1
|
Motsoeneng BM, Dhar N, Nunes MC, Krammer F, Madhi SA, Moore PL, Richardson SI. Hemagglutinin Stalk-Specific Fc-Mediated Functions Are Associated With Protection Against Influenza Illness After Seasonal Influenza Vaccination. J Infect Dis 2024; 230:1329-1336. [PMID: 38743692 DOI: 10.1093/infdis/jiae241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 04/25/2024] [Accepted: 05/08/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Future vaccine candidates aim to elicit antibodies against the conserved hemagglutinin stalk domain. Understanding the protective mechanism of these antibodies, which mediate broad neutralization and Fc-mediated functions, following seasonal vaccination is critical. METHODS Plasma samples were obtained from pregnant women with or without HIV-1 enrolled in a randomised trial (138 trivalent inactivated vaccine [TIV] and 145 placebo recipients). Twenty-three influenza cases were confirmed within 6 months postpartum. We measured H1 stalk-specific antibody-dependent cellular phagocytosis (ADCP), complement deposition (ADCD) and cellular cytotoxicity (ADCC) at enrolment and 1-month postvaccination. RESULTS Lower H1 stalk-specific ADCP and ADCD activity was detected for participants with confirmed influenza compared with individuals without illness 1-month postvaccination. Pre-existing ADCP scores ≥250 reduced the odds of A/H1N1 infection (odds ratio [OR], 0.11; P = .01) with an 83% likelihood of risk reduction. Following TIV, ADCD scores of ≥25 and ≥15 significantly reduced the odds against A/H1N1 (OR, 0.10; P = .01) and non-group 1 (OR, 0.06; P = .0004) influenza virus infections, respectively. These ADCD scores were associated with >84% likelihood of risk reduction. CONCLUSIONS Overall, H1 stalk-specific Fc effector function correlates with protection against influenza illness following influenza vaccination during pregnancy. These findings provide insight into the protective mechanisms of hemagglutinin stalk antibodies. CLINICAL TRIALS REGISTRATION NCT01306669 and NCT01306682 (ClinicalTrials.gov).
Collapse
Affiliation(s)
- Boitumelo M Motsoeneng
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa
| | - Nisha Dhar
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Marta C Nunes
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Center of Excellence in Respiratory Pathogens, Hospices Civils de Lyon and Centre International de Recherche en Infectiologie, Équipe Santé Publique, Épidémiologie et Écologie Évolutive des Maladies Infectieuses (PHE3ID), Inserm U1111, CNRS UMR5308, École Normale Supérieure de Lyon, Université Claude Bernard, Lyon, France
| | - Florian Krammer
- Department of Microbiology, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Department of Pathology, Molecular and Cell Based Medicine, Icahn School of Medicine at Mount Sinai, New York, New York, USA
- Center for Vaccine Research and Pandemic Preparedness, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Shabir A Madhi
- South African Medical Research Council Vaccines and Infectious Diseases Analytics Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- Department of Science and Innovation/National Research Foundation, South African Research Chair Initiative in Vaccine Preventable Diseases Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- African Leadership in Vaccinology Expertise, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - Penny L Moore
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa
- Centre for the AIDS Programme of Research in South Africa, University of KwaZulu Natal, Durban, South Africa
| | - Simone I Richardson
- South African Medical Research Council Antibody Immunity Research Unit, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
- HIV Virology Section, Centre for HIV and STIs, National Institute for Communicable Diseases, National Health Laboratory Services, Johannesburg, South Africa
| |
Collapse
|
2
|
Holmes S, Li H, Shen X, Martin M, Tuck R, Chen Y, Giorgi EE, Kirshner HF, Berry M, Van Italie E, Venkatayogi S, Martin Beem JS, Edwards RJ, Mansouri K, Singh A, Kuykendall C, Gurley T, Anthony Moody M, DeNayer N, Demarco T, Denny TN, Wang Y, Evangelous TD, Clinton JT, Hora B, Wagh K, Seaman MS, Saunders KO, Solomotis N, Misamore J, Lewis MG, Wiehe K, Montefiori DC, Shaw GM, Williams WB. Neonatal immunity associated with heterologous HIV-1 neutralizing antibody induction in SHIV-infected Rhesus Macaques. Nat Commun 2024; 15:10302. [PMID: 39604409 PMCID: PMC11603298 DOI: 10.1038/s41467-024-54753-6] [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: 02/23/2024] [Accepted: 11/20/2024] [Indexed: 11/29/2024] Open
Abstract
The details of the pediatric immune system that supports induction of antibodies capable of neutralizing geographically-diverse or heterologous HIV-1 is currently unclear. Here we explore the pediatric immune environment in neonatal macaque undergoing Simian-HIV infection. Simian-HIV infection of 11 pairs of therapy-naive dams and infant rhesus macaques for 24 months results in heterologous HIV-1 neutralizing antibodies in 64% of young macaques compared to 18% of adult macaques. Heterologous HIV-1 neutralizing antibodies emerge by 12 months post-infection in young macaques, in association with lower expression of immunosuppressive genes, fewer germinal center CD4 + T regulatory cells, and a lower ratio of CD4 + T follicular regulatory to helper cells. Antibodies from peripheral blood B cells in two young macaques following SHIV infection neutralize 13% of 119 heterologous HIV-1 strains and map to regions of canonical broadly neutralizing antibody epitopes on the envelope surface protein. Here we show that pediatric immunity to SHIV infection in a macaque model may inform vaccine strategies to induce effective HIV-1 neutralizing antibodies in infants and children prior to viral exposure.
Collapse
Affiliation(s)
- Sommer Holmes
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Hui Li
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Xiaoying Shen
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Mitchell Martin
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ryan Tuck
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Yue Chen
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | | | | | - Madison Berry
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Elizabeth Van Italie
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Sravani Venkatayogi
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Joshua S Martin Beem
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Robert J Edwards
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Katayoun Mansouri
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Ajay Singh
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Cindy Kuykendall
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thaddeus Gurley
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - M Anthony Moody
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | - Nicole DeNayer
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Todd Demarco
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Thomas N Denny
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - Yunfei Wang
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Tyler D Evangelous
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - John T Clinton
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Bhavna Hora
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
| | - Kshitij Wagh
- Los Alamos National Laboratory, Los Alamos, NM, USA
| | - Michael S Seaman
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
| | - Kevin O Saunders
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | | | | | | | - Kevin Wiehe
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Medicine, Duke University School of Medicine, Durham, NC, USA
| | - David C Montefiori
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - George M Shaw
- Departments of Medicine and Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Wilton B Williams
- Duke Human Vaccine Institute, Duke University School of Medicine, Durham, NC, USA.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Integrative Immunobiology, Duke University School of Medicine, Durham, NC, USA.
| |
Collapse
|
3
|
England RN, Drapeau EM, Alameh MG, Hosseinzadeh R, Weissman D, Hensley SE. Evaluation of mRNA-LNP and adjuvanted protein SARS-CoV-2 vaccines in a maternal antibody mouse model. NPJ Vaccines 2024; 9:110. [PMID: 38890316 PMCID: PMC11189435 DOI: 10.1038/s41541-024-00901-4] [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: 04/12/2023] [Accepted: 06/06/2024] [Indexed: 06/20/2024] Open
Abstract
Maternal antibodies (matAbs) protect against a myriad of pathogens early in life; however, these antibodies can also inhibit de novo immune responses against some vaccine platforms. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) matAbs are efficiently transferred during pregnancy and protect infants against subsequent SARS-CoV-2 infections. It is unknown if matAbs inhibit immune responses elicited by different types of SARS-CoV-2 vaccines. Here, we established a mouse model to determine if SARS-CoV-2 spike-specific matAbs inhibit immune responses elicited by recombinant protein and nucleoside-modified mRNA-lipid nanoparticle (mRNA-LNP) vaccines. We found that SARS-CoV-2 mRNA-LNP vaccines elicited robust de novo antibody responses in mouse pups in the presence of matAbs. Recombinant protein vaccines were also able to circumvent the inhibitory effects of matAbs when adjuvants were co-administered. While additional studies need to be completed in humans, our studies raise the possibility that mRNA-LNP-based and adjuvanted protein-based SARS-CoV-2 vaccines have the potential to be effective when delivered very early in life.
Collapse
Affiliation(s)
- Ross N England
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pediatrics, Children's Hospital of Philadelphia, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth M Drapeau
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mohamad-Gabriel Alameh
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
4
|
Zhang HQ, Zhang QY, Yuan ZM, Zhang B. The potential epidemic threat of Ebola virus and the development of a preventive vaccine. JOURNAL OF BIOSAFETY AND BIOSECURITY 2023; 5:67-78. [DOI: 10.1016/j.jobb.2023.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2025] Open
|
5
|
Serum IgM antibody response to Clostridioides difficile polysaccharide PS-II vaccination in pony foals. Anaerobe 2022; 77:102635. [PMID: 36064161 DOI: 10.1016/j.anaerobe.2022.102635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Revised: 08/16/2022] [Accepted: 08/28/2022] [Indexed: 11/21/2022]
Abstract
OBJECTIVES Clostridioides difficile (formerly Clostridium difficile) is associated with colitis in foals and mature horses. C. difficile exposes specific phosphorylated polysaccharides (PSs), named PS-I, PS-II and PS-III. These cell-surface PSs are potential vaccine targets, especially the hexasaccharide phosphate PS-II, that has been found in all C. difficile ribotypes examined. Since we previously identified anti-PS-II circulating antibodies in horses, we postulated that vaccinating foals with PS-II may prevent colonization by C. difficile. In this study, we aim to evaluate the IgM antibody responses in foals to PS-II. METHODS To evaluate the reactogenicity and immunogenicity of C. difficile PS-II in foals, three-to four-month-old foals were vaccinated intramuscularly three times at intervals of three weeks with 100 μg/dose (3 foals) or 500 μg/dose (3 foals) of purified PS-II antigen with aluminum hydroxide adjuvant, or with a placebo preparation (2 foals) containing adjuvant alone. RESULTS No injection site swelling, pain or fever was observed after vaccination. Two of the three foals receiving 100 μg/dose, and three out of three foals receiving 500 μg/dose of PS-II responded with increases in serum IgM antibodies. No control foals that received the placebo had IgM responses to PS-II. There was a trend towards a higher response rate in foals receiving 500 μg PS-II one week after second vaccination when compared to control foals and towards higher concentrations of serum IgM antibodies in foals receiving 500 μg PS-II. CONCLUSIONS No adverse reactions were observed following vaccination with PS-II in foals; Serum IgM immune responses were induced by vaccination. A polysaccharide-based vaccine for C. difficile in horses deserves further investigation.
Collapse
|
6
|
van Haren SD, Pedersen GK, Kumar A, Ruckwardt TJ, Moin S, Moore IN, Minai M, Liu M, Pak J, Borriello F, Doss-Gollin S, Beijnen EMS, Ahmed S, Helmel M, Andersen P, Graham BS, Steen H, Christensen D, Levy O. CAF08 adjuvant enables single dose protection against respiratory syncytial virus infection in murine newborns. Nat Commun 2022; 13:4234. [PMID: 35918315 PMCID: PMC9346114 DOI: 10.1038/s41467-022-31709-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Accepted: 06/30/2022] [Indexed: 11/09/2022] Open
Abstract
Respiratory syncytial virus is a leading cause of morbidity and mortality in children, due in part to their distinct immune system, characterized by impaired induction of Th 1 immunity. Here we show application of cationic adjuvant formulation CAF08, a liposomal vaccine formulation tailored to induce Th 1 immunity in early life via synergistic engagement of Toll-like Receptor 7/8 and the C-type lectin receptor Mincle. We apply quantitative phosphoproteomics to human dendritic cells and reveal a role for Protein Kinase C-δ for enhanced Th1 cytokine production in neonatal dendritic cells and identify signaling events resulting in antigen cross-presentation. In a murine in vivo model a single immunization at birth with CAF08-adjuvanted RSV pre-fusion antigen protects newborn mice from RSV infection by induction of antigen-specific CD8+ T-cells and Th1 cells. Overall, we describe a pediatric adjuvant formulation and characterize its mechanism of action providing a promising avenue for development of early life vaccines against RSV and other respiratory viral pathogens.
Collapse
Affiliation(s)
- Simon D van Haren
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA.
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| | - Gabriel K Pedersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Azad Kumar
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Tracy J Ruckwardt
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Syed Moin
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Ian N Moore
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mahnaz Minai
- Infectious Disease Pathogenesis Section, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Mark Liu
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Jensen Pak
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Francesco Borriello
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Department of Translational Medical Sciences and Center for Basic and Clinical Immunology Research (CISI), University of Naples Federico II, Naples, Italy
- Generate Biomedicines, Cambridge, MA, USA
| | - Simon Doss-Gollin
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Elisabeth M S Beijnen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
| | - Saima Ahmed
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Michaela Helmel
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Peter Andersen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
- Department of Immunology and Microbiology, University of Copenhagen, Copenhagen, Denmark
| | - Barney S Graham
- Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Hanno Steen
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pathology, Boston Children's Hospital and Harvard Medical School, Boston, MA, USA
| | - Dennis Christensen
- Center for Vaccine Research, Statens Serum Institut, Copenhagen, Denmark
| | - Ofer Levy
- Precision Vaccines Program, Division of Infectious Diseases, Boston Children's Hospital, Boston, MA, USA
- Department of Pediatrics, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Boston, MA, USA
| |
Collapse
|
7
|
Ajmeriya S, Kumar A, Karmakar S, Rana S, Singh H. Neutralizing Antibodies and Antibody-Dependent Enhancement in COVID-19: A Perspective. J Indian Inst Sci 2022; 102:671-687. [PMID: 35136306 PMCID: PMC8814804 DOI: 10.1007/s41745-021-00268-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Accepted: 09/28/2021] [Indexed: 12/14/2022]
Abstract
Antibody-dependent enhancement (ADE) is an alternative route of viral entry in the susceptible host cell. In this process, antiviral antibodies enhance the entry access of virus in the cells via interaction with the complement or Fc receptors leading to the worsening of infection. SARS-CoV-2 variants pose a general concern for the efficacy of neutralizing antibodies that may fail to neutralize infection, raising the possibility of a more severe form of COVID-19. Data from various studies on respiratory viruses raise the speculation that antibodies elicited against SARS-CoV-2 and during COVID-19 recovery could potentially exacerbate the infection through ADE at sub-neutralizing concentrations; this may contribute to disease pathogenesis. It is, therefore, of utmost importance to study the effectiveness of the anti-SARS-CoV-2 antibodies in COVID-19-infected subjects. Theoretically, ADE remains a general concern for the efficacy of antibodies elicited during infection, most notably in convalescent plasma therapy and in response to vaccines where it could be counterproductive.
Collapse
Affiliation(s)
- Swati Ajmeriya
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Amit Kumar
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Subhradip Karmakar
- Department of Biochemistry, All India Institute of Medical Sciences, AIIMS, Room no 3020, Ansari Nagar, New Delhi, 110029 India
| | - Shweta Rana
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| | - Harpreet Singh
- Division of Biomedical Informatics, ICMR-AIIMS Computational Genomics Center, Indian Council of Medical Research (ICMR), Ansari Nagar, New Delhi, 110029 India
| |
Collapse
|
8
|
Husseini N, Beard SC, Hodgins DC, Barnes C, Chik E, Mallard BA. Immuno-phenotyping of Canadian Beef Cattle: Adaptation of the High Immune Response Methodology for Utilization in Beef Cattle. Transl Anim Sci 2022; 6:txac006. [PMID: 35261968 PMCID: PMC8896012 DOI: 10.1093/tas/txac006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Indexed: 11/23/2022] Open
Abstract
The high immune response (HIR) methodology measures the genetic performance of the adaptive immune system to identify and breed animals with balanced and robust immunity. The HIR methodology has previously been used in dairy and swine to reduce disease but has not been fully investigated in beef cattle. The first objective of the current study was to examine whether the HIR methodology as standardized for use in dairy cattle was appropriate for use in beef cattle. The second objective was to determine the earliest age for immune response phenotyping of beef calves. In this study, beef calves (n = 295) of various ages, as well as mature beef cows (n = 170) of mixed breeds, were immunized using test antigens to assess their antibody- (AMIR) and cell-mediated immune responses (CMIR). Heritability for AMIR and CMIR was estimated at 0.43 and 0.18, respectively. The HIR methodology was appropriate for use in beef cattle; beef calves as young as 2–3 wk of age were capable of mounting AMIR responses comparable with those seen historically in mature Holstein dairy cows. Three-week-old beef calves mounted CMIR responses comparable with those of Holstein cows, but 9-mo-old calves and mature beef cows had significantly higher CMIR responses than Holsteins. The HIR methodology can be used to measure both AMIR and CMIR in beef calves as young as 3 wk of age.
Collapse
|
9
|
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
|
10
|
Wickramasinghe LC, van Wijngaarden P, Tsantikos E, Hibbs ML. The immunological link between neonatal lung and eye disease. Clin Transl Immunology 2021; 10:e1322. [PMID: 34466225 PMCID: PMC8387470 DOI: 10.1002/cti2.1322] [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: 12/22/2020] [Revised: 06/02/2021] [Accepted: 07/13/2021] [Indexed: 01/02/2023] Open
Abstract
Bronchopulmonary dysplasia (BPD) and retinopathy of prematurity (ROP) are two neonatal diseases of major clinical importance, arising in large part as a consequence of supplemental oxygen therapy used to promote the survival of preterm infants. The presence of coincident inflammation in the lungs and eyes of neonates receiving oxygen therapy indicates that a dysregulated immune response serves as a potential common pathogenic factor for both diseases. This review examines the current state of knowledge of immunological dysregulation in BPD and ROP, identifying similarities in the cellular subsets and inflammatory cytokines that are found in the alveoli and retina during the active phase of these diseases, indicating possible mechanistic overlap. In addition, we highlight gaps in the understanding of whether these responses emerge independently in the lung and retina as a consequence of oxygen exposure or arise because of inflammatory spill-over from the lung. As BPD and ROP are anatomically distinct, they are often considered discreet disease entities and are therefore treated separately. We propose that an improved understanding of the relationship between BPD and ROP is key to the identification of novel therapeutic targets to treat or prevent both conditions simultaneously.
Collapse
Affiliation(s)
- Lakshanie C Wickramasinghe
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Peter van Wijngaarden
- OphthalmologyDepartment of SurgeryUniversity of MelbourneMelbourneVICAustralia
- Centre for Eye Research AustraliaRoyal Victorian Eye and Ear HospitalEast MelbourneVICAustralia
| | - Evelyn Tsantikos
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| | - Margaret L Hibbs
- Leukocyte Signalling LaboratoryDepartment of Immunology and PathologyCentral Clinical SchoolMonash UniversityMelbourneVICAustralia
| |
Collapse
|
11
|
de Groot N, Fariñas F, Cabrera-Gómez CG, Pallares FJ, Ramis G. Weaning causes a prolonged but transient change in immune gene expression in the intestine of piglets. J Anim Sci 2021; 99:6153447. [PMID: 33640983 PMCID: PMC8051849 DOI: 10.1093/jas/skab065] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 02/24/2021] [Indexed: 12/24/2022] Open
Abstract
Controlling gut inflammation is important in managing gut disorders in the piglet after weaning. Establishing patterns of inflammation markers in the time subsequent to weaning is important for future research to determine whether interventions are effective in controlling gut inflammation. The objective of this study was to evaluate the intestinal inflammatory response during the postweaning period in piglets. A 45-d study included 108 piglets (weaned at 22 d, body weight 5.53 ± 1.19 kg), distributed in 12 pens with nine pigs per pen. Histomorphometry, gene expression of pro- and anti-inflammatory cytokines, and the quantity of immunoglobulin (Ig) A producing cells were measured in jejunum, ileum, and colon on days 0, 15, 30, and 45 postweaning. Cytokine gene expression in peripheral blood mononuclear cells and Ig quantities were analyzed in blood from piglets on days 0, 15, 30, and 45 postweaning. Histomorphometrical results showed a lower villus length directly after weaning. Results demonstrated a postweaning intestinal inflammation response for at least 15 d postweaning by upregulation of IgA producing cells and IFN-γ, IL-1α, IL-8, IL-10, IL-12α, and TGF-β in jejunum, ileum, and colon. IgM and IgA were upregulated at day 30 postweaning. IgG was downregulated at day 15 postweaning. The results indicate that weaning in piglets is associated with a prolonged and transient response in gene expression of pro- and anti-inflammatory cytokines and IgA producing cells in the intestine.
Collapse
Affiliation(s)
- Nienke de Groot
- Trouw Nutrition Innovation, Amersfoort 3811 MH, The Netherlands.,Instituto de Inmunología Clínica y Enfermedades Infecciosas, Málaga, Spain
| | - Fernando Fariñas
- Dpto. Producción Animal, Facultad de Veterinaria, Universidad de Murcia, Murcia, Spain
| | | | - Francisco J Pallares
- Dpto. Anatomía y Anatomía Patológica Comparadas, Universidad de Murcia, Murcia, Spain
| | - Guillermo Ramis
- Dpto. Producción Animal, Facultad de Veterinaria, Universidad de Murcia, Murcia, Spain
| |
Collapse
|
12
|
Sartoretti J, Eberhardt CS. The Potential Role of Nonhuman Primate Models to Better Comprehend Early Life Immunity and Maternal Antibody Transfer. Vaccines (Basel) 2021; 9:vaccines9040306. [PMID: 33804886 PMCID: PMC8063815 DOI: 10.3390/vaccines9040306] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 03/14/2021] [Accepted: 03/17/2021] [Indexed: 02/04/2023] Open
Abstract
Early life immunity is a complex field of research and there are still gaps in knowledge regarding the detailed mechanism of maternal antibody transfer, the impact of maternal antibodies on infant vaccine responses and the ontogeny of human early life immunity. A comprehensive understanding is necessary to identify requirements for early life vaccines and to improve early childhood immunization. New immunological methods have facilitated performing research in the youngest, however, some questions can only be addressed in animal models. To date, mostly murine models are used to study neonatal and infant immunity since they are well-described, easy to use and cost effective. Given their limitations especially in the transfer biology of maternal antibodies and the lack of infectivity of numerous human pathogens, this opinion piece discusses the potential and prerequisites of the nonhuman primate model in studying early life immunity and maternal antibody transfer.
Collapse
Affiliation(s)
- Julie Sartoretti
- Center for Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland;
- Department of Woman, Child and Adolescent Medicine, Geneva University Hospitals and Faculty of Medicine, 6 rue Willy-Donze, 1211 Geneve 4, Switzerland
| | - Christiane S. Eberhardt
- Center for Vaccinology, Department of Pathology and Immunology, Faculty of Medicine, University of Geneva, 1211 Geneva 4, Switzerland;
- Department of Woman, Child and Adolescent Medicine, Geneva University Hospitals and Faculty of Medicine, 6 rue Willy-Donze, 1211 Geneve 4, Switzerland
- Center for Vaccinology, University Hospitals of Geneva, 1205 Geneva, Switzerland
- Emory Vaccine Center, Emory University, Atlanta, GA 30322, USA
- Correspondence:
| |
Collapse
|
13
|
Cuervo W, Sordillo LM, Abuelo A. Oxidative Stress Compromises Lymphocyte Function in Neonatal Dairy Calves. Antioxidants (Basel) 2021; 10:antiox10020255. [PMID: 33562350 PMCID: PMC7915147 DOI: 10.3390/antiox10020255] [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: 01/13/2021] [Revised: 01/31/2021] [Accepted: 02/03/2021] [Indexed: 12/02/2022] Open
Abstract
Dairy calves are unable to mount an effective immune response during their first weeks of life, which contributes to increased disease susceptibility during this period. Oxidative stress (OS) diminishes the immune cell capabilities of humans and adult cows, and dairy calves also experience OS during their first month of life. However, the impact that OS may have on neonatal calf immunity remains unexplored. Thus, we aimed to evaluate the impact of OS on newborn calf lymphocyte functions. For this, we conducted two experiments. First, we assessed the association of OS status throughout the first month of age and the circulating concentrations of the cytokines interferon-gamma (IFN-γ) and interleukin (IL) 4, as well as the expression of cytokine-encoding genes IFNG, IL2, IL4, and IL10 in peripheral mononuclear blood cells (PBMCs) of 12 calves. Subsequently, we isolated PBMCs from another 6 neonatal calves to investigate in vitro the effect of OS on immune responses in terms of activation of lymphocytes, cytokine expression, and antibody production following stimulation with phorbol 12-myristate 13-acetate or bovine herpesvirus-1. The results were compared statistically through mixed models. Calves exposed to high OS status in their first month of age showed higher concentrations of IL-4 and expression of IL4 and IL10 and lower concentrations of IFN-γ and expression of IFNG and IL2 than calves exposed to lower OS. In vitro, OS reduced lymphocyte activation, production of antibodies, and protein and gene expression of key cytokines. Collectively, our results demonstrate that OS can compromise some immune responses of newborn calves. Hence, further studies are needed to explore the mechanisms of how OS affects the different lymphocyte subsets and the potential of ameliorating OS in newborn calves as a strategy to augment the functional capacity of calf immune cells, as well as enhance calves’ resistance to infections.
Collapse
|
14
|
Semmes EC, Chen JL, Goswami R, Burt TD, Permar SR, Fouda GG. Understanding Early-Life Adaptive Immunity to Guide Interventions for Pediatric Health. Front Immunol 2021; 11:595297. [PMID: 33552052 PMCID: PMC7858666 DOI: 10.3389/fimmu.2020.595297] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2020] [Accepted: 12/04/2020] [Indexed: 01/16/2023] Open
Abstract
Infants are capable of mounting adaptive immune responses, but their ability to develop long-lasting immunity is limited. Understanding the particularities of the neonatal adaptive immune system is therefore critical to guide the design of immune-based interventions, including vaccines, in early life. In this review, we present a thorough summary of T cell, B cell, and humoral immunity in early life and discuss infant adaptive immune responses to pathogens and vaccines. We focus on the differences between T and B cell responses in early life and adulthood, which hinder the generation of long-lasting adaptive immune responses in infancy. We discuss how knowledge of early life adaptive immunity can be applied when developing vaccine strategies for this unique period of immune development. In particular, we emphasize the use of novel vaccine adjuvants and optimization of infant vaccine schedules. We also propose integrating maternal and infant immunization strategies to ensure optimal neonatal protection through passive maternal antibody transfer while avoiding hindering infant vaccine responses. Our review highlights that the infant adaptive immune system is functionally distinct and uniquely regulated compared to later life and that these particularities should be considered when designing interventions to promote pediatric health.
Collapse
Affiliation(s)
- Eleanor C. Semmes
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
- Medical Scientist Training Program, Duke University, Durham, NC, United States
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
| | - Jui-Lin Chen
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| | - Ria Goswami
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
| | - Trevor D. Burt
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
- Division of Neonatology, Department of Pediatrics, Duke University, Durham, NC, United States
| | - Sallie R. Permar
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
| | - Genevieve G. Fouda
- Duke Human Vaccine Institute, Duke University, Durham, NC, United States
- Children’s Health and Discovery Initiative, Department of Pediatrics, Duke University, Durham, NC, United States
| |
Collapse
|
15
|
Ginseng Stem-Leaf Saponins in Combination with Selenium Promote the Immune Response in Neonatal Mice with Maternal Antibody. Vaccines (Basel) 2020; 8:vaccines8040755. [PMID: 33322647 PMCID: PMC7768402 DOI: 10.3390/vaccines8040755] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Revised: 12/10/2020] [Accepted: 12/10/2020] [Indexed: 12/24/2022] Open
Abstract
Neonates acquire from their mothers maternal antibody (MatAb) which results in poor immune response to vaccination. We previously demonstrated that ginseng stem-leaf saponins in combination with selenium (GSe) had adjuvant effect on the immune response to an attenuated pseudorabies virus (aPrV) vaccine. The present study was to evaluate GSe for its effect on the immune response to aPrV vaccine in neonatal mice with MatAb. Results showed that GSe had adjuvant effect on the immune response to aPrV vaccine in neonates. When GSe was co-administered with aPrV vaccine (aP-GSe), specific gB antibody, Th1 cytokines (IL-2, IL-12 and IFN-γ) and Th2 cytokines (IL-4, IL-6 and IL-10) responses were significantly increased in association with enhanced protection of vaccinated neonates against the lethal PrV challenge even though MatAb existed when compared to the neonates immunized with aPrV vaccine alone. GSe-enhanced immune response depended on its use in the primary immunization. The mechanisms underlying the adjuvant effect of GSe may be due to more innate immune related pathways activated by GSe. Transcriptome analysis of splenocytes from neonates immunized with aP-GSe, aPrV or saline solution showed that there were 3976 differentially expressed genes (DEGs) in aP-GSe group while 5959 DEGs in aPrV group when compared to the control. Gene ontology (GO) terms and Kyoto encyclopedia of genes and genomes (KEGG) pathways analysis showed that innate immune responses and cytokine productions related terms or pathways were predominantly enriched in aP-GSe group, such as “NOD-like receptor signaling pathway”, “Natural killer cell mediated cytotoxicity”, “NF-κB signaling pathway”, “cytokine-cytokine receptor interaction”, and “Th1 and Th2 cell differentiation”. Considering the potent adjuvant effect of GSe on aPrV vaccine in neonatal mice with MatAb, it deserves further investigation in piglets.
Collapse
|
16
|
Protecting the most vulnerable from hand, foot, and mouth disease. THE LANCET. INFECTIOUS DISEASES 2020; 21:308-309. [PMID: 33031751 DOI: 10.1016/s1473-3099(20)30452-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 05/22/2020] [Indexed: 01/31/2023]
|
17
|
Ramos L, Lunney JK, Gonzalez-Juarrero M. Neonatal and infant immunity for tuberculosis vaccine development: importance of age-matched animal models. Dis Model Mech 2020; 13:dmm045740. [PMID: 32988990 PMCID: PMC7520460 DOI: 10.1242/dmm.045740] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Neonatal and infant immunity differs from that of adults in both the innate and adaptive arms, which are critical contributors to immune-mediated clearance of infection and memory responses elicited during vaccination. The tuberculosis (TB) research community has openly admitted to a vacuum of knowledge about neonatal and infant immune responses to Mycobacterium tuberculosis (Mtb) infection, especially in the functional and phenotypic attributes of memory T cell responses elicited by the only available vaccine for TB, the Bacillus Calmette-Guérin (BCG) vaccine. Although BCG vaccination has variable efficacy in preventing pulmonary TB during adolescence and adulthood, 80% of endemic TB countries still administer BCG at birth because it has a good safety profile and protects children from severe forms of TB. As such, new vaccines must work in conjunction with BCG at birth and, thus, it is essential to understand how BCG shapes the immune system during the first months of life. However, many aspects of the neonatal and infant immune response elicited by vaccination with BCG remain unknown, as only a handful of studies have followed BCG responses in infants. Furthermore, most animal models currently used to study TB vaccine candidates rely on adult-aged animals. This presents unique challenges when transitioning to human trials in neonates or infants. In this Review, we focus on vaccine development in the field of TB and compare the relative utility of animal models used thus far to study neonatal and infant immunity. We encourage the development of neonatal animal models for TB, especially the use of pigs.
Collapse
Affiliation(s)
- Laylaa Ramos
- Mycobacteria Research Laboratories, Microbiology Immunology and Pathology Department, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| | - Joan K Lunney
- Animal Parasitic Diseases Laboratory, BARC, NEA, ARS, USDA Building 1040, Room 103, Beltsville, MD 20705, USA
| | - Mercedes Gonzalez-Juarrero
- Mycobacteria Research Laboratories, Microbiology Immunology and Pathology Department, Colorado State University, 1682 Campus Delivery, Fort Collins, CO 80523, USA
| |
Collapse
|
18
|
Willis E, Pardi N, Parkhouse K, Mui BL, Tam YK, Weissman D, Hensley SE. Nucleoside-modified mRNA vaccination partially overcomes maternal antibody inhibition of de novo immune responses in mice. Sci Transl Med 2020; 12:eaav5701. [PMID: 31915303 PMCID: PMC7339908 DOI: 10.1126/scitranslmed.aav5701] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 06/21/2019] [Accepted: 09/23/2019] [Indexed: 12/12/2022]
Abstract
Maternal antibodies provide short-term protection to infants against many infections. However, they can inhibit de novo antibody responses in infants elicited by infections or vaccination, leading to increased long-term susceptibility to infectious diseases. Thus, there is a need to develop vaccines that are able to elicit protective immune responses in the presence of antigen-specific maternal antibodies. Here, we used a mouse model to demonstrate that influenza virus-specific maternal antibodies inhibited de novo antibody responses in mouse pups elicited by influenza virus infection or administration of conventional influenza vaccines. We found that a recently developed influenza vaccine, nucleoside-modified mRNA encapsulated in lipid nanoparticles (mRNA-LNP), partially overcame this inhibition by maternal antibodies. The mRNA-LNP influenza vaccine established long-lived germinal centers in the mouse pups and elicited stronger antibody responses than did a conventional influenza vaccine approved for use in humans. Vaccination with mRNA-LNP vaccines may offer a promising strategy for generating robust immune responses in infants in the presence of maternal antibodies.
Collapse
Affiliation(s)
- Elinor Willis
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Norbert Pardi
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Kaela Parkhouse
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC V6T 1Z3, Canada
| | - Drew Weissman
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Scott E Hensley
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
19
|
Park S, Nixon CE, Pond-Tor S, Kabyemela ER, Fried M, Duffy PE, Kurtis JD, Friedman JF. Impact of maternally derived antibodies to Plasmodium falciparum Schizont Egress Antigen-1 on the endogenous production of anti-PfSEA-1 in offspring. Vaccine 2019; 37:5044-5050. [PMID: 31288996 PMCID: PMC6677924 DOI: 10.1016/j.vaccine.2019.06.084] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 06/07/2019] [Accepted: 06/27/2019] [Indexed: 02/03/2023]
Abstract
Background We evaluated whether maternally-derived antibodies to a malarial vaccine candidate, Plasmodium falciparum Schizont Egress Antigen-1 (PfSEA-1), in cord blood interfered with the development of infant anti-PfSEA-1 antibodies in response to natural exposure. Methods We followed 630 Tanzanian infants who were measured their antibodies against PfSEA-1 (aa 810-1023; PfSEA-1A) at birth and 6, 12, 18, and 24 months of age, and examined the changes in anti-PfSEA-1A antibody levels in response to parasitemia, and evaluated whether maternally-derived anti-PfSEA-1A antibodies in cord blood modified infant anti-PfSEA-1A immune responses. Results Infants who experienced parasitemia during the first 6 months of life had significantly higher anti-PfSEA-1A antibodies at 6 and 12 months of age compared to uninfected infants. Maternally-derived anti-PfSEA-1A antibodies in cord blood significantly modified this effect during the first 6 months. During this period, infant anti-PfSEA-1A antibody levels were significantly associated with their P. falciparum exposure when they were born with low, but not higher, maternally-derived anti-PfSEA-1A antibody levels in cord blood. Nevertheless, during the first 6 months of life, maternally-derived anti-PfSEA-1A antibodies in cord blood did not abrogate the parasitemia driven development of infant anti-PfSEA-1A: parasitemia were significantly correlated with anti-PfSEA-1A antibody levels at 6 months of age in the infants born with low maternally-derived anti-PfSEA-1A antibody levels in cord blood and borderline significantly correlated in those infants born with middle and high levels. Conclusions Maternal vaccination with PfSEA-1A is unlikely to interfere with the development of naturally acquired anti-PfSEA-1A immune responses following exposure during infancy.
Collapse
Affiliation(s)
- Sangshin Park
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States; Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States; Graduate School of Urban Public Health, University of Seoul, Seoul 02504, Republic of Korea.
| | - Christina E Nixon
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States; Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Sunthorn Pond-Tor
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States; Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Edward R Kabyemela
- Mother Offspring Malaria Studies (MOMS) Project, Seattle Biomedical Research Institute, Seattle, WA 98109, United States; Muheza Designated District Hospital, Muheza, Tanzania; Tumaini University, Moshi, Tanzania
| | - Michal Fried
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20892, United States
| | - Patrick E Duffy
- Laboratory of Malaria Immunology and Vaccinology, National Institute of Allergy and Infectious Diseases, NIH, Rockville, MD 20892, United States
| | - Jonathan D Kurtis
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States; Department of Pathology and Laboratory Medicine, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| | - Jennifer F Friedman
- Center for International Health Research, Rhode Island Hospital, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States; Department of Pediatrics, The Warren Alpert Medical School of Brown University, Providence, RI 02903, United States
| |
Collapse
|
20
|
Xu H, Ziani W, Shao J, Doyle-Meyers LA, Russell-Lodrigue KE, Ratterree MS, Veazey RS, Wang X. Impaired Development and Expansion of Germinal Center Follicular Th Cells in Simian Immunodeficiency Virus-Infected Neonatal Macaques. THE JOURNAL OF IMMUNOLOGY 2018; 201:1994-2003. [PMID: 30104244 DOI: 10.4049/jimmunol.1800235] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Accepted: 07/15/2018] [Indexed: 12/16/2022]
Abstract
Germinal center (GC) CD4+ follicular Th (Tfh) cells are critical for cognate B cell help in humoral immune responses to pathogenic infections. Although Tfh cells are expanded or depleted in HIV/SIV-infected adults, the effects of pediatric HIV/SIV infection on Tfh cells remain unclear. In this study, we examined changes in lymphoid follicle formation in lymph nodes focusing on GC Tfh cells, B cell development, and differentiation in SIV-infected neonatal rhesus macaques (Macaca mulatta) compared with age-matched cohorts. Our data showed that follicles and GCs of normal infants rapidly formed in the first few weeks of age, in parallel with increasing GC Tfh cells in various lymphoid tissues. In contrast, GC development and GC Tfh cells were markedly impaired in SIV-infected infants. There was a very low frequency of GC Tfh cells throughout SIV infection in neonates and subsequent infants, accompanied by high viremia, reduction of B cell proliferation/resting memory B cells, and displayed proinflammatory unresponsiveness. These findings indicate neonatal HIV/SIV infection compromises the development of GC Tfh cells, likely contributing to ineffective Ab responses, high viremia, and eventually rapid disease progression to AIDS.
Collapse
Affiliation(s)
- Huanbin Xu
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Widade Ziani
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Jiasheng Shao
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Lara A Doyle-Meyers
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Kasi E Russell-Lodrigue
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Marion S Ratterree
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Ronald S Veazey
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| | - Xiaolei Wang
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433
| |
Collapse
|
21
|
Labastida-Conde RG, Ramírez-Pliego O, Peleteiro-Olmedo M, Lopez-Guerrero DV, Badillo-Godinez OD, Gutiérrez-Xicoténcatl MDL, Rosas-Salgado G, González-Fernández Á, Esquivel-Guadarrama FR, Santana MA. Flagellin is a Th1 polarizing factor for human CD4 + T cells and induces protection in a murine neonatal vaccination model of rotavirus infection. Vaccine 2018; 36:4188-4197. [PMID: 29891347 DOI: 10.1016/j.vaccine.2018.06.005] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 05/29/2018] [Accepted: 06/03/2018] [Indexed: 02/07/2023]
Abstract
Neonates have an increased susceptibility to infections, particularly those caused by intracellular pathogens, leading to high morbidity and mortality rates. This is partly because of a poor response of neonatal CD4+ T cells, leading to deficient antibody production and a low production of IFN-γ, resulting in deficient elimination of intracellular pathogens. The poor memory response of human neonates has underpinned the need for improving vaccine formulations. Molecular adjuvants that improve the response of neonatal lymphocytes, such as the ligands of toll-like receptors (TLRs), are attractive candidates. Among them, flagellin, the TLR5 ligand, is effective at very low doses; prior immunity to flagellin does not impair its adjuvant activity. Human CD4+ and CD8+ T cells express TLR5. We found that flagellin induces the expression of IFN-γ, IL-1β and IL-12 in mononuclear cells from human neonate and adult donors. When human naïve CD4+ T cells were activated in the presence of flagellin, there was high level of expression of IFN-γ in both neonates and adults. Furthermore, flagellin induced IFN-γ production in Th1 cells obtained from adult donors; in the Th2 population, it inhibited IL-4 cytokine production. Flagellin also promoted expression of the IFN-γ receptor in naive CD4+ T cells from neonates and adults. To test the adjuvant capacity of flagellin in vivo, we used a murine neonate vaccination model for infection with rotavirus, a pathogen responsible for severe diarrhea in young infants. Using the conserved VP6 antigen, we observed an 80% protection against rotavirus infection in the presence of flagellin, but only in those mice previously primed in the neonatal period. Our data suggest that flagellin could be an attractive adjuvant for achieving a Th1 response.
Collapse
Affiliation(s)
| | - Oscar Ramírez-Pliego
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - Mercedes Peleteiro-Olmedo
- Inmunología, Centro de Investigaciones Biomédicas (CINBIO), Centro Singular de Investigación de Galicia, Instituto de Investigación Sanitaria Galicia Sur, Universidad de Vigo, Campus Universitario de Vigo, 36310 Vigo, Spain
| | | | | | | | - Gabriela Rosas-Salgado
- Facultad de Medicina, Universidad Autonoma del Estado de Morelos, Cuernavaca, Morelos, Mexico
| | - África González-Fernández
- Inmunología, Centro de Investigaciones Biomédicas (CINBIO), Centro Singular de Investigación de Galicia, Instituto de Investigación Sanitaria Galicia Sur, Universidad de Vigo, Campus Universitario de Vigo, 36310 Vigo, Spain
| | | | - M Angélica Santana
- Centro de Investigación en Dinámica Celular (IICBA), Universidad Autónoma del Estado de Morelos, Cuernavaca, Morelos, Mexico.
| |
Collapse
|
22
|
Lynn DJ, Pulendran B. The potential of the microbiota to influence vaccine responses. J Leukoc Biol 2017; 103:225-231. [PMID: 28864446 DOI: 10.1189/jlb.5mr0617-216r] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 07/21/2017] [Accepted: 08/01/2017] [Indexed: 12/25/2022] Open
Abstract
After clean water, vaccines are the primary public health intervention providing protection against serious infectious diseases. Antigen-specific antibody-mediated responses play a critical role in the protection conferred by vaccination; however these responses are highly variable among individuals. In addition, vaccine immunogenicity is frequently impaired in developing world populations, for reasons that are poorly understood. Although the factors that are associated with interindividual variation in vaccine responses are likely manifold, emerging evidence from mouse models and studies in human populations now suggests that the gut microbiome plays a key role in shaping systemic immune responses to both orally and parenterally administered vaccines. Herein, we review the evidence to date that the microbiota can influence vaccine responses and discuss the potential mechanisms through which these effects may be mediated. In addition, we highlight the gaps in this evidence and suggest future directions for research.
Collapse
Affiliation(s)
- David J Lynn
- Infection and Immunity Theme, South Australian Health and Medical Research Institute, North Terrace, Adelaide, South Australia, Australia.,School of Medicine, Flinders University, Bedford Park, South Australia, Australia
| | - Bali Pulendran
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, California, USA.,Department of Pathology, Stanford University School of Medicine, Stanford, California, USA.,Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
23
|
Sun J, Sun B, Gao Y, He F, Yang L, Wang M, Zhou W. Composition and Variation Analysis of the T Cell Receptor β-Chain Complementarity Determining Region 3 Repertoire in Neonatal Sepsis. Scand J Immunol 2017; 86:418-423. [PMID: 28891256 DOI: 10.1111/sji.12614] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 09/04/2017] [Indexed: 12/19/2022]
Abstract
T cell receptor (TCR) diversity is clearly related to protection from infection. However, the characteristics of TCR diversity in neonates are not clear. In this study, we investigated the TCR diversity of neonates with sepsis. Twenty neonates with severe sepsis and eight matched neonates without infection were enrolled in the study. For the neonates with sepsis, EDTA-anticoagulated blood was collected on day 1 after the diagnosis of sepsis and on day 7 of treatment. For the neonates without infection, blood was collected one time. DNA was extracted from peripheral blood mononuclear cells. The complementarity determining region 3 (CDR3) gene was analysed by multiplex PCR and high-throughput sequencing. The CDR3 types and lengths were similar in patients and healthy controls. There was a significant difference in VJ gene usage among the three groups. Compared to the healthy neonates, the neonates with sepsis had different VJ pairs and generated different clonotypes. Although the TCR β-chain diversity was generally lower in the neonates with sepsis, there was no significant difference in TCR β-chain diversity between the patients and the healthy controls. Our data showed the characteristics of the TCR repertoire in neonates with sepsis, which represents a potentially valuable data set. This result is useful for understanding neonatal susceptibility to infection.
Collapse
Affiliation(s)
- J Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - B Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, Shanghai, China
| | - Y Gao
- Department of Ultrasound, Children's Hospital of Fudan University, Shanghai, China
| | - F He
- BGI Genomics, BGI-Shenzhen, Shenzhen, China
| | - L Yang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - M Wang
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - W Zhou
- Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| |
Collapse
|
24
|
Efficacy of the marker vaccine rAdV-SFV-E2 against classical swine fever in the presence of maternally derived antibodies to rAdV-SFV-E2 or C-strain. Vet Microbiol 2016; 196:50-54. [DOI: 10.1016/j.vetmic.2016.10.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 09/30/2016] [Accepted: 10/04/2016] [Indexed: 02/06/2023]
|
25
|
Balka G, Dreckmann K, Papp G, Kraft C. Vaccination of piglets at 2 and 3 weeks of age with Ingelvac PRRSFLEX® EU provides protection against heterologous field challenge in the face of homologous maternally derived antibodies. Porcine Health Manag 2016; 2:24. [PMID: 28405450 PMCID: PMC5382426 DOI: 10.1186/s40813-016-0037-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2015] [Accepted: 07/14/2016] [Indexed: 01/13/2023] Open
Abstract
Background Due to difficulties in eradicating porcine reproductive and respiratory syndrome (PRRS) linked to biosecurity challenges, transmission of the virus and the lack of efficient DIVA vaccines, successful control of PRRS requires a combination of strict management measures and vaccination of both sows and piglets. The present study aimed to assess the efficacy of a recently developed MLV vaccine (Ingelvac PRRSFLEX® EU) in piglets at 2 and 3-weeks of age in the presence of homologous maternally derived antibodies as the dams were vaccinated with the same vaccine strain (ReproCyc® PRRS EU). Methods The study was carried out on a Hungarian farrow to finish farm naturally infected with PRRSv. The study was designed as a blind, placebo controlled side by side trial. ORF5 sequence similarity of the vaccine strain and the resident field strain was 87.8 %. PRRS specific real-time quantitative PCR was performed from serum samples to measure both the viral load and the frequency of virus positive animals. Results At the time of the natural infection observed in the control group at 10–12 weeks of age, the number of viraemic animals did not increase significantly in the vaccinated group. To understand the infection dynamics, positive PCR samples with low Ct values were sequenced (ORF5) and the data analysis indicated the circulation of wild type virus in both groups, however wild type virus was only found in non-vaccinated animals. Conclusions Our data indicate that piglets vaccinated at as early as 2 weeks of age with Ingelvac PRRSFLEX® EU were protected both in terms of proportion of viraemic animals and viraemia levels. It has to be highlighted that these results were achieved in piglets with high levels of homologous maternally derived antibodies (MDA) at the time of vaccination.
Collapse
Affiliation(s)
- Gyula Balka
- Department of Pathology, University of Veterinary Medicine, István u. 2, H-1078 Budapest, Hungary
| | - Karla Dreckmann
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, Bemeroder Str. 31, 30559 Hannover, Germany
| | - György Papp
- Jászapáti 2000 Mezőgazdasági Zrt., H-5130 Jászapáti, Hungary
| | - Christian Kraft
- Boehringer Ingelheim Veterinary Research Center GmbH & Co. KG, Bemeroder Str. 31, 30559 Hannover, Germany
| |
Collapse
|
26
|
Villa-Mancera A, Hernández-Guzmán K, Olivares-Pérez J, Molina-Mendoza P. Influence of four commercial porcine circovirus type 2 (PCV2) vaccines on the improvement of production parameters in pigs with maternally derived antibodies. Livest Sci 2016. [DOI: 10.1016/j.livsci.2016.05.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
27
|
Human milk oligosaccharides: The role in the fine-tuning of innate immune responses. Carbohydr Res 2016; 432:62-70. [PMID: 27448325 DOI: 10.1016/j.carres.2016.07.009] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 07/05/2016] [Accepted: 07/06/2016] [Indexed: 01/12/2023]
Abstract
In order to secure the health of newborns over the period of immune immaturity during the first months of life, a mother provides her offspring with passive protection: bioactive molecules transferred through the placenta and breast milk. It is well known that human milk contains immunoglobulins (Ig), immune cells and diverse cytokines, which affect newborn directly or indirectly and contribute to the maturation of the immune system. However, in addition to the above-stated molecules, human milk oligosaccharides (HMOs), a complex mixture of free indigestible carbohydrates with multiple functions, play exceptional roles in the functioning of the infants' immune system. These biological molecules have been studied over decades, however, interest in HMOs does not seem to have abated. Although biological activities of oligosaccharides from human milk have been explicitly reviewed, information regarding the role of HMOs in inflammation remains rather fragmented. The purpose of this review is to compile existing knowledge about the role of certain species of HMOs, including fucosylated, galactosylated and sialylated oligosaccharides, and their signaling pathways in immunity and inflammation. The advances in applying this information to the treatment of diseases in infants as well as adults were also reviewed here.
Collapse
|
28
|
Development of immune organs and functioning in humans and test animals: Implications for immune intervention studies. Reprod Toxicol 2016; 64:180-90. [PMID: 27282947 DOI: 10.1016/j.reprotox.2016.06.002] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Revised: 05/10/2016] [Accepted: 06/03/2016] [Indexed: 12/23/2022]
Abstract
A healthy immune status is mostly determined during early life stages and many immune-related diseases may find their origin in utero and the first years of life. Therefore, immune health optimization may be most effective during early life. This review is an inventory of immune organ maturation events in relation to developmental timeframes in minipig, rat, mouse and human. It is concluded that time windows of immune organ development in rodents can be translated to human, but minipig reflects the human timeframes better; however the lack of prenatal maternal-fetal immune interaction in minipig may cause less responsiveness to prenatal intervention. It is too early to conclude which immune parameters are most appropriate, because there are not enough comparative immune parameters. Filling these gaps will increase the predictability of results observed in experimental animals, and guide future intervention studies by assessing relevant parameters in the right corresponding developmental time frames.
Collapse
|
29
|
Affiliation(s)
- B Abela-Ridder
- Department of the Control of Neglected Tropical Diseases, World Health Organization, Avenue Appia 20, CH-1211, Genève 27, Switzerland e-mail:
| |
Collapse
|
30
|
Neonatal vaccination with bacillus Calmette-Guérin and hepatitis B vaccines modulates hippocampal synaptic plasticity in rats. J Neuroimmunol 2015; 288:1-12. [PMID: 26531688 DOI: 10.1016/j.jneuroim.2015.08.019] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 08/08/2015] [Accepted: 08/19/2015] [Indexed: 11/21/2022]
Abstract
Immune activation can exert multiple effects on synaptic transmission. Our study demonstrates the influence of neonatal vaccination on hippocampal synaptic plasticity in rats under normal physiological conditions. The results revealed that neonatal BCG vaccination enhanced synaptic plasticity. In contrast, HBV hampered it. Furthermore, we found that the cytokine balance shifted in favour of the T helper type 1/T helper type 2 immune response in BCG/HBV-vaccinated rats in the periphery. The peripheral IFN-γ:IL-4 ratio was positively correlated with BDNF and IGF-1 in the hippocampus. BCG raised IFN-γ, IL-4, BDNF and IGF-1 and reduced IL-1β, IL-6, and TNF-α in the hippocampus, whereas, HBV triggered the opposite effects.
Collapse
|
31
|
Morters MK, McNabb S, Horton DL, Fooks AR, Schoeman JP, Whay HR, Wood JLN, Cleaveland S. Effective vaccination against rabies in puppies in rabies endemic regions. Vet Rec 2015; 177:150. [PMID: 26109286 PMCID: PMC4552936 DOI: 10.1136/vr.102975] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/19/2015] [Indexed: 12/25/2022]
Abstract
In rabies endemic regions, a proportionally higher incidence of rabies is often reported in dogs younger than 12 months of age, which includes puppies less than 3 months of age; this presents a serious risk to public health. The higher incidence of rabies in young dogs may be the effect of low vaccination coverage in this age class, partly as a result of the perception that immature immune systems and maternal antibodies inhibit seroconversion to rabies vaccine in puppies less than three months of age. Therefore, to test this perception, the authors report the virus neutralising antibody titres from 27 dogs that were vaccinated with high quality, inactivated rabies vaccine aged three months of age and under as part of larger serological studies undertaken in Gauteng Province, South Africa, and the Serengeti District, Tanzania. All of these dogs seroconverted to a single dose of vaccine with no adverse reactions reported and with postvaccinal peak titres ranging from 2.0 IU/ml to 90.5 IU/ml. In light of these results, and the risk of human beings contracting rabies from close contact with puppies, the authors recommend that all dogs in rabies endemic regions, including those less than three months of age, are vaccinated with high quality, inactivated vaccine.
Collapse
Affiliation(s)
- M K Morters
- Disease Dynamics Unit, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - S McNabb
- The Neighbourhood Vet, East Dulwich, London, UK
| | - D L Horton
- School of Veterinary Medicine, University of Surrey, Guildford, UK
| | - A R Fooks
- Animal and Plant Health Agency, Weybridge, UK; and Department of Clinical Infection, Microbiology and Immunology, University of Liverpool, Liverpool, UK
| | - J P Schoeman
- Department of Companion Animal Clinical Studies, Faculty of Veterinary Science, University of Pretoria, Pretoria, South Africa
| | - H R Whay
- School of Veterinary Sciences, University of Bristol, Bristol, UK
| | - J L N Wood
- Disease Dynamics Unit, Department of Veterinary Medicine, University of Cambridge, Cambridge, UK
| | - S Cleaveland
- Institute of Biodiversity, Animal Health and Comparative Medicine, University of Glasgow, Glasgow, UK
| |
Collapse
|
32
|
Pyo HM, Hlasny M, Zhou Y. Influence of maternally-derived antibodies on live attenuated influenza vaccine efficacy in pigs. Vaccine 2015; 33:3667-72. [PMID: 26092308 DOI: 10.1016/j.vaccine.2015.06.044] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 06/03/2015] [Accepted: 06/05/2015] [Indexed: 01/10/2023]
Abstract
Vaccination during pregnancy is practiced in swine farms as one measure to control swine influenza virus (SIV) infection in piglets at an early age. Vaccine-induced maternal antibodies transfer to piglets through colostrum and stabilize the herd: however, maternally derived antibodies (MDA) interfere with immune response following influenza vaccination in piglets at the later stage of life. In addition, MDA is related to enhanced respiratory disease in SIV infection. Previously, we have developed a bivalent live attenuated influenza vaccine (LAIV) which harbors both H1 and H3 HAs. We demonstrated vaccination of this LAIV provided protection to homologous and heterologous SIV infection in pigs. In this study we aimed to investigate the influence of MDA on LAIV efficacy. To this end, SIV sero-negative sows were vaccinated with a commercial vaccine. After parturition, nursery piglets were vaccinated with LAIV intranasally or intramuscularly, and were then challenged with SIV. We report that MDA hampered serum antibody response induced by intramuscular vaccination but not by intranasal vaccination of the LAIV. Viral challenge in the presence of MDA caused exacerbated respiratory disease in unvaccinated piglets. In contrast, all LAIV vaccinated piglets were protected from homologous viral infection regardless of the route of vaccination and the presence of MDA. Our results demonstrated that LAIV conferred protection in the presence of MDA without inciting exacerbated respiratory disease.
Collapse
Affiliation(s)
- Hyun Mi Pyo
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, Canada S7N 5E3
| | - Magda Hlasny
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, Canada S7N 5E3
| | - Yan Zhou
- Vaccine and Infectious Disease Organization-International Vaccine Centre, University of Saskatchewan, 120 Veterinary Road, Saskatoon, SK, Canada S7N 5E3.
| |
Collapse
|
33
|
The role of pre-existing cross-reactive antibodies in determining the efficacy of vaccination in humans: study protocol for a randomized controlled trial. Trials 2015; 16:147. [PMID: 25872531 PMCID: PMC4404017 DOI: 10.1186/s13063-015-0651-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2014] [Accepted: 03/13/2015] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Epidemic viral diseases have become more prevalent.. Among the various strategies to prevent such epidemics, vaccination is the most cost-effective. However, populations that are immunized are typically already exposed to multiple previous vaccinations or natural infections. Studies from this and other laboratories have revealed that pre-existing dengue antibodies can either inhibit or enhance subsequent dengue infection depending on the pre-existing antibody levels. While cross-reactive antibody is potentially pathogenic in dengue, how it impacts immune response to vaccination is unclear. Aggregated at the site of vaccination and the respective draining lymph nodes are antigen-presenting and immune regulatory cells that express Fc receptors and play pivotal roles in determining the magnitude and polarity of the immune response. Vaccine uptake by these antigen-presenting cells may thus be either inhibited or enhanced when vaccines are opsonized with cross-reactive antibodies. DESIGN In view of the limited knowledge on how cross-reactive antibodies affect vaccination outcome, we propose a study that exploits the known cross reactivity between Japanese encephalitis (JE) virus antibody and yellow fever (YF) vaccine. We hypothesize that cross-reactive antibodies impact antibody response to YF at the point vaccination in a concentration-dependent manner by altering both vaccine uptake and the innate immune response by antigen presenting cells. We will structure an open-label clinical trial on sequential vaccination with JE and YF vaccines, with different time intervals between vaccinations. This would test immune response to YF vaccination in subjects with different titer of cross-reactive JE vaccine-derived antibodies. The clinical materials obtained in the trial will drive basic laboratory investigations directed at elucidating how heterologous antibody affect vaccination at the molecular level. YF neutralizing antibody titer will be measured using plaque reduction neutralization test against the vaccine strain YF17D. Innate immune response will be characterized genetically using either microarray or digital PCR (or both). The innate immune response will also be characterized at the protein and metabolite level using Luminex bead technology and lipidomic/metabolomic approaches. DISCUSSION This proposed study represents one of the first to examine the role of cross-reactive antibodies in modulating immune responses to vaccines, the findings of which may re-shape vaccination strategy. TRIAL REGISTRATION Clinical Trials.gov registration number: NCT01943305 (3 September 2013).
Collapse
|
34
|
Chen Q, Ross AC. α-Galactosylceramide stimulates splenic lymphocyte proliferation in vitro and increases antibody production in vivo in late neonatal-age mice. Clin Exp Immunol 2015; 179:188-96. [PMID: 25178151 DOI: 10.1111/cei.12447] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/26/2014] [Indexed: 12/11/2022] Open
Abstract
The neonatal stage is characterized by weak responses to various infections and vaccines, thus the development of efficient formulas to improve vaccine effectiveness is of high priority. The glycolipid alpha galactosylceramide (αGalCer) is known as a potent immune modulator due mainly to natural killer (NK) T cell activation. Using a mouse tetanus toxoid (TT) immunization model, we observed that neonatal mice given αGalCer at the time of primary immunization on postnatal day (pnd) 17 had a significantly higher TT-specific immunoglobulin (Ig)M response as well as a memory IgG response, while αGalCer given on pnd 7 resulted in only marginal boosting. Consistently, immunostaining of the spleen sections from αGalCer-treated pnd 17 immunized neonates showed a higher number of Ki67(+) cells in the splenic germinal centre area, suggesting a stronger response after immunization. In-vitro kinetic studies revealed that spleen cells from newborn to pnd 7 neonates did not respond to αGalCer stimulation, whereas cell proliferation was increased markedly by αGalCer after pnd 7, and became dramatic around neonatal pnd 17-18, which was accompanied by increased B, T and NK T cell populations in the spleen. In addition, in pnd 17 spleen cells, αGalCer significantly stimulated the production of NK T cytokines, interleukin (IL)-4 and interferon (IFN)-γ, and promoted the proliferation of CD23(+) B cells, a subset of B cells enriched in germinal centres. These data suggest that αGalCer is an effective immune stimulus in the late neonatal stage, and thus may be useful in translational studies to test as a potential adjuvant to achieve a more efficient response to immunization.
Collapse
Affiliation(s)
- Q Chen
- Department of Nutritional Sciences, The Pennsylvania State University, University Park, PA, USA
| | | |
Collapse
|
35
|
Canine distemper virus DNA vaccination of mink can overcome interference by maternal antibodies. Vaccine 2015; 33:1375-81. [PMID: 25637861 DOI: 10.1016/j.vaccine.2015.01.029] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2014] [Revised: 11/22/2014] [Accepted: 01/09/2015] [Indexed: 11/24/2022]
Abstract
Canine distemper virus (CDV) is highly contagious and can cause severe disease against which conventional live vaccines are ineffective in the presence of maternal antibodies. Vaccination in the presences of maternal antibodies was challenged by vaccination of 5 days old and 3 weeks old mink kits with CDV DNA vaccines. Virus neutralising (VN) antibody responses were induced in mink kits vaccinated with a plasmid encoding the haemaglutinin protein (H) of CDV (n=5, pCDV-H) or a combination of the H, fusion (F) and nucleoprotein (N) of CDV (n=5, pCDV-HFN). These DNA vaccinated kits were protected against virulent experimental infection with field strains of CDV. The pCDV-H was more efficient in inducing protective immunity in the presence of maternal antibodies compared to the pCDV-HFN. The results show that DNA vaccination with the pCDV-H or pCDV-HFN (n=4) only given once at 5 days of age induces virus specific immune response in neonatal mink and protection against virulent CDV exposure later in life.
Collapse
|
36
|
Perkins GA, Wagner B. The development of equine immunity: Current knowledge on immunology in the young horse. Equine Vet J 2015; 47:267-74. [DOI: 10.1111/evj.12387] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 11/09/2014] [Indexed: 01/21/2023]
Affiliation(s)
- G. A. Perkins
- Department of Clinical Sciences; College of Veterinary Medicine; Cornell University; Ithaca New York USA
| | - B. Wagner
- Department of Population Medicine and Diagnostic Sciences; College of Veterinary Medicine; Cornell University; Ithaca New York USA
| |
Collapse
|
37
|
Bielefeldt-Ohmann H, Prow NA, Wang W, Tan CSE, Coyle M, Douma A, Hobson-Peters J, Kidd L, Hall RA, Petrovsky N. Safety and immunogenicity of a delta inulin-adjuvanted inactivated Japanese encephalitis virus vaccine in pregnant mares and foals. Vet Res 2014; 45:130. [PMID: 25516480 PMCID: PMC4268807 DOI: 10.1186/s13567-014-0130-7] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 12/04/2014] [Indexed: 11/10/2022] Open
Abstract
In 2011, following severe flooding in Eastern Australia, an unprecedented epidemic of equine encephalitis occurred in South-Eastern Australia, caused by Murray Valley encephalitis virus (MVEV) and a new variant strain of Kunjin virus, a subtype of West Nile virus (WNVKUN). This prompted us to assess whether a delta inulin-adjuvanted, inactivated cell culture-derived Japanese encephalitis virus (JEV) vaccine (JE-ADVAX™) could be used in horses, including pregnant mares and foals, to not only induce immunity to JEV, but also elicit cross-protective antibodies against MVEV and WNVKUN. Foals, 74–152 days old, received two injections of JE-ADVAX™. The vaccine was safe and well-tolerated and induced a strong JEV-neutralizing antibody response in all foals. MVEV and WNVKUN antibody cross-reactivity was seen in 33% and 42% of the immunized foals, respectively. JE-ADVAX™ was also safe and well-tolerated in pregnant mares and induced high JEV-neutralizing titers. The neutralizing activity was passively transferred to their foals via colostrum. Foals that acquired passive immunity to JEV via maternal antibodies then were immunized with JE-ADVAX™ at 36–83 days of age, showed evidence of maternal antibody interference with low peak antibody titers post-immunization when compared to immunized foals of JEV-naïve dams. Nevertheless, when given a single JE-ADVAX™ booster immunization as yearlings, these animals developed a rapid and robust JEV-neutralizing antibody response, indicating that they were successfully primed to JEV when immunized as foals, despite the presence of maternal antibodies. Overall, JE-ADVAX™ appears safe and well-tolerated in pregnant mares and young foals and induces protective levels of JEV neutralizing antibodies with partial cross-neutralization of MVEV and WNVKUN.
Collapse
Affiliation(s)
- Helle Bielefeldt-Ohmann
- School of Veterinary Science, University of Queensland, Gatton Campus, Gatton 4343, Qld, Australia.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Abstract
Enteric viral infections in domestic animals cause significant economic losses. The recent emergence of virulent enteric coronaviruses [porcine epidemic diarrhea virus (PEDV)] in North America and Asia, for which no vaccines are available, remains a challenge for the global swine industry. Vaccination strategies against rotavirus and coronavirus (transmissible gastroenteritis virus) infections are reviewed. These vaccination principles are applicable against emerging enteric infections such as PEDV. Maternal vaccines to induce lactogenic immunity, and their transmission to suckling neonates via colostrum and milk, are critical for early passive protection. Subsequently, in weaned animals, oral vaccines incorporating novel mucosal adjuvants (e.g., vitamin A, probiotics) may provide active protection when maternal immunity wanes. Understanding intestinal and systemic immune responses to experimental rotavirus and transmissible gastroenteritis virus vaccines and infection in pigs provides a basis and model for the development of safe and effective vaccines for young animals and children against established and emerging enteric infections.
Collapse
Affiliation(s)
- Kuldeep S Chattha
- Canadian Food Inspection Agency, Lethbridge, Alberta T1H 6P7, Canada;
| | | | | |
Collapse
|
39
|
Gervassi A, Lejarcegui N, Dross S, Jacobson A, Itaya G, Kidzeru E, Gantt S, Jaspan H, Horton H. Myeloid derived suppressor cells are present at high frequency in neonates and suppress in vitro T cell responses. PLoS One 2014; 9:e107816. [PMID: 25248150 PMCID: PMC4172591 DOI: 10.1371/journal.pone.0107816] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 08/04/2014] [Indexed: 12/26/2022] Open
Abstract
Over 4 million infants die each year from infections, many of which are vaccine-preventable. Young infants respond relatively poorly to many infections and vaccines, but the basis of reduced immunity in infants is ill defined. We sought to investigate whether myeloid-derived suppressor cells (MDSC) represent one potential impediment to protective immunity in early life, which may help inform strategies for effective vaccination prior to pathogen exposure. We enrolled healthy neonates and children in the first 2 years of life along with healthy adult controls to examine the frequency and function of MDSC, a cell population able to potently suppress T cell responses. We found that MDSC, which are rarely seen in healthy adults, are present in high numbers in neonates and their frequency rapidly decreases during the first months of life. We determined that these neonatal MDSC are of granulocytic origin (G-MDSC), and suppress both CD4+ and CD8+ T cell proliferative responses in a contact-dependent manner and gamma interferon production. Understanding the role G-MDSC play in infant immunity could improve vaccine responsiveness in newborns and reduce mortality due to early-life infections.
Collapse
Affiliation(s)
- Ana Gervassi
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Nicholas Lejarcegui
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Sandra Dross
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- University of Washington Department of Global Health, Seattle, Washington, United States of America
| | - Amanda Jacobson
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Grace Itaya
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Elvis Kidzeru
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Soren Gantt
- University of British Columbia Department of Pediatrics and Child and Family Research Institute, Vancouver, Canada
| | - Heather Jaspan
- University of Washington Seattle Children's Hospital, Seattle, Washington, United States of America
- Division of Immunology, Institute of Infectious Diseases and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | - Helen Horton
- Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- University of Washington Department of Global Health, Seattle, Washington, United States of America
- University of Washington Department of Medicine, Seattle, Washington, United States of America
- * E-mail:
| |
Collapse
|
40
|
Abstract
Neonates have little immunological memory and a developing immune system, which increases their vulnerability to infectious agents. Recent advances in the understanding of neonatal immunity indicate that both innate and adaptive responses are dependent on precursor frequency of lymphocytes, antigenic dose and mode of exposure. Studies in neonatal mouse models and human umbilical cord blood cells demonstrate the capability of neonatal immune cells to produce immune responses similar to adults in some aspects but not others. This review focuses mainly on the developmental and functional mechanisms of the human neonatal immune system. In particular, the mechanism of innate and adaptive immunity and the role of neutrophils, antigen presenting cells, differences in subclasses of T lymphocytes (Th1, Th2, Tregs) and B cells are discussed. In addition, we have included the recent developments in the neonatal mouse immune system. Understanding neonatal immunity is essential to development of therapeutic vaccines to combat newly emerging infectious agents.
Collapse
Affiliation(s)
- Saleem Basha
- Center for Infectious Disease and Immunology, Rochester General Hospital Research Institute, 1425 Portland Avenue, Rochester, NY 14621, USA
| | | | | |
Collapse
|
41
|
Maternal T-lymphocytes in equine colostrum express a primarily inflammatory phenotype. Vet Immunol Immunopathol 2014; 161:141-50. [PMID: 25174977 DOI: 10.1016/j.vetimm.2014.07.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2014] [Revised: 07/22/2014] [Accepted: 07/25/2014] [Indexed: 01/02/2023]
Abstract
The purpose of this study was to characterize maternal immune cells in colostrum of mares. Cell phenotypes and cytokine secretion from mare peripheral blood mononuclear cells (PBMC) and cells from colostrum were analyzed by flow cytometry and by multiplex cytokine analysis. Equine colostral leukocytes were composed of mainly CD8(+) and CD4(+) lymphocytes. CD8(+) cells were significantly enriched in colostrum compared to PBMC (n=35). Colostral T-cells (n=13) responded to stimulation with PMA/ionomycin with a significantly higher magnitude of IL-17 (p=0.037) and similar IFN-γ concentrations (p=0.305), while IL-4 (p=0.0002) and IL-10 (p=0.0002) production was decreased compared to PBMC. CD4(+) and CD8(+) T-cells in colostrum produced IFN-γ (n=4). The findings show that colostrum T-cells can produce all four cytokines investigated here but most cells are polarized toward IL-17 and IFN-γ production and an inflammatory phenotype. Maternal T-cells likely migrate to the colostrum in a selective manner and may have specific roles in neonatal immune development.
Collapse
|
42
|
Klein R, Lourenço M, Moutinho F, Takahira R, Lopes R, Martins R, Machado L, Silveira V, Ferreira H. Imunidade celular em caninos neonatos - do nascimento ao 45° dia de idade. ARQ BRAS MED VET ZOO 2014. [DOI: 10.1590/1678-41625985] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
O objetivo do presente trabalho foi acompanhar o desenvolvimento imunológico dos neonatos caninos, a fim de avaliar a imunidade celular pela análise dos leucócitos e linfócitos totais e das subpopulações de linfócitos T (CD4+ e CD8+) pela técnica de citometria de fluxo. Foram utilizados 30 cães neonatos de ambos os sexos, sem raça definida, aos três, 10, 17, 24, 31, 38 e 45 dias de idade. A contagem de leucócitos totais aos 45 dias (11.639±3.574) foi significativamente maior que no terceiro dia de idade (8.740±1.812) (P<0,05); não houve diferença entre a contagem total de linfócitos aos 45 dias em relação ao terceiro dia de idade. Quanto às subpopulações de LT CD4+ e LT CD8+, os percentuais de LT CD4+, aos três dias de idade (24,9±16,8%), foram inferiores quando comparados à média entre o 10°, o 24° e o 31°dia (35,5%), e os de CD8+, ao terceiro dia, menores em relação às médias do 10° e do 31° dia de idade. Pode-se concluir que as subpopulações de LT CD4+ e CD8+ sofrem oscilações durante o desenvolvimento pós-natal, sendo estas crescentes em relação aos níveis obtidos aos três dias de idade. A relação CD4+:CD8+ mostrou superioridade para o primeiro tipo celular, sendo que a maior relação entre CD4+ e CD8+ ocorreu no terceiro dia de idade. Com base nos resultados obtidos neste estudo, notaram-se as diferenças semanais nas populações linfocitárias, o que demonstra a dinâmica dessas células durante o período neonatal.
Collapse
|
43
|
Mayer AE, Johnson JB, Parks GD. The neutralizing capacity of antibodies elicited by parainfluenza virus infection of African Green Monkeys is dependent on complement. Virology 2014; 460-461:23-33. [PMID: 25010267 DOI: 10.1016/j.virol.2014.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Revised: 03/29/2014] [Accepted: 05/04/2014] [Indexed: 11/27/2022]
Abstract
The African Green Monkey (AGM) model was used to analyze the role of complement in neutralization of parainfluenza virus. Parainfluenza virus 5 (PIV5) and human parainfluenza virus type 2 were effectively neutralized in vitro by naïve AGM sera, but neutralizing capacity was lost by heat-inactivation. The mechanism of neutralization involved formation of massive aggregates, with no evidence of virion lysis. Following inoculation of the respiratory tract with a PIV5 vector expressing HIV gp160, AGM produced high levels of serum and tracheal antibodies against gp120 and the viral F and HN proteins. However, in the absence of complement these anti-PIV5 antibodies had very poor neutralizing capacity. Virions showed extensive deposition of IgG and C1q with post- but not pre-immune sera. These results highlight the importance of complement in the initial antibody response to parainfluenza viruses, with implications for understanding infant immune responses and design of vaccine strategies for these pediatric pathogens.
Collapse
Affiliation(s)
- Anne E Mayer
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - John B Johnson
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | - Griffith D Parks
- Department of Microbiology and Immunology, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA.
| |
Collapse
|
44
|
Marcotte EL, Ritz B, Cockburn M, Yu F, Heck JE. Exposure to infections and risk of leukemia in young children. Cancer Epidemiol Biomarkers Prev 2014; 23:1195-203. [PMID: 24793957 DOI: 10.1158/1055-9965.epi-13-1330] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Epidemiologic studies indicate that infections in early childhood may protect against pediatric acute lymphoblastic leukemia (ALL). METHODS We identified 3,402 ALL cases among children 0 to 5 years of age using the California Cancer Registry. From California birth records we randomly selected controls in a 20:1 ratio and frequency matched them to cases by birth year. We investigated markers of exposure to infections, including month of birth, timing of birth in relation to influenza and respiratory syncytial virus (RSV) seasons, and birth order based on data from California birth certificates and national infection surveillance systems. RESULTS We observed an increased risk of ALL for spring and summer births, and for those first exposed to an influenza or RSV season at nine to twelve months of age compared with those exposed during the first three months of life, and this association was stronger among first born children [odds ratios (OR), 1.44 and 95% confidence intervals (CI), 1.13-1.82, for influenza exposure at nine to twelve months of age]. Decreased risk was observed with increasing birth order among non-Hispanic whites but not Hispanics (OR, 0.76 and 95% CI, 0.59-096, for fourth or higher birth order among whites). CONCLUSION Our results support the hypothesis that infections in early childhood decrease risk of ALL. IMPACT Our findings implicate early life exposure to infections as protective factors for ALL in young children.
Collapse
Affiliation(s)
- Erin L Marcotte
- Authors' Affiliations: Department of Pediatrics, Division of Epidemiology and Clinical Research, University of Minnesota, Minneapolis, Minnesota; Departments of
| | | | - Myles Cockburn
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, California
| | - Fei Yu
- Biostatistics, Fielding School of Public Health, University of California; and
| | | |
Collapse
|
45
|
Thorstensson R, Trollfors B, Al-Tawil N, Jahnmatz M, Bergström J, Ljungman M, Törner A, Wehlin L, Van Broekhoven A, Bosman F, Debrie AS, Mielcarek N, Locht C. A phase I clinical study of a live attenuated Bordetella pertussis vaccine--BPZE1; a single centre, double-blind, placebo-controlled, dose-escalating study of BPZE1 given intranasally to healthy adult male volunteers. PLoS One 2014; 9:e83449. [PMID: 24421886 PMCID: PMC3885431 DOI: 10.1371/journal.pone.0083449] [Citation(s) in RCA: 100] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2013] [Accepted: 11/01/2013] [Indexed: 11/30/2022] Open
Abstract
Background Acellular pertussis vaccines do not control pertussis. A new approach to offer protection to infants is necessary. BPZE1, a genetically modified Bordetella pertussis strain, was developed as a live attenuated nasal pertussis vaccine by genetically eliminating or detoxifying 3 toxins. Methods We performed a double-blind, placebo-controlled, dose-escalating study of BPZE1 given intranasally for the first time to human volunteers, the first trial of a live attenuated bacterial vaccine specifically designed for the respiratory tract. 12 subjects per dose group received 103, 105 or 107 colony-forming units as droplets with half of the dose in each nostril. 12 controls received the diluent. Local and systemic safety and immune responses were assessed during 6 months, and nasopharyngeal colonization with BPZE1 was determined with repeated cultures during the first 4 weeks after vaccination. Results Colonization was seen in one subject in the low dose, one in the medium dose and five in the high dose group. Significant increases in immune responses against pertussis antigens were seen in all colonized subjects. There was one serious adverse event not related to the vaccine. Other adverse events were trivial and occurred with similar frequency in the placebo and vaccine groups. Conclusions BPZE1 is safe in healthy adults and able to transiently colonize the nasopharynx. It induces immune responses in all colonized individuals. BPZE1 can thus undergo further clinical development, including dose optimization and trials in younger age groups. Trial Registration ClinicalTrials.gov NCT01188512
Collapse
Affiliation(s)
| | - Birger Trollfors
- Swedish Institute for Communicable Disease Control, Solna, Sweden
| | - Nabil Al-Tawil
- Karolinska Trial Alliance, Karolinska University Hospital, Stockholm, Sweden
| | - Maja Jahnmatz
- Swedish Institute for Communicable Disease Control, Solna, Sweden
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Jakob Bergström
- Swedish Institute for Communicable Disease Control, Solna, Sweden
| | | | - Anna Törner
- Swedish Institute for Communicable Disease Control, Solna, Sweden
| | - Lena Wehlin
- Swedish Institute for Communicable Disease Control, Solna, Sweden
| | | | - Fons Bosman
- Q-Biologicals, BioIncubator, Zwijnaarde, Belgium
| | - Anne-Sophie Debrie
- Inserm, Lille, France
- National Center for Scientific Research, Lille, France
- Université Lille-Nord de France, Lille, France
- Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
| | - Nathalie Mielcarek
- Inserm, Lille, France
- National Center for Scientific Research, Lille, France
- Université Lille-Nord de France, Lille, France
- Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
| | - Camille Locht
- Inserm, Lille, France
- National Center for Scientific Research, Lille, France
- Université Lille-Nord de France, Lille, France
- Center for Infection and Immunity of Lille, Institut Pasteur de Lille, Lille, France
| |
Collapse
|
46
|
Haake M, Palzer A, Rist B, Weissenbacher-Lang C, Fachinger V, Eggen A, Ritzmann M, Eddicks M. Influence of age on the effectiveness of PCV2 vaccination in piglets with high levels of maternally derived antibodies. Vet Microbiol 2014; 168:272-80. [DOI: 10.1016/j.vetmic.2013.11.012] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2013] [Revised: 11/02/2013] [Accepted: 11/04/2013] [Indexed: 02/08/2023]
|
47
|
Reber A, Katz J. Immunological assessment of influenza vaccines and immune correlates of protection. Expert Rev Vaccines 2013; 12:519-36. [PMID: 23659300 DOI: 10.1586/erv.13.35] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Influenza vaccines remain the primary public health tool in reducing the ever-present burden of influenza and its complications. In seeking more immunogenic, more effective and more broadly cross-protective influenza vaccines, the landscape of influenza vaccines is rapidly expanding, both in near-term advances and next-generation vaccine design. Although the first influenza vaccines were licensed over 60 years ago, the hemagglutination-inhibition antibody titer is currently the only universally accepted immune correlate of protection against influenza. However, hemagglutination-inhibition titers appear to be less effective at predicting protection in populations at high risk for severe influenza disease; older adults, young children and those with certain medical conditions. The lack of knowledge and validated methods to measure alternate immune markers of protection against influenza remain a substantial barrier to the development of more immunogenic, broadly cross-reactive and effective influenza vaccines. Here, the authors review the knowledge of immune effectors of protection against influenza and discuss assessment methods for a broader range of immunological parameters that could be considered in the evaluation of traditional or new-generation influenza vaccines.
Collapse
Affiliation(s)
- Adrian Reber
- Influenza Division, National Center for Immunization and Respiratory Diseases, Centers for Disease Control and Prevention, 1600 Clifton Road Atlanta, GA 30333, USA
| | | |
Collapse
|
48
|
O'Connor TG, Winter MA, Hunn J, Carnahan J, Pressman EK, Glover V, Robertson-Blackmore E, Moynihan JA, Lee FEH, Caserta MT. Prenatal maternal anxiety predicts reduced adaptive immunity in infants. Brain Behav Immun 2013; 32:21-8. [PMID: 23439080 PMCID: PMC3686987 DOI: 10.1016/j.bbi.2013.02.002] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2012] [Revised: 01/30/2013] [Accepted: 02/11/2013] [Indexed: 01/29/2023] Open
Abstract
Prenatal anxiety has been linked with altered immune function in offspring in animal studies, but the relevance for human health is unknown. We examined prenatal maternal anxiety as a predictor of adaptive immunity in infants at 2 and 6 months of age as part of a prospective longitudinal study. The humoral immune response to hepatitis B vaccine was assessed at 2 months (n=80) and 6 months (n=76) of age. Prenatal anxiety predicted lower hepatitis B antibody titers at 6 months of age independent of obstetric and socio-demographic covariates; the effects were limited to those infants who had not completed the 3-dose vaccine series (for transformed titer values, r=-.36, p<.05). Cell-mediated immune responses at 2 (n=56) and 6 (n=54) months of age were examined by ELISpot assays for interferon(IFN)-γ, interleukin(IL)-2, and IL-4 responder cell frequencies to three antigens: hepatitis B surface antigen, tetanus toxoid, and phytohaemagglutinin (PHA). Prenatal maternal anxiety was associated with reduced IFN-γ and increased IL-4 responder cell frequencies at 6 months of age, independent of obstetric and socio-demographic covariates. No effect of prenatal anxiety was found on adaptive immunity at 2 months of age. The findings provide the first demonstration in humans that prenatal anxiety alters adaptive immunity in the infant.
Collapse
Affiliation(s)
- Thomas G O'Connor
- Department of Psychiatry, University of Rochester Medical Center, Rochester, NY 14642, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Bodewes R, Fraaij PLA, Osterhaus ADME, Rimmelzwaan GF. Pediatric influenza vaccination: understanding the T-cell response. Expert Rev Vaccines 2013; 11:963-71. [PMID: 23002977 DOI: 10.1586/erv.12.69] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Influenza A virus-specific T cells are highly cross-reactive and contribute to heterosubtypic immunity, which may afford protection against novel pandemic strains of influenza virus. However, the magnitude and nature of virus-specific T-cell responses induced by natural infections and/or vaccination in young children is poorly understood. Host factors, such as the development of the immune system during childhood and environmental factors such as exposure rates to influenza viruses and interference by vaccination contribute to shaping the magnitude and specificity of the T-cell response. Here, the authors review several of these factors, including the differences between T-cell responses of young children and adults, the age-dependent frequency of virus-specific T cells and the impact of annual childhood influenza vaccination. In addition, the authors summarize all currently available studies in which influenza vaccine-induced T-cell responses were evaluated. The authors discuss these findings in the light of developing vaccines and vaccination strategies aiming at the induction of protective immunity to seasonal and pandemic influenza viruses of antigenically distinct subtypes.
Collapse
Affiliation(s)
- Rogier Bodewes
- Department of Virology, Erasmus MC, Rotterdam, The Netherlands
| | | | | | | |
Collapse
|
50
|
Agnandji ST, Lell B, Fernandes JF, Abossolo BP, Methogo BGNO, Kabwende AL, Adegnika AA, Mordmüller B, Issifou S, Kremsner PG, Sacarlal J, Aide P, Lanaspa M, Aponte JJ, Machevo S, Acacio S, Bulo H, Sigauque B, Macete E, Alonso P, Abdulla S, Salim N, Minja R, Mpina M, Ahmed S, Ali AM, Mtoro AT, Hamad AS, Mutani P, Tanner M, Tinto H, D'Alessandro U, Sorgho H, Valea I, Bihoun B, Guiraud I, Kaboré B, Sombié O, Guiguemdé RT, Ouédraogo JB, Hamel MJ, Kariuki S, Oneko M, Odero C, Otieno K, Awino N, McMorrow M, Muturi-Kioi V, Laserson KF, Slutsker L, Otieno W, Otieno L, Otsyula N, Gondi S, Otieno A, Owira V, Oguk E, Odongo G, Woods JB, Ogutu B, Njuguna P, Chilengi R, Akoo P, Kerubo C, Maingi C, Lang T, Olotu A, Bejon P, Marsh K, Mwambingu G, Owusu-Agyei S, Asante KP, Osei-Kwakye K, Boahen O, Dosoo D, Asante I, Adjei G, Kwara E, Chandramohan D, Greenwood B, Lusingu J, Gesase S, Malabeja A, Abdul O, Mahende C, Liheluka E, Malle L, Lemnge M, Theander TG, Drakeley C, Ansong D, Agbenyega T, Adjei S, Boateng HO, Rettig T, Bawa J, Sylverken J, Sambian D, Sarfo A, Agyekum A, et alAgnandji ST, Lell B, Fernandes JF, Abossolo BP, Methogo BGNO, Kabwende AL, Adegnika AA, Mordmüller B, Issifou S, Kremsner PG, Sacarlal J, Aide P, Lanaspa M, Aponte JJ, Machevo S, Acacio S, Bulo H, Sigauque B, Macete E, Alonso P, Abdulla S, Salim N, Minja R, Mpina M, Ahmed S, Ali AM, Mtoro AT, Hamad AS, Mutani P, Tanner M, Tinto H, D'Alessandro U, Sorgho H, Valea I, Bihoun B, Guiraud I, Kaboré B, Sombié O, Guiguemdé RT, Ouédraogo JB, Hamel MJ, Kariuki S, Oneko M, Odero C, Otieno K, Awino N, McMorrow M, Muturi-Kioi V, Laserson KF, Slutsker L, Otieno W, Otieno L, Otsyula N, Gondi S, Otieno A, Owira V, Oguk E, Odongo G, Woods JB, Ogutu B, Njuguna P, Chilengi R, Akoo P, Kerubo C, Maingi C, Lang T, Olotu A, Bejon P, Marsh K, Mwambingu G, Owusu-Agyei S, Asante KP, Osei-Kwakye K, Boahen O, Dosoo D, Asante I, Adjei G, Kwara E, Chandramohan D, Greenwood B, Lusingu J, Gesase S, Malabeja A, Abdul O, Mahende C, Liheluka E, Malle L, Lemnge M, Theander TG, Drakeley C, Ansong D, Agbenyega T, Adjei S, Boateng HO, Rettig T, Bawa J, Sylverken J, Sambian D, Sarfo A, Agyekum A, Martinson F, Hoffman I, Mvalo T, Kamthunzi P, Nkomo R, Tembo T, Tegha G, Tsidya M, Kilembe J, Chawinga C, Ballou WR, Cohen J, Guerra Y, Jongert E, Lapierre D, Leach A, Lievens M, Ofori-Anyinam O, Olivier A, Vekemans J, Carter T, Kaslow D, Leboulleux D, Loucq C, Radford A, Savarese B, Schellenberg D, Sillman M, Vansadia P. A phase 3 trial of RTS,S/AS01 malaria vaccine in African infants. N Engl J Med 2012; 367:2284-95. [PMID: 23136909 PMCID: PMC10915853 DOI: 10.1056/nejmoa1208394] [Show More Authors] [Citation(s) in RCA: 567] [Impact Index Per Article: 43.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
BACKGROUND The candidate malaria vaccine RTS,S/AS01 reduced episodes of both clinical and severe malaria in children 5 to 17 months of age by approximately 50% in an ongoing phase 3 trial. We studied infants 6 to 12 weeks of age recruited for the same trial. METHODS We administered RTS,S/AS01 or a comparator vaccine to 6537 infants who were 6 to 12 weeks of age at the time of the first vaccination in conjunction with Expanded Program on Immunization (EPI) vaccines in a three-dose monthly schedule. Vaccine efficacy against the first or only episode of clinical malaria during the 12 months after vaccination, a coprimary end point, was analyzed with the use of Cox regression. Vaccine efficacy against all malaria episodes, vaccine efficacy against severe malaria, safety, and immunogenicity were also assessed. RESULTS The incidence of the first or only episode of clinical malaria in the intention-to-treat population during the 14 months after the first dose of vaccine was 0.31 per person-year in the RTS,S/AS01 group and 0.40 per person-year in the control group, for a vaccine efficacy of 30.1% (95% confidence interval [CI], 23.6 to 36.1). Vaccine efficacy in the per-protocol population was 31.3% (97.5% CI, 23.6 to 38.3). Vaccine efficacy against severe malaria was 26.0% (95% CI, -7.4 to 48.6) in the intention-to-treat population and 36.6% (95% CI, 4.6 to 57.7) in the per-protocol population. Serious adverse events occurred with a similar frequency in the two study groups. One month after administration of the third dose of RTS,S/AS01, 99.7% of children were positive for anti-circumsporozoite antibodies, with a geometric mean titer of 209 EU per milliliter (95% CI, 197 to 222). CONCLUSIONS The RTS,S/AS01 vaccine coadministered with EPI vaccines provided modest protection against both clinical and severe malaria in young infants. (Funded by GlaxoSmithKline Biologicals and the PATH Malaria Vaccine Initiative; RTS,S ClinicalTrials.gov number, NCT00866619.).
Collapse
|