influenced by the pertussis vaccination. CONCLUSION The data underscore the importance of full vaccination in pregnancy and support the recommendation of COVID-19 immunization for pregnant women. The lower level of vaccine-induced neutralizing antibodies for the Delta variant indicates insufficient protection for mother and newborn and highlights the need for development of effective vaccine strategies.
Collapse
Affiliation(s)
- Ching-Ju Shen
- Department of Obstetrics and Gynecology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 807, Taiwan
| | - Yi-Chen Fu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan; (Y.-C.F.); (Y.-P.L.)
| | - Yen-Pin Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan; (Y.-C.F.); (Y.-P.L.)
| | - Ching-Fen Shen
- Department of Pediatrics, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan 704, Taiwan;
| | - Der-Ji Sun
- Department of Obstetrics and Gynecology, Pojen Hospital, Kaohsiung 804, Taiwan;
| | - Huan-Yun Chen
- Department of Obstetrics and Gynecology, Kaohsiung Chang Gung Memorial Hospital, Kaohsiung 833, Taiwan;
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 300, Taiwan; (Y.-C.F.); (Y.-P.L.)
| |
Collapse
|
34
|
Kumar P. A Perspective on the Success and Failure of BCG. Front Immunol 2022; 12:778028. [PMID: 34970263 PMCID: PMC8712472 DOI: 10.3389/fimmu.2021.778028] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 11/18/2021] [Indexed: 11/13/2022] Open
Abstract
TB continues to be one of the major public health threats. BCG is the only available vaccine against TB and confers significant protection against the childhood disease. However, the protective efficacy of BCG against adult pulmonary TB, which represents a larger burden of disease, is highly variable. It has been suggested that prior exposure to environmental mycobacteria (EMb) mitigates the anti-TB efficacy of BCG by blocking its duplication or masking its immunogenicity. However, its effectiveness against childhood TB and failure of repeated administration to provide additional benefit against pulmonary TB, suggest of some other mechanisms for the variable efficacy of BCG against the pulmonary disease. Importantly, TB is a heterogeneous disease occurring in different forms and having distinct mechanisms of pathogenesis. While inability of the immune system to contain the bacilli is responsible for TB pathogenesis in infants, an aggravated immune response to Mtb has been blamed for the development of adult pulmonary TB. Available data suggest that EMb play a key role in heightening the immune response against Mtb. In this article, differential efficacy of BCG against childhood and adult TB is explained by taking into account the heterogeneity of TB, mechanisms of TB pathogenesis, and the effect of EMb on anti-Mtb immunity. It is believed that a refined understanding of the success and failure of BCG will help in the development of effective anti-TB vaccines.
Collapse
Affiliation(s)
- Pawan Kumar
- Department of Preventive Oncology, Dr. B. R. Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
35
|
Gabis LV, Attia OL, Goldman M, Barak N, Tefera P, Shefer S, Shaham M, Lerman-Sagie T. The myth of vaccination and autism spectrum. Eur J Paediatr Neurol 2022; 36:151-158. [PMID: 34996019 PMCID: PMC8694782 DOI: 10.1016/j.ejpn.2021.12.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 12/12/2021] [Accepted: 12/14/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Among all of the studied potential causes of autism, vaccines have received some of the most scrutiny and have been the topic of many evidence-based studies. These efforts have led the great majority of scientists, physicians, and public health researchers to refute causation between vaccines and autism. RATIONALE This presumed association and concern has been a major contributor to parents' refusal to immunize their children and has become a major threat to public health in secluded populations over the last two decades, even prior to the COVID-19 pandemic. With the emergence of COVID-19 immunizations, sentiments towards this topic were addressed as a public health concern that may influence the ability to overcome the Corona virus worldwide. SCIENTIFIC REVIEW OF DATA Despite the overwhelming data demonstrating that there is no link between vaccines and autism, many parents are hesitant to immunize their children because of the alleged association. Other contributing factors to the myths and conspiracy theories surrounding the association between vaccines and autism include the fact that the diagnosis of autism is typically made after the age of receiving the main childhood immunizations, as well as the occasional occurrence of regression after the age of first year vaccinations. In spite of vast evidence that the main contribution to the increase in incidence is from improvement of the diagnostic process, this rapid and publicized rise in autism diagnoses feeds parental concerns regarding any medical intervention that may be associated with the health of their children. RECOMMENDATIONS It is plausible that with more evidence-based studies linking autism to specific etiologies the myth will diminish and disappear eventually. In an era where conspiracy theories are prevalent on social media, it is critical that evidence-based studies relating autism to specific etiologies be made public, and that information concerning autism diagnosis and causes be made more readily available through social media and parental organizations.
Collapse
Affiliation(s)
- Lidia V Gabis
- Sackler School of Medicine at Tel Aviv University, Israel; Maccabi Health Services, Israel.
| | - Odelia Leon Attia
- Weinberg Developmental Center, at Safra Children's Hospital, Tel Hashomer, Israel.
| | - Mia Goldman
- Sackler School of Medicine at Tel Aviv University, Israel.
| | - Noy Barak
- Department of Industrial Engineering at Tel-Aviv University, Israel.
| | - Paula Tefera
- Sackler School of Medicine at Tel Aviv University, Israel.
| | - Shahar Shefer
- Weinberg Developmental Center, at Safra Children's Hospital, Tel Hashomer, Israel.
| | - Meirav Shaham
- Department of Statistics at University of Haifa, Haifa, Israel.
| | - Tally Lerman-Sagie
- Sackler School of Medicine at Tel Aviv University, Israel; Pediatric Neurology Unit at Wolfson Medical Center, Holon, Israel.
| |
Collapse
|
36
|
Otsuka KS, Nielson C, Firpo MA, Park AH, Beaudin AE. Early Life Inflammation and the Developing Hematopoietic and Immune Systems: The Cochlea as a Sensitive Indicator of Disruption. Cells 2021; 10:cells10123596. [PMID: 34944105 PMCID: PMC8700005 DOI: 10.3390/cells10123596] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/02/2021] [Accepted: 12/13/2021] [Indexed: 12/22/2022] Open
Abstract
Emerging evidence indicates that perinatal infection and inflammation can influence the developing immune system and may ultimately affect long-term health and disease outcomes in offspring by perturbing tissue and immune homeostasis. We posit that perinatal inflammation influences immune outcomes in offspring by perturbing (1) the development and function of fetal-derived immune cells that regulate tissue development and homeostasis, and (2) the establishment and function of developing hematopoietic stem cells (HSCs) that continually generate immune cells across the lifespan. To disentangle the complexities of these interlinked systems, we propose the cochlea as an ideal model tissue to investigate how perinatal infection affects immune, tissue, and stem cell development. The cochlea contains complex tissue architecture and a rich immune milieu that is established during early life. A wide range of congenital infections cause cochlea dysfunction and sensorineural hearing loss (SNHL), likely attributable to early life inflammation. Furthermore, we show that both immune cells and bone marrow hematopoietic progenitors can be simultaneously analyzed within neonatal cochlear samples. Future work investigating the pathogenesis of SNHL in the context of congenital infection will therefore provide critical information on how perinatal inflammation drives disease susceptibility in offspring.
Collapse
Affiliation(s)
- Kelly S. Otsuka
- Department of Pathology, University of Utah School of Medicine, Salt Lake City, UT 84112, USA;
| | - Christopher Nielson
- Division of Otolaryngology—Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (C.N.); (A.H.P.)
| | - Matthew A. Firpo
- Department of Surgery, University of Utah, Salt Lake City, UT 84112, USA;
| | - Albert H. Park
- Division of Otolaryngology—Head and Neck Surgery, University of Utah School of Medicine, Salt Lake City, UT 84112, USA; (C.N.); (A.H.P.)
| | - Anna E. Beaudin
- Division of Hematology and Hematologic Malignancies, University of Utah School of Medicine, Salt Lake City, UT 84112, USA
- Correspondence:
| |
Collapse
|
37
|
Zhu W, Wu Y, Liu H, Jiang C, Huo L. Gut-Lung Axis: Microbial Crosstalk in Pediatric Respiratory Tract Infections. Front Immunol 2021; 12:741233. [PMID: 34867963 PMCID: PMC8637285 DOI: 10.3389/fimmu.2021.741233] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 10/28/2021] [Indexed: 12/12/2022] Open
Abstract
The gut microbiota is an important regulator for maintaining the organ microenvironment through effects on the gut-vital organs axis. Respiratory tract infections are one of the most widespread and harmful diseases, especially in the last 2 years. Many lines of evidence indicate that the gut microbiota and its metabolites can be considered in therapeutic strategies to effectively prevent and treat respiratory diseases. However, due to the different gut microbiota composition in children compared to adults and the dynamic development of the immature immune system, studies on the interaction between children's intestinal flora and respiratory infections are still lacking. Here, we describe the changes in the gut microbiota of children with respiratory tract infections and explain the relationship between the microbiota of children with their immune function and disease development. In addition, we will provide perspectives on the direct manipulation of intestinal microbes to prevent or treat pediatric respiratory infections.
Collapse
Affiliation(s)
- Wenxia Zhu
- Shanghai Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yilin Wu
- Shanghai Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hui Liu
- Shanghai Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Caini Jiang
- Shanghai Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Lili Huo
- Shanghai Hospital of Integrated Traditional Chinese and Western Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
38
|
Choudhury SM, Ma X, Dang W, Li Y, Zheng H. Recent Development of Ruminant Vaccine Against Viral Diseases. Front Vet Sci 2021; 8:697194. [PMID: 34805327 PMCID: PMC8595237 DOI: 10.3389/fvets.2021.697194] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 10/04/2021] [Indexed: 01/21/2023] Open
Abstract
Pathogens of viral origin produce a large variety of infectious diseases in livestock. It is essential to establish the best practices in animal care and an efficient way to stop and prevent infectious diseases that impact animal husbandry. So far, the greatest way to combat the disease is to adopt a vaccine policy. In the fight against infectious diseases, vaccines are very popular. Vaccination's fundamental concept is to utilize particular antigens, either endogenous or exogenous to induce immunity against the antigens or cells. In light of how past emerging and reemerging infectious diseases and pandemics were handled, examining the vaccination methods and technological platforms utilized for the animals may provide some useful insights. New vaccine manufacturing methods have evolved because of developments in technology and medicine and our broad knowledge of immunology, molecular biology, microbiology, and biochemistry, among other basic science disciplines. Genetic engineering, proteomics, and other advanced technologies have aided in implementing novel vaccine theories, resulting in the discovery of new ruminant vaccines and the improvement of existing ones. Subunit vaccines, recombinant vaccines, DNA vaccines, and vectored vaccines are increasingly gaining scientific and public attention as the next generation of vaccines and are being seen as viable replacements to conventional vaccines. The current review looks at the effects and implications of recent ruminant vaccine advances in terms of evolving microbiology, immunology, and molecular biology.
Collapse
Affiliation(s)
- Sk Mohiuddin Choudhury
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - XuSheng Ma
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - Wen Dang
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| | - YuanYuan Li
- Gansu Agricultural University, Lanzhou, China
| | - HaiXue Zheng
- State Key Laboratory of Veterinary Etiological Biology, National Foot and Mouth Disease Reference Laboratory, Key Laboratory of Animal Virology of Ministry of Agriculture, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, China
| |
Collapse
|
39
|
Citron MP, McAnulty J, Callahan C, Knapp W, Fontenot J, Morales P, Flynn JA, Douglas CM, Espeseth AS. Transplacental Antibody Transfer of Respiratory Syncytial Virus Specific IgG in Non-Human Primate Mother-Infant Pairs. Pathogens 2021; 10:pathogens10111441. [PMID: 34832599 PMCID: PMC8624788 DOI: 10.3390/pathogens10111441] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/30/2021] [Accepted: 11/03/2021] [Indexed: 12/14/2022] Open
Abstract
One approach to protect new-borns against respiratory syncytial virus (RSV) is to vaccinate pregnant women in the last trimester of pregnancy. The boosting of circulating antibodies which can be transferred to the foetus would offer immune protection against the virus and ultimately the disease. Since non-human primates (NHPs) have similar reproductive anatomy, physiology, and antibody architecture and kinetics to humans, we utilized this preclinical species to evaluate maternal immunization (MI) using an RSV F subunit vaccine. Three species of NHPs known for their ability to be infected with human RSV in experimental challenge studies were tested for RSV-specific antibodies. African green monkeys had the highest overall antibody levels of the old-world monkeys evaluated and they gave birth to offspring with anti-RSV titers that were proportional to their mother. These higher overall antibody levels are associated with greater durability found in their offspring. Immunization of RSV seropositive AGMs during late pregnancy boosts RSV titers, which consequentially results in significantly higher titers in the vaccinated new-borns compared to the new-borns of unvaccinated mothers. These findings, accomplished in small treatment group sizes, demonstrate a model that provides an efficient, resource sparing and translatable preclinical in vivo system for evaluating vaccine candidates for maternal immunization.
Collapse
Affiliation(s)
- Michael P. Citron
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
- Correspondence:
| | - Jessica McAnulty
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Cheryl Callahan
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Walter Knapp
- Safety Assessment and Laboratory Animal Resources, Merck & Co., Inc., Kenilworth, NJ 07033, USA;
| | - Jane Fontenot
- The New Iberia Research Center, University of Louisiana at Lafayette, New Iberia, LA 70560, USA;
| | - Pablo Morales
- The Mannheimer Foundation, Homestead, FL 33034, USA;
| | - Jessica A. Flynn
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Cameron M. Douglas
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| | - Amy S. Espeseth
- Infectious Disease & Vaccines, Merck & Co., Inc., Kenilworth, NJ 07033, USA; (J.M.); (C.C.); (J.A.F.); (C.M.D.); (A.S.E.)
| |
Collapse
|
40
|
Are Primary Health Care Features Associated with Reduced Late Neonatal Mortality in Brazil? An Ecological Study. Matern Child Health J 2021; 26:1790-1799. [PMID: 34731357 DOI: 10.1007/s10995-021-03269-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/08/2021] [Indexed: 10/19/2022]
Abstract
OBJECTIVE To analyze the effect and efficiency of the characteristics of PHC facilities' structures and the work process of PHC teams on late neonatal mortality (LNM). METHODS This ecological time-series study adopted 3.764 Brazilian municipalities as analysis units. The independent variables were sorted into three hierarchical levels and four blocks. The distal level consisted of economic and demographic variables; the intermediate level comprised health coverage and demand for services; and the proximal level included structure and work process. The dependent variable was LNM. A linear mixed-effects regression analysis with a hierarchical approach was performed, estimating the crude (β) and adjusted (alpha = 5%) regression coefficients. Data involution analysis and municipalities were the decision-making unit according to their strata. RESULTS LNM was directly associated with the number of live births and unemployment rate. LNM was inversely associated with the year, per capita income, the community health worker's strategy coverage, vaginal delivery, household visits, and available vaccines. In the 2002-2014 period, the number of municipalities efficient in reducing LNM dropped from 38 to 27. In 2014, a more significant investment occurred in the number of vaginal deliveries in almost all strata to make inefficient municipalities efficient. CONCLUSION FOR PRACTICE The deaths of children aged 7-28 days are affected by the characteristics of the PHC structure and work process.
Collapse
|
41
|
Chaudhari T. Vaccinations in the newborn. Best Pract Res Clin Obstet Gynaecol 2021; 76:66-82. [DOI: 10.1016/j.bpobgyn.2020.09.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2020] [Revised: 09/15/2020] [Accepted: 09/26/2020] [Indexed: 02/07/2023]
|
42
|
Immune Responses against SARS-CoV-2-Questions and Experiences. Biomedicines 2021; 9:biomedicines9101342. [PMID: 34680460 PMCID: PMC8533170 DOI: 10.3390/biomedicines9101342] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 09/17/2021] [Accepted: 09/18/2021] [Indexed: 02/07/2023] Open
Abstract
Understanding immune reactivity against SARS-CoV-2 is essential for coping with the COVID-19 pandemic. Herein, we discuss experiences and open questions about the complex immune responses to SARS-CoV-2. Some people react excellently without experiencing any clinical symptoms, they do not get sick, and they do not pass the virus on to anyone else ("sterilizing" immunity). Others produce antibodies and do not get COVID-19 but transmit the virus to others ("protective" immunity). Some people get sick but recover. A varying percentage develops respiratory failure, systemic symptoms, clotting disorders, cytokine storms, or multi-organ failure; they subsequently decease. Some develop long COVID, a new pathologic entity similar to fatigue syndrome or autoimmunity. In reality, COVID-19 is considered more of a systemic immune-vascular disease than a pulmonic disease, involving many tissues and the central nervous system. To fully comprehend the complex clinical manifestations, a profound understanding of the immune responses to SARS-CoV-2 is a good way to improve clinical management of COVID-19. Although neutralizing antibodies are an established approach to recognize an immune status, cellular immunity plays at least an equivalent or an even more important role. However, reliable methods to estimate the SARS-CoV-2-specific T cell capacity are not available for clinical routines. This deficit is important because an unknown percentage of people may exist with good memory T cell responsibility but a low number of or completely lacking peripheral antibodies against SARS-CoV-2. Apart from natural immune responses, vaccination against SARS-CoV-2 turned out to be very effective and much safer than naturally acquired immunity. Nevertheless, besides unwanted side effects of the currently available vector and mRNA preparations, concerns remain whether these vaccines will be strong enough to defeat the pandemic. Altogether, herein we discuss important questions, and try to give answers based on the current knowledge and preliminary data from our laboratories.
Collapse
|
43
|
Shook LL, Fallah PN, Silberman JN, Edlow AG. COVID-19 Vaccination in Pregnancy and Lactation: Current Research and Gaps in Understanding. Front Cell Infect Microbiol 2021; 11:735394. [PMID: 34604115 PMCID: PMC8481914 DOI: 10.3389/fcimb.2021.735394] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 08/31/2021] [Indexed: 01/21/2023] Open
Abstract
The COVID-19 pandemic has demonstrated the urgent need to develop vaccine strategies optimized for pregnant people and their newborns, as both populations are at risk of developing severe disease. Although not included in COVID-19 vaccine development trials, pregnant people have had access to these vaccines since their initial release in the US and abroad. The rapid development and distribution of novel COVID-19 vaccines to people at risk, including those who are pregnant and lactating, presents an unprecedented opportunity to further our understanding of vaccine-induced immunity in these populations. In this review, we aim to summarize the literature to date on COVID-19 vaccination in pregnancy and lactation and highlight opportunities for investigation that may inform future maternal vaccine development and implementation strategies.
Collapse
Affiliation(s)
- Lydia L. Shook
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| | - Parisa N. Fallah
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Jason N. Silberman
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
| | - Andrea G. Edlow
- Department of Obstetrics and Gynecology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States
- Vincent Center for Reproductive Biology, Massachusetts General Hospital, Boston, MA, United States
| |
Collapse
|
44
|
Khan A, Sayedahmed EE, Singh VK, Mishra A, Dorta-Estremera S, Nookala S, Canaday DH, Chen M, Wang J, Sastry KJ, Mittal SK, Jagannath C. A recombinant bovine adenoviral mucosal vaccine expressing mycobacterial antigen-85B generates robust protection against tuberculosis in mice. Cell Rep Med 2021; 2:100372. [PMID: 34467249 PMCID: PMC8385328 DOI: 10.1016/j.xcrm.2021.100372] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 02/16/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023]
Abstract
Although the BCG vaccine offers partial protection, tuberculosis remains a leading cause of infectious disease death, killing ∼1.5 million people annually. We developed mucosal vaccines expressing the autophagy-inducing peptide C5 and mycobacterial Ag85B-p25 epitope using replication-defective human adenovirus (HAdv85C5) and bovine adenovirus (BAdv85C5) vectors. BAdv85C5-infected dendritic cells (DCs) expressed a robust transcriptome of genes regulating antigen processing compared to HAdv85C5-infected DCs. BAdv85C5-infected DCs showed enhanced galectin-3/8 and autophagy-dependent in vitro Ag85B-p25 epitope presentation to CD4 T cells. BCG-vaccinated mice were intranasally boosted using HAdv85C5 or BAdv85C5 followed by infection using aerosolized Mycobacterium tuberculosis (Mtb). BAdv85C5 protected mice against tuberculosis both as a booster after BCG vaccine (>1.4-log10 reduction in Mtb lung burden) and as a single intranasal dose (>0.5-log10 reduction). Protection was associated with robust CD4 and CD8 effector (TEM), central memory (TCM), and CD103+/CD69+ lung-resident memory (TRM) T cell expansion, revealing BAdv85C5 as a promising mucosal vaccine for tuberculosis.
Collapse
Affiliation(s)
- Arshad Khan
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| | - Ekramy E. Sayedahmed
- Department of Comparative Pathobiology and Purdue Institute of Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Vipul K. Singh
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| | - Abhishek Mishra
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| | | | - Sita Nookala
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - David H. Canaday
- Department of Medicine, Case Western Reserve University and Cleveland Veterans Affairs, Cleveland, OH, USA
| | - Min Chen
- Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Jin Wang
- Immunobiology and Transplant Science Center, Houston Methodist Research Institute, and Department of Surgery, Weill Cornell Medical College, Houston, TX, USA
| | - K. Jagannadha Sastry
- Department of Thoracic Head and Neck Medical Oncology, MD Anderson Cancer Center, Houston, TX, USA
| | - Suresh K. Mittal
- Department of Comparative Pathobiology and Purdue Institute of Inflammation, Immunology, and Infectious Disease, College of Veterinary Medicine, Purdue University, West Lafayette, IN, USA
| | - Chinnaswamy Jagannath
- Department of Pathology and Genomic Medicine, Houston Methodist Academic Institute, Houston Methodist Research Institute & Weill Cornell Medical College, Houston, TX, USA
| |
Collapse
|
45
|
Saso A, Kampmann B, Roetynck S. Vaccine-Induced Cellular Immunity against Bordetella pertussis: Harnessing Lessons from Animal and Human Studies to Improve Design and Testing of Novel Pertussis Vaccines. Vaccines (Basel) 2021; 9:877. [PMID: 34452002 PMCID: PMC8402596 DOI: 10.3390/vaccines9080877] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 07/28/2021] [Indexed: 12/14/2022] Open
Abstract
Pertussis ('whooping cough') is a severe respiratory tract infection that primarily affects young children and unimmunised infants. Despite widespread vaccine coverage, it remains one of the least well-controlled vaccine-preventable diseases, with a recent resurgence even in highly vaccinated populations. Although the exact underlying reasons are still not clear, emerging evidence suggests that a key factor is the replacement of the whole-cell (wP) by the acellular pertussis (aP) vaccine, which is less reactogenic but may induce suboptimal and waning immunity. Differences between vaccines are hypothesised to be cell-mediated, with polarisation of Th1/Th2/Th17 responses determined by the composition of the pertussis vaccine given in infancy. Moreover, aP vaccines elicit strong antibody responses but fail to protect against nasal colonisation and/or transmission, in animal models, thereby potentially leading to inadequate herd immunity. Our review summarises current knowledge on vaccine-induced cellular immune responses, based on mucosal and systemic data collected within experimental animal and human vaccine studies. In addition, we describe key factors that may influence cell-mediated immunity and how antigen-specific responses are measured quantitatively and qualitatively, at both cellular and molecular levels. Finally, we discuss how we can harness this emerging knowledge and novel tools to inform the design and testing of the next generation of improved infant pertussis vaccines.
Collapse
Affiliation(s)
- Anja Saso
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Beate Kampmann
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| | - Sophie Roetynck
- The Vaccine Centre, Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1 7HT, UK; (B.K.); (S.R.)
- Vaccines and Immunity Theme, MRC Unit, The Gambia at London School of Hygiene & Tropical Medicine, Banjul P.O. Box 273, The Gambia
| |
Collapse
|
46
|
Wen S, Wu Z, Zhong S, Li M, Shu Y. Factors influencing the immunogenicity of influenza vaccines. Hum Vaccin Immunother 2021; 17:2706-2718. [PMID: 33705263 DOI: 10.1080/21645515.2021.1875761] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Annual vaccination is the best prevention of influenza. However, the immunogenicity of influenza vaccines varies among different populations. It is important to fully identify the factors that may affect the immunogenicity of the vaccines to provide best protection for vaccine recipients. This paper reviews the factors that may influence the immunogenicity of influenza vaccines from the aspects of vaccine factors, adjuvants, individual factors, repeated vaccination, and genetic factors. The confirmed or hypothesized molecular mechanisms of these factors have also been briefly summarized.
Collapse
Affiliation(s)
- Simin Wen
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Zhengyu Wu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Shuyi Zhong
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Mao Li
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China
| | - Yuelong Shu
- School of Public Health (Shenzhen), Sun Yat-sen University, Guangdong, China.,National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Prevention and Control, Beijing, China
| |
Collapse
|
47
|
Clemens EA, Alexander-Miller MA. Understanding Antibody Responses in Early Life: Baby Steps towards Developing an Effective Influenza Vaccine. Viruses 2021; 13:v13071392. [PMID: 34372597 PMCID: PMC8310046 DOI: 10.3390/v13071392] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Accepted: 07/13/2021] [Indexed: 01/01/2023] Open
Abstract
The immune system of young infants is both quantitatively and qualitatively distinct from that of adults, with diminished responsiveness leaving these individuals vulnerable to infection. Because of this, young infants suffer increased morbidity and mortality from respiratory pathogens such as influenza viruses. The impaired generation of robust and persistent antibody responses in these individuals makes overcoming this increased vulnerability through vaccination challenging. Because of this, an effective vaccine against influenza viruses in infants under 6 months is not available. Furthermore, vaccination against influenza viruses is challenging even in adults due to the high antigenic variability across viral strains, allowing immune evasion even after induction of robust immune responses. This has led to substantial interest in understanding how specific antibody responses are formed to variable and conserved components of influenza viruses, as immune responses tend to strongly favor recognition of variable epitopes. Elicitation of broadly protective antibody in young infants, therefore, requires that both the unique characteristics of young infant immunity as well as the antibody immunodominance present among epitopes be effectively addressed. Here, we review our current understanding of the antibody response in newborns and young infants and discuss recent developments in vaccination strategies that can modulate both magnitude and epitope specificity of IAV-specific antibody.
Collapse
|
48
|
Akhtar M, Nizam NN, Basher SR, Hossain L, Akter S, Bhuiyan TR, Qadri F, Lundgren A. dmLT Adjuvant Enhances Cytokine Responses to T Cell Stimuli, Whole Cell Vaccine Antigens and Lipopolysaccharide in Both Adults and Infants. Front Immunol 2021; 12:654872. [PMID: 34054818 PMCID: PMC8160295 DOI: 10.3389/fimmu.2021.654872] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 04/19/2021] [Indexed: 02/02/2023] Open
Abstract
Enhancement of mucosal immune responses in children and infants using novel adjuvants such as double mutant heat labile toxin (dmLT) is an important goal in the enteric vaccine field. dmLT has been shown to enhance mucosal IgA responses to the oral inactivated enterotoxigenic Escherichia coli (ETEC) vaccine ETVAX. dmLT can enhance IL-17A production from adult T cells, which may increase the production and secretion of mucosal IgA antibodies. However, the adjuvant mechanism remains to be fully elucidated and might differ between infants and adults due to age-related differences in the development of the immune system. The main objective of this study was to determine how dmLT influences antigen presenting cells and T cells from infants compared to adults, and the role of IL-1β for mediating the adjuvant activity. Peripheral blood mononuclear cells (PBMCs) from Bangladeshi infants (6-11 months) and adults (18-40 years) were stimulated with the mitogen phytohaemagglutinin (PHA), the superantigen Staphylococcal enterotoxin B (SEB), ETVAX whole cell component (WCC) or E. coli lipopolysaccharide (LPS) ± dmLT, and cytokine production was measured using ELISA and electrochemiluminescence assays. The adjuvant dmLT significantly enhanced SEB- and PHA-induced IL-17A, but not IFN-γ responses, in PBMCs from both infants and adults. Blocking experiments using an IL-1 receptor antagonist demonstrated the importance of IL-1 signaling for the adjuvant effect. dmLT, ETVAX WCC and LPS induced dose-dependent IL-1β responses of comparable magnitudes in infant and adult cells. Depletion experiments suggested that IL-1β was mainly produced by monocytes. dmLT enhanced IL-1β responses to low doses of WCC and LPS, and the adjuvant effect appeared over a wider dose-range of WCC in infants. dmLT and WCC also induced IL-6, IL-23 and IL-12p70 production in both age groups and dmLT tended to particularly enhance IL-23 responses to WCC. Our results show that dmLT can induce IL-1β as well as other cytokines, which in turn may enhance IL-17A and potentially modulate other immunological responses in both infants and adults. Thus, dmLT may have an important function in promoting immune responses to the ETVAX vaccine, as well as other whole cell- or LPS-based vaccines in infants in low- and middle-income countries.
Collapse
Affiliation(s)
- Marjahan Akhtar
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Nuder Nower Nizam
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Salima Raiyan Basher
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Lazina Hossain
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Sarmin Akter
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Taufiqur Rahman Bhuiyan
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Firdausi Qadri
- Infectious Diseases Division, icddr,b (International Centre for Diarrhoeal Disease Research, Bangladesh), Dhaka, Bangladesh
| | - Anna Lundgren
- Department of Microbiology and Immunology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
49
|
England R, Pak J, Liu M, Rao S, Ozonoff A, Levy O, van Haren SD. Human Blood Plasma Shapes Distinct Neonatal TLR-Mediated Dendritic Cell Activation via Expression of the MicroRNA Let-7g. Immunohorizons 2021; 5:246-256. [PMID: 33931496 PMCID: PMC11688640 DOI: 10.4049/immunohorizons.2000081] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 03/19/2021] [Indexed: 11/19/2022] Open
Abstract
The newborn innate immune system is characterized as functionally distinct, resulting in impaired proinflammatory responses to many stimuli and a bias toward Th2 development. Although the magnitude of impairment can be partially overcome, for instance through activation of TLR7/8 in newborn dendritic cells, the newborn innate response remains distinct from that of adults. Using human in vitro modeling of newborn and adult dendritic cells, we investigated the role of extracellular and intracellular regulators in driving age-specific responses to TLR7/8 stimulation. MicroRNA expression profiling and plasma switch experiments identified Let-7g as a novel regulator of newborn innate immunity. Activation-induced expression of Let-7g in monocyte-derived dendritic cells (MoDCs) is driven by newborn plasma and reduces expression of costimulatory receptors CD86, MHC class I, and CCR7 and secretion of IFN-α and sCD40L. Conversely, an increase in secretion of the Th2-polarizing cytokine IL-12p40 is observed. Overexpression of Let-7g in adult MoDCs resulted in the same observations. Small interfering RNA-mediated ablation of Let-7g levels in newborn MoDCs resulted in an adult-like phenotype. In conclusion, this study reveals for the first time (to our knowledge) that age-specific differences in human plasma induce the microRNA Let-7g as a key mediator of the newborn innate immune phenotype. These observations shed new light on the mechanisms of immune ontogeny and may inform approaches to discover age-specific immunomodulators, such as adjuvants.
Collapse
Affiliation(s)
- Ross England
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Jensen Pak
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Mark Liu
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Shun Rao
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Al Ozonoff
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA
- Harvard Medical School, Boston, MA; and
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA;
- Harvard Medical School, Boston, MA; and
| |
Collapse
|
50
|
Analysis of Pneumonia Occurrence in Relation to Climate Change in Tanga, Tanzania. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2021; 18:ijerph18094731. [PMID: 33946714 PMCID: PMC8125699 DOI: 10.3390/ijerph18094731] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/22/2021] [Accepted: 04/27/2021] [Indexed: 11/17/2022]
Abstract
In 2018, 70% of global fatalities due to pneumonia occurred in about fifteen countries, with Tanzania being among the top eight countries contributing to these deaths. Environmental and individual factors contributing to these deaths may be multifaceted, but they have not yet been explored in Tanzania. Therefore, in this study, we explore the association between climate change and the occurrence of pneumonia in the Tanga Region, Tanzania. A time series study design was employed using meteorological and health data of the Tanga Region collected from January 2016 to December 2018 from the Tanzania Meteorological Authority and Health Management Information System, respectively. The generalized negative binomial regression technique was used to explore the associations between climate indicators (i.e., precipitation, humidity, and temperature) and the occurrence of pneumonia. There were trend differences in climate indicators and the occurrence of pneumonia between the Tanga and Handeni districts. We found a positive association between humidity and increased rates of non-severe pneumonia (incidence rate ratio (IRR) = 1.01; 95% CI: 1.01–1.02; p ≤ 0.05) and severe pneumonia (IRR = 1.02; 95% CI: 1.01–1.03; p ≤ 0.05). There was also a significant association between cold temperatures and the rate of severe pneumonia in Tanga (IRR = 1.21; 95% CI: 1.11–1.33; p ≤ 0.001). Other factors that were associated with pneumonia included age and district of residence. We found a positive relationship between humidity, temperature, and incidence of pneumonia in the Tanga Region. Policies focusing on prevention and control, as well as promotion strategies relating to climate change-related health effects should be developed and implemented.
Collapse
|