1
|
Kakh M, Doroudchi M, Talepoor A. Induction of Regulatory T Cells After Virus Infection and Vaccination. Immunology 2025. [PMID: 40329764 DOI: 10.1111/imm.13927] [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: 09/24/2024] [Revised: 03/04/2025] [Accepted: 03/17/2025] [Indexed: 05/08/2025] Open
Abstract
Vaccines have been proven to be one of the safest and most effective ways to prevent and combat diseases. However, the main focus has been on the evaluation of the potency of effector mechanisms and the lack of adverse effects of vaccine candidates. Recently, the importance of induced regulatory mechanisms of the immune system after vaccination has come to light. With the increase in our knowledge about these regulatory mechanisms including the regulatory T cells (Tregs), we have come to understand the significance of this arm of the immune system in controlling immunopathology and/or diminishing the effectiveness of vaccines, especially viral vaccines. Tregs play a dual role during infectious diseases by limiting immune-mediated pathology and also contributing to chronic pathogen persistence by decreasing effector immunity and clearance of infection. Tregs may also affect immune responses after vaccination primarily by inhibiting antigen presenting cell function such as cytokine secretion and co-stimulatory molecule expression as well as effector T (Teff) and B cell function. In this article, we review the current knowledge on the induction of Tregs after several life-threatening virus infections and their available vaccines to bring them to the spotlight and emphasise that studying viral-induced antigen-specific Tregs will help us improve the effectiveness and decrease the immunopathology or side effects of viral vaccines. Trial Registration: ClinicalTrials.gov identifier: NCT04357444.
Collapse
Affiliation(s)
- MansourehKarimi Kakh
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mehrnoosh Doroudchi
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - AtefeGhamar Talepoor
- Department of Immunology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Autoimmune Diseases Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
2
|
Domnich A, Trombetta CS, Fallani E, Salvatore M. Immunogenicity and safety of the MF59-adjuvanted seasonal influenza vaccine in non-elderly adults: A systematic review and meta-analysis. PLoS One 2024; 19:e0310677. [PMID: 39775353 PMCID: PMC11684710 DOI: 10.1371/journal.pone.0310677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/27/2024] [Indexed: 01/11/2025] Open
Abstract
OBJECTIVE In Europe, the age indication for the MF59-adjuvanted quadrivalent influenza vaccine (aQIV) has recently been extended from ≥65 to ≥50 years. Considering that the earliest approval of its trivalent formulation (aTIV) in Italy was for people aged ≥12 years, we aimed to systematically appraise data on the immunogenicity, efficacy, and safety of aTIV/aQIV in non-elderly adults. METHODS A systematic literature review was conducted according to the available guidelines and studies were searched in MEDLINE, Biological Abstracts, Web of Science, Cochrane Library and clinical trial registries. Studies on absolute and relative immunogenicity, efficacy, effectiveness, and safety of aTIV/aQIV in non-elderly adults (<65 years) were potentially eligible. These endpoints were analyzed by virus (sub)types and characteristics of vaccinees. Fixed- and random-effects meta-analyses were performed for data synthesis. Protocol registration: CRD42024512472. RESULTS Twenty-four publications were analyzed. aTIV/aQIV was more immunogenic than non-adjuvanted vaccines towards vaccine-like strains: the absolute differences in seroconversion rates were 8.8% (95% CI: 3.7%, 14.0%), 13.1% (95% CI: 6.7%, 19.6%) and 11.7% (95% CI: 7.2%, 16.2%) for A(H1N1), A(H3N2), and B strains, respectively. This immunogenicity advantage was more pronounced in immunosuppressed adults. Additionally, aTIV/aQIV was more immunogenic than non-adjuvanted counterparts towards heterologous A(H3N2) strains with a 10.7% (95% CI: 3.2%, 18.2%) difference in seroconversion rates. Data on antibody persistence and efficacy were limited and inconclusive. Overall, aTIV/aQIV was judged safe and well tolerated, although reactogenic events were more frequent in aTIV/aQIV recipients versus comparators. Serious adverse events were uncommon and no difference (risk ratio 1.02; 95% CI: 0.64, 1.63) between aTIV/aQIV and non-adjuvanted formulations was found. CONCLUSIONS In non-elderly adults, aTIV/aQIV is safe and generally more immunogenic than non-adjuvanted standard-dose vaccines.
Collapse
Affiliation(s)
- Alexander Domnich
- Hygiene Unit, San Martino Policlinico Hospital—IRCCS for Oncology and Neurosciences, Genoa, Italy
| | | | | | - Marco Salvatore
- Seqirus S.r.l., Monteriggioni (Siena), Italy
- Department of Life Sciences; University of Siena, Siena, Italy
| |
Collapse
|
3
|
Sann S, Kleinewietfeld M, Cantaert T. Balancing functions of regulatory T cells in mosquito-borne viral infections. Emerg Microbes Infect 2024; 13:2304061. [PMID: 38192073 PMCID: PMC10812859 DOI: 10.1080/22221751.2024.2304061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/07/2024] [Indexed: 01/10/2024]
Abstract
Mosquito-borne viral infections are on the rise worldwide and can lead to severe symptoms such as haemorrhage, encephalitis, arthritis or microcephaly. A protective immune response following mosquito-borne viral infections requires the generation of a controlled and balanced immune response leading to viral clearance without immunopathology. Here, regulatory T cells play a central role in restoring immune homeostasis. In current review, we aim to provide an overview and summary of the phenotypes of FOXP3+ Tregs in various mosquito-borne arboviral disease, their association with disease severity and their functional characteristics. Furthermore, we discuss the role of cytokines and Tregs in the immunopathogenesis of mosquito-borne infections. Lastly, we discuss possible novel lines of research which could provide additional insight into the role of Tregs in mosquito-borne viral infections in order to develop novel therapeutic approaches or vaccination strategies.
Collapse
Affiliation(s)
- Sotheary Sann
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
| | - Markus Kleinewietfeld
- Department of Immunology, Biomedical Research Institute, Hasselt University, Diepenbeek, Belgium
- VIB Laboratory of Translational Immunomodulation, VIB Center for Inflammation Research (IRC), Hasselt University, Diepenbeek, Belgium
| | - Tineke Cantaert
- Immunology Unit, Institut Pasteur du Cambodge, Pasteur Network, Phnom Penh, Cambodia
| |
Collapse
|
4
|
Kütük T, Onbaşilar İ, Oskay-Halaçli S, Babaoğlu B, Ayhan S, Yalçin SS. Investigation of the Hepatitis-B Vaccine's Immune Response in a Non-Alcoholic Fatty Liver Disease Mouse Model. Vaccines (Basel) 2024; 12:934. [PMID: 39204057 PMCID: PMC11359425 DOI: 10.3390/vaccines12080934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
This study aimed to investigate the immunogenicity of the hepatitis B virus (HBV) vaccine by applying a normal and high-dose hepatitis B virus vaccination program in the mice modeling of non-alcoholic fatty liver disease (NAFLD). NAFLD was induced in mouse livers via diet. At the 10-week mark, both groups were divided into 3 subgroups. While the standard dose vaccination program was applied on days 0, 7, and 21, two high-dose programs were applied: one was applied on days 0 and 7, and the other was applied on days 0, 7, and 21. All mice were euthanized. Blood samples from anti-HB titers; T follicular helper, T follicular regulatory, CD27+, and CD38+ cells; and the liver, spleen, and thymus were taken for histopathologic evaluation. NAFLD subgroups receiving high doses showed higher hepatocyte ballooning scores than normal-dose subgroup. There were differences in CD27+ and CD27+CD38+ cells in animals fed on different diets, without any differences or interactions in terms of vaccine protocols. In the NAFLD group, a negative correlation was observed between anti-HB titers and T helper and CD27+ cells, while a positive correlation was observed with CD38+ cells. NAFLD induced changes in immune parameters in mice, but there was no difference in vaccine efficacy among the applied vaccine protocols. Based on this study's results, high-dose vaccination protocols are not recommended in cases of NAFLD, as they do not enhance efficacy and may lead to increased liver damage.
Collapse
Affiliation(s)
- Tuğba Kütük
- Vaccinology Department, Institute of Vaccinology, Hacettepe University, Ankara 06430, Türkiye; (T.K.); (S.S.Y.)
- Turkish Medicines and Medical Devices Agency, Ankara 06500, Türkiye
| | - İlyas Onbaşilar
- Vaccinology Department, Institute of Vaccinology, Hacettepe University, Ankara 06430, Türkiye; (T.K.); (S.S.Y.)
- Health Science Institute, Hacettepe University, Ankara 06430, Türkiye
- Transgenic Animal Technologies Research and Application Center, Hacettepe University, Ankara 06430, Türkiye
| | - Sevil Oskay-Halaçli
- Department of Basic Sciences of Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06430, Türkiye; (S.O.-H.); (S.A.)
| | - Berrin Babaoğlu
- Department of Pathology, Hacettepe University, Ankara 06430, Türkiye;
| | - Selda Ayhan
- Department of Basic Sciences of Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06430, Türkiye; (S.O.-H.); (S.A.)
| | - Sıddika Songül Yalçin
- Vaccinology Department, Institute of Vaccinology, Hacettepe University, Ankara 06430, Türkiye; (T.K.); (S.S.Y.)
- Department of Social Pediatrics, Institute of Child Health, Hacettepe University, Ankara 06430, Türkiye
| |
Collapse
|
5
|
Sanchez-Felipe L, Alpizar YA, Ma J, Coelmont L, Dallmeier K. YF17D-based vaccines - standing on the shoulders of a giant. Eur J Immunol 2024; 54:e2250133. [PMID: 38571392 DOI: 10.1002/eji.202250133] [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: 02/21/2023] [Revised: 02/11/2024] [Accepted: 02/16/2024] [Indexed: 04/05/2024]
Abstract
Live-attenuated yellow fever vaccine (YF17D) was developed in the 1930s as the first ever empirically derived human vaccine. Ninety years later, it is still a benchmark for vaccines made today. YF17D triggers a particularly broad and polyfunctional response engaging multiple arms of innate, humoral and cellular immunity. This unique immunogenicity translates into an extraordinary vaccine efficacy and outstanding longevity of protection, possibly by single-dose immunization. More recently, progress in molecular virology and synthetic biology allowed engineering of YF17D as a powerful vector and promising platform for the development of novel recombinant live vaccines, including two licensed vaccines against Japanese encephalitis and dengue, even in paediatric use. Likewise, numerous chimeric and transgenic preclinical candidates have been described. These include prophylactic vaccines against emerging viral infections (e.g. Lassa, Zika and SARS-CoV-2) and parasitic diseases (e.g. malaria), as well as therapeutic applications targeting persistent infections (e.g. HIV and chronic hepatitis), and cancer. Efforts to overcome historical safety concerns and manufacturing challenges are ongoing and pave the way for wider use of YF17D-based vaccines. In this review, we summarize recent insights regarding YF17D as vaccine platform, and how YF17D-based vaccines may complement as well as differentiate from other emerging modalities in response to unmet medical needs and for pandemic preparedness.
Collapse
Affiliation(s)
- Lorena Sanchez-Felipe
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Yeranddy A Alpizar
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Ji Ma
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Lotte Coelmont
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| | - Kai Dallmeier
- KU Leuven Department of Microbiology, Immunology and Transplantation, Rega Institute, Molecular Vaccinology and Vaccine Discovery, Leuven, Belgium
| |
Collapse
|
6
|
Laganà A, Visalli G, Di Pietro A, Facciolà A. Vaccinomics and adversomics: key elements for a personalized vaccinology. Clin Exp Vaccine Res 2024; 13:105-120. [PMID: 38752004 PMCID: PMC11091437 DOI: 10.7774/cevr.2024.13.2.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 02/07/2024] [Accepted: 03/12/2024] [Indexed: 05/18/2024] Open
Abstract
Vaccines are one of the most important and effective tools in the prevention of infectious diseases and research about all the aspects of vaccinology are essential to increase the number of available vaccines more and more safe and effective. Despite the unquestionable value of vaccinations, vaccine hesitancy has spread worldwide compromising the success of vaccinations. Currently, the main purpose of vaccination campaigns is the immunization of whole populations with the same vaccine formulations and schedules for all individuals. A personalized vaccinology approach could improve modern vaccinology counteracting vaccine hesitancy and giving great benefits for human health. This ambitious purpose would be possible by facing and deepening the areas of vaccinomics and adversomics, two innovative areas of study investigating the role of a series of variables able to influence the immune response to vaccinations and the development of serious side effects, respectively. We reviewed the recent scientific knowledge about these innovative sciences focusing on genetic and non-genetic basis involved in the individual response to vaccines in terms of both immune response and side effects.
Collapse
Affiliation(s)
- Antonio Laganà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- Istituto Clinico Polispecialistico C.O.T., Cure Ortopediche Traumatologiche S.P.A., Messina, Italy
| | - Giuseppa Visalli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Angela Di Pietro
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Alessio Facciolà
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| |
Collapse
|
7
|
Di Chiara C, Cantarutti A, Raffaella Petrara M, Bonfante F, Benetti E, Boracchini R, Bosa L, Carmona F, Cosma C, Cotugno N, Le Prevost M, Martini G, Meneghel A, Pagliari M, Palma P, Ruffoni E, Zin A, De Rossi A, Giaquinto C, Donà D, Padoan A. Stronger and durable SARS-CoV-2 immune response to mRNA vaccines in 5-11 years old children with prior COVID-19. Vaccine 2024; 42:263-270. [PMID: 38071105 DOI: 10.1016/j.vaccine.2023.12.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/08/2023] [Accepted: 12/01/2023] [Indexed: 01/01/2024]
Abstract
BACKGROUND AND OBJECTIVES mRNA vaccines elicit a durable humoral response to SARS-CoV-2 in adults, whereas evidence in children is scarce. This study aimed to assess the early and long-term immune response to the mRNA vaccine in children with or without previous SARS-CoV-2 infection. METHODS In a multicentre prospective observational study, we profiled the immune response to the Pfizer BioNTech (BNT162b2) vaccine in 5-11-year-old children attending the University Pediatric Hospital of Padua and Bambino-Gesù Hospital in Rome (Italy) from December-2021 to February-2023. Blood samples were collected pre-, 1-, and 6-months after vaccination. Neutralizing antibodies (NAbs) and anti-spike-receptor-binding-domain (anti-S-RBD) IgG titers were analyzed through Plaque Reduction Neutralization Test (PRNT) and chemiluminescent immune-enzymatic assay (CLIA), respectively. Immune cell phenotypes were analyzed by flow cytometry. RESULTS Sixty children (26 [43 %] female, median age = 8 years [IQR = 7-10.7]) were enrolled in the study, including 46 children with a laboratory-confirmed previous COVID-19 (SARS-CoV-2-recovered) and 14 SARS-CoV-2-naïve participants defined as the absence of antigen-specific antibodies before vaccination. SARS-CoV-2-recovered participants recorded higher anti-S-RBD IgG and Wild-type and Omicron BA.2 NAbs titers than SARS-CoV-2-naïve participants at both 1- and 6-months after vaccination. Antibody titers correlated with T (Tregs) and B (Bregs) regulatory cell frequencies in SARS-CoV-2-recovered children. Both SARS-CoV-2-recovered and SARS-CoV-2-naïve participants decreased antibody titers by approximately 100 to 250 % from 1 to 6 months. While children with immunocompromising underlying conditions developed immune responses comparable to those of healthy children, solid organ transplant recipients exhibited lower levels of NAbs and anti-S-RBD IgG titers, as well as reduced frequencies of Tregs and Bregs. CONCLUSIONS mRNA vaccination triggered a higher production of specific anti-SARS-CoV-2 antibodies along with increased levels of regulatory cells in children with previous SARS-CoV-2 infection up to the following 6 months. These findings provide insights into boosting pre-existing immunity.
Collapse
Affiliation(s)
- Costanza Di Chiara
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy; Penta - Child Health Research, Corso Stati Uniti, 4 - 35127 Padua, Italy.
| | - Anna Cantarutti
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, Laboratory of Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1 - 20126 Milan, Italy.
| | - Maria Raffaella Petrara
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2 - 35124 Padua, Italy.
| | - Francesco Bonfante
- Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10 - 35020 Legnaro (Padua), Italy.
| | - Elisa Benetti
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy.
| | - Riccardo Boracchini
- Department of Statistics and Quantitative Methods, Division of Biostatistics, Epidemiology and Public Health, Laboratory of Healthcare Research and Pharmacoepidemiology, University of Milano-Bicocca, Piazza dell'Ateneo Nuovo, 1 - 20126 Milan, Italy.
| | - Luca Bosa
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Francesco Carmona
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64 - 35128 Padua, Italy.
| | - Chiara Cosma
- Department of Laboratory Medicine, University-Hospital of Padova, Via Giambattista Belzoni, 160 - 35121 Padua, Italy.
| | - Nicola Cotugno
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata", Piazza Sant'Onofrio, 4 - 00165 Rome, Italy.
| | - Marthe Le Prevost
- Medical Research Council Clinical Trials Unit at University College London, 90 High Holborn, WC1V 6LJ London, United Kingdom.
| | - Giorgia Martini
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Alessandra Meneghel
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Matteo Pagliari
- Division of Comparative Biomedical Sciences, Istituto Zooprofilattico Sperimentale delle Venezie, Viale dell'Università, 10 - 35020 Legnaro (Padua), Italy.
| | - Paolo Palma
- Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Systems Medicine, University of Rome "Tor Vergata", Piazza Sant'Onofrio, 4 - 00165 Rome, Italy.
| | - Elena Ruffoni
- Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64 - 35128 Padua, Italy.
| | - Annachiara Zin
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy.
| | - Anita De Rossi
- Oncology and Immunology Section, Department of Surgery, Oncology and Gastroenterology, University of Padova, Via Giustiniani, 2 - 35124 Padua, Italy; Immunology and Diagnostic Molecular Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, Via Gattamelata, 64 - 35128 Padua, Italy.
| | - Carlo Giaquinto
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy; Penta - Child Health Research, Corso Stati Uniti, 4 - 35127 Padua, Italy.
| | - Daniele Donà
- Department for Women's and Children's Health, University of Padova, Via Giustiniani, 3 - 35128 Padua, Italy; Penta - Child Health Research, Corso Stati Uniti, 4 - 35127 Padua, Italy.
| | - Andrea Padoan
- Department of Medicine-DIMED, University of Padova, Via Giustiniani 2, 35128 Padua, Italy.
| |
Collapse
|
8
|
Choi JP, Ayoub G, Ham J, Huh Y, Choi SE, Hwang YK, Noh JY, Kim SH, Song JY, Kim ES, Chang YS. Exercise With a Novel Digital Device Increased Serum Anti-influenza Antibody Titers After Influenza Vaccination. Immune Netw 2023; 23:e18. [PMID: 37179746 PMCID: PMC10166655 DOI: 10.4110/in.2023.23.e18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 12/19/2022] [Accepted: 12/28/2022] [Indexed: 05/15/2023] Open
Abstract
It has been reported that some exercise could enhance the anti-viral antibody titers after vaccination including influenza and coronavirus disease 2019 vaccines. We developed SAT-008, a novel digital device, consists of physical activities and activities related to the autonomic nervous system. We assessed the feasibility of SAT-008 to boost host immunity after an influenza vaccination by a randomized, open-label, and controlled study on adults administered influenza vaccines in the previous year. Among 32 participants, the SAT-008 showed a significant increase in the anti-influenza antibody titers assessed by hemagglutination-inhibition test against antigen subtype B Yamagata lineage after 4 wk of vaccination and subtype B Victoria lineage after 12 wk (p<0.05). There was no difference in the antibody titers against subtype "A." The SAT-008 also showed significant increase in the plasma cytokine levels of IL-10, IL-1β, and IL-6 at weeks 4 and 12 after the vaccination (p<0.05). A new approach using the digital device may boost host immunity against virus via vaccine adjuvant-like effects. Trial Registration ClinicalTrials.gov Identifier: NCT04916145.
Collapse
Affiliation(s)
- Jun-Pyo Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Korea
| | | | - Jarang Ham
- S-Alpha Therapeutics, Inc., Seoul 06628, Korea
| | | | | | - Yu-Kyoung Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Korea
| | - Ji Yun Noh
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Sae-Hoon Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Korea
| | - Joon Young Song
- Division of Infectious Diseases, Department of Internal Medicine, Korea University Guro Hospital, Korea University College of Medicine, Seoul 08308, Korea
| | - Eu Suk Kim
- Division of Infectious Diseases, Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Korea
| | - Yoon-Seok Chang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam 13620, Korea
| |
Collapse
|
9
|
Opposite Effects of mRNA-Based and Adenovirus-Vectored SARS-CoV-2 Vaccines on Regulatory T Cells: A Pilot Study. Biomedicines 2023; 11:biomedicines11020511. [PMID: 36831046 PMCID: PMC9953737 DOI: 10.3390/biomedicines11020511] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/27/2023] [Accepted: 01/28/2023] [Indexed: 02/12/2023] Open
Abstract
New-generation mRNA and adenovirus vectored vaccines against SARS-CoV-2 spike protein are endowed with immunogenic, inflammatory and immunomodulatory properties. Recently, BioNTech developed a noninflammatory tolerogenic mRNA vaccine (MOGm1Ψ) that induces in mice robust expansion of antigen-specific regulatory T (Treg) cells. The Pfizer/BioNTech BNT162b2 mRNA vaccine against SARS-CoV-2 is identical to MOGm1Ψ except for the lipid carrier, which differs for containing lipid nanoparticles rather than lipoplex. Here we report that vaccination with BNT162b2 led to an increase in the frequency and absolute count of CD4posCD25highCD127low putative Treg cells; in sharp contrast, vaccination with the adenovirus-vectored ChAdOx1 nCoV-19 vaccine led to a significant decrease of CD4posCD25high cells. This pilot study is very preliminary, suffers from important limitations and, frustratingly, very hardly can be refined in Italy because of the >90% vaccination coverage. Thus, the provocative perspective that BNT162b2 and MOGm1Ψ may share the capacity to promote expansion of Treg cells deserves confirmatory studies in other settings.
Collapse
|
10
|
Barnwal A, Basu B, Tripathi A, Soni N, Mishra D, Banerjee A, Kumar R, Vrati S, Bhattacharyya J. SARS-CoV-2 Spike Protein-Activated Dendritic Cell-Derived Extracellular Vesicles Induce Antiviral Immunity in Mice. ACS Biomater Sci Eng 2022; 8:5338-5348. [PMID: 36445062 PMCID: PMC9717688 DOI: 10.1021/acsbiomaterials.2c01094] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/17/2022] [Indexed: 11/30/2022]
Abstract
The onset and spread of the SARS-CoV-2 virus have created an unprecedented universal crisis. Although vaccines have been developed against the parental SARS-CoV-2, outbreaks of the disease still occur through the appearance of different variants, suggesting a continuous need for improved and effective therapeutic strategies. Therefore, we developed a novel nanovesicle presenting Spike protein on the surface of the dendritic cell-derived extracellular vesicles (DEVs) for use as a potential vaccine platform against SARS-CoV-2. DEVs express peptide/MHC-I (pMHC-I) complexes, CCR-7, on their surface. The immunogenicity and efficacy of the Spike-activated DEVs were tested in mice and compared with free Spike protein. A 1/10 Spike equivalent dose of DEVs showed a superior potency in inducing anti-Spike IgG titers in blood of mice when compared to dendritic cells or free Spike protein treatment. Moreover, DEV-induced sera effectively reduced viral infection by 55-60% within 15 days of booster dose administration. Furthermore, a 1/10 Spike equivalent dose of DEV-treated mice was found to be equally effective in inducing CD19+CD38+ T-cells in the spleen and lymph node; CD8 cells in the bone marrow, spleen, and lymph node; and CD4+CD25+ T-cells in the spleen and lymph node after 90 days of treatment. Thus, our results support the immunogenic nature of DEVs, demonstrating that a low dose of DEVs induces antibodies to inhibit SARS-CoV-2 infection in vitro, therefore warranting further investigations.
Collapse
Affiliation(s)
- Anjali Barnwal
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Science, New Delhi 110029, India
| | - Brohmomoy Basu
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Aarti Tripathi
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Naina Soni
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Debasish Mishra
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Arup Banerjee
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Rajesh Kumar
- Translational Health Science & Technology Institute, Faridabad 121001, Haryana, India
| | - Sudhanshu Vrati
- Laboratory of Virology, Regional Centre for Biotechnology, Faridabad 121001, Haryana, India
| | - Jayanta Bhattacharyya
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, New Delhi 110016, India
- Department of Biomedical Engineering, All India Institute of Medical Science, New Delhi 110029, India
| |
Collapse
|
11
|
Widyasari K, Jang J, Lee S, Kang T, Kim S. Evaluation of the T cell and B cell response following the administration of COVID-19 vaccines in Korea. JOURNAL OF MICROBIOLOGY, IMMUNOLOGY, AND INFECTION = WEI MIAN YU GAN RAN ZA ZHI 2022; 55:1013-1024. [PMID: 36261313 PMCID: PMC9516877 DOI: 10.1016/j.jmii.2022.09.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 12/27/2022]
Abstract
BACKGROUND The coronavirus disease (COVID-19) has been a worldwide concern since 2019. Vaccines are predicted to be crucial in preventing further outbreaks. The development and kinetics of immune responses determine the efficacy of COVID-19 vaccines. METHODS We measured interferon-gamma (IFN-γ) levels upon administering homologous adenovirus vector-based (ChAdOx1-S [AZ], Ad26.COV2.S [JAN]), mRNA-based (BNT162b2 [PF]; mRNA-1273 [MO]), and heterologous (AZ/PF) vaccines in healthy Korean individuals using two IFN-γ release assays: the Covi-FERON ELISA and T-SPOT Discovery SARS-CoV-2 assay. B cell responses were evaluated by assessing the production of neutralizing antibodies by surrogate virus neutralization assay. The immune response among the vaccine groups was compared after adjusting the vaccination dose and interactions between each group. RESULTS AZ triggered the highest T cell response after the first dose but showed instability after the second. PF and MO yielded stable and higher increments of T and B cell responses compared to AZ. MO yielded a higher immune response than PF. JAN yielded T and B cell responses at lower levels than the other vaccines. The booster dose triggered significant increases in the T and B cell responses and is therefore needed to protect against SARS-CoV-2 given the possibility of waning immune responses. CONCLUSION Administering two doses of mRNA vaccines provides the most effective results among the administered vaccines in triggering the immune response specific to SARS-CoV-2 in healthy Korean individuals. Administration of booster doses demonstrated a significant increase in the immune response and may provide longer protection against SARS-CoV-2.
Collapse
Affiliation(s)
- Kristin Widyasari
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, South Korea
| | - Jieun Jang
- Gyeongnam Center for Infectious Disease Control and Prevention, Changwon 51154, South Korea
| | - Seungjun Lee
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, South Korea,Gyeongsang National University College of Medicine, Gyeongsang Institute of Health Sciences, Jinju 52727, South Korea
| | - Taejoon Kang
- Bionanotechnology Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, South Korea,School of Pharmacy, Sungkyunkwan University, Suwon, 16419, South Korea
| | - Sunjoo Kim
- Department of Laboratory Medicine, Gyeongsang National University Changwon Hospital, Changwon, 51472, South Korea,Gyeongnam Center for Infectious Disease Control and Prevention, Changwon 51154, South Korea,Gyeongsang National University College of Medicine, Gyeongsang Institute of Health Sciences, Jinju 52727, South Korea,Corresponding author. Department of Laboratory Medicine, Gyeongsang University Changwon Hospital, Samjungja-ro 11, Seongsan-gu, Changwon-si, Gyeongnam 51472, South Korea. Fax: +82 55 214 3087
| |
Collapse
|
12
|
Distinct immunological and molecular signatures underpinning influenza vaccine responsiveness in the elderly. Nat Commun 2022; 13:6894. [PMID: 36371426 PMCID: PMC9653450 DOI: 10.1038/s41467-022-34487-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 10/26/2022] [Indexed: 11/13/2022] Open
Abstract
Seasonal influenza outbreaks, especially in high-risk groups such as the elderly, represent an important public health problem. Prevailing inadequate efficacy of seasonal vaccines is a crucial bottleneck. Understanding the immunological and molecular mechanisms underpinning differential influenza vaccine responsiveness is essential to improve vaccination strategies. Here we show comprehensive characterization of the immune response of randomly selected elderly participants (≥ 65 years), immunized with the adjuvanted influenza vaccine Fluad. In-depth analyses by serology, multi-parametric flow cytometry, multiplex and transcriptome analysis, coupled to bioinformatics and mathematical modelling, reveal distinguishing immunological and molecular features between responders and non-responders defined by vaccine-induced seroconversion. Non-responders are specifically characterized by multiple suppressive immune mechanisms. The generated comprehensive high dimensional dataset enables the identification of putative mechanisms and nodes responsible for vaccine non-responsiveness independently of confounding age-related effects, with the potential to facilitate development of tailored vaccination strategies for the elderly.
Collapse
|
13
|
Tormo N, Navalpotro D, Martínez-Serrano M, Moreno M, Grosson F, Tur I, Guna MR, Soriano P, Tornero A, Gimeno C. Commercial Interferon-gamma release assay to assess the immune response to first and second doses of mRNA vaccine in previously COVID-19 infected versus uninfected individuals. Diagn Microbiol Infect Dis 2022; 102:115573. [PMID: 35121268 PMCID: PMC8502494 DOI: 10.1016/j.diagmicrobio.2021.115573] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/29/2021] [Accepted: 10/04/2021] [Indexed: 01/01/2023]
Abstract
We analysed immunological response during vaccination by using quantitative anti-spike IgG antibodies (qAbs) and Interferon-gamma (IFNγ) production by SARS-CoV-2-specific CD4+ and CD8+ T cells (QuantiFERON® assay). Blood samples were collected at four time points: a day before the reception of first (T0) and second (T1) BNT162b2 doses, 14 (T2) and 28 days (T3) after second dose. Fifty individuals were included: 34 previously infected by SARS-CoV-2 (CoV2+) and 16 that were not (CoV2-). Among CoV2+, we only observed significant differences after the first dose in both qAbs and IFNγ+ T cells. CoV2- showed differences after each dose, and the response was lower than CoV2+. Older people presented a higher response in CoV2+, while in CoV2, young people responded best. Our results suggest that the second BNT162b2 vaccine dose is not a priority in people with previous COVID-19. QuantiFERON® is a good option to monitor T-cell immunity to SARS-CoV-2.
Collapse
Affiliation(s)
- Nuria Tormo
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain.
| | - David Navalpotro
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain
| | | | - Marta Moreno
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain
| | - Fernando Grosson
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain
| | - Irene Tur
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain
| | - Maria Remedios Guna
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain
| | - Pepa Soriano
- Control Commission of Nursing Homes, Consorcio Hospital General Universitario, Valencia, Spain
| | - Ana Tornero
- Control Commission of Nursing Homes, Consorcio Hospital General Universitario, Valencia, Spain
| | - Concepción Gimeno
- Microbiology Department, Consorcio Hospital General Universitario, Valencia, Spain
| |
Collapse
|
14
|
Bovay A, Fuertes Marraco SA, Speiser DE. Yellow fever virus vaccination: an emblematic model to elucidate robust human immune responses. Hum Vaccin Immunother 2021; 17:2471-2481. [PMID: 33909542 PMCID: PMC8475614 DOI: 10.1080/21645515.2021.1891752] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
By preventing infectious diseases, vaccines contribute substantially to public health. Besides, they offer great opportunities to investigate human immune responses. This is particularly true for live-attenuated virus vaccines which cause resolving acute infections and induce robust immunity. The fact that one can precisely schedule the time-point of vaccination enables complete characterization of the immune response over time, short-term and over many years. The live-attenuated Yellow Fever virus vaccine strain YF-17D was developed in the 1930's and gave rise to the 17D-204 and 17DD vaccine sub-strains, administered to over 600 million individuals worldwide. YF vaccination causes a systemic viral infection, which induces neutralizing antibodies that last for a lifetime. It also induces a strong T cell response resembling the ones of acute infections, in contrast to most other vaccines. In spite of its use since 1937, learning how YF vaccination stimulates such strong and persistent immune responses has gained substantial knowledge only in the last decades. Here we summarize the current state of knowledge on the immune response to YF vaccination, and discuss its contribution as a human model to address complex questions on optimal immune responses.
Collapse
Affiliation(s)
- Amandine Bovay
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Silvia A Fuertes Marraco
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Daniel E Speiser
- Department of Oncology, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Mohamed MMB, Wickman TJ, Fogo AB, Velez JCQ. De Novo Immunoglobulin A Vasculitis Following Exposure to SARS-CoV-2 Immunization. Ochsner J 2021; 21:395-401. [PMID: 34984055 PMCID: PMC8675622 DOI: 10.31486/toj.21.0083] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Background: Immunizations have been previously described as potential triggering events for the development of certain glomerular diseases. However, glomerular disease occurrences are being reported after exposure to a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccine. Case Report: A 50-year-old male presented to a nephrology clinic for evaluation of persistent proteinuria. Six weeks prior to evaluation, the patient had reported developing a rash 2 weeks after receiving the first dose of a SARS-CoV-2 vaccine (BNT162b2 mRNA, Pfizer, Inc). His primary care provider treated the rash with corticosteroids, leading to partial improvement of the skin lesions. Three weeks after the first vaccine injection, the patient received his scheduled second vaccine injection. Within 2 days, the rash reappeared. This time, the lesions were more severe in nature. Skin biopsy revealed immunoglobulin A (IgA)-dominant leukocytoclastic vasculitis. After the patient completed 2 weeks of oral corticosteroids, urinalysis revealed proteinuria, and consultation with nephrology was requested. On examination, healing papules were noted on his legs. Serum creatinine 2 weeks after the second dose of vaccine was 0.9 mg/dL. Microscopic examination of the urinary sediment revealed acanthocytes. Urine protein to creatinine ratio 3 weeks after the second dose of vaccine was 1.1 g/day. Serum complements were normal, and all pertinent serology was negative. Kidney biopsy findings were consistent with IgA nephropathy. Conclusion: The clinical presentation and pathologic findings in this case strongly suggest that the Pfizer SARS-CoV-2 vaccine can trigger a clinical syndrome compatible with Henoch-Schönlein purpura. The recurrence of the rash following the second dose argues for a definite causal association by the Naranjo criteria.
Collapse
Affiliation(s)
| | | | - Agnes B. Fogo
- Department of Pathology, Microbiology and Immunology, Vanderbilt Medical Center, Nashville, TN
| | - Juan Carlos Q. Velez
- Department of Nephrology, Ochsner Clinic Foundation, New Orleans, LA
- The University of Queensland Faculty of Medicine, Ochsner Clinical School, Brisbane, Queensland, Australia
| |
Collapse
|
16
|
Omersel J, Karas Kuželički N. Vaccinomics and Adversomics in the Era of Precision Medicine: A Review Based on HBV, MMR, HPV, and COVID-19 Vaccines. J Clin Med 2020; 9:E3561. [PMID: 33167413 PMCID: PMC7694388 DOI: 10.3390/jcm9113561] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/31/2020] [Accepted: 11/02/2020] [Indexed: 02/06/2023] Open
Abstract
Precision medicine approaches based on pharmacogenomics are now being successfully implemented to enable physicians to predict more efficient treatments and prevention strategies for a given disease based on the genetic background of the patient. This approach has already been proposed for vaccines, but research is lagging behind the needs of society, and precision medicine is far from being implemented here. While vaccinomics concerns the effectiveness of vaccines, adversomics concerns their side effects. This area has great potential to address public concerns about vaccine safety and to promote increased public confidence, higher vaccination rates, and fewer serious adverse events in genetically predisposed individuals. The aim here is to explore the contemporary scientific literature related to the vaccinomic and adversomic aspects of the three most-controversial vaccines: those against hepatitis B, against measles, mumps, and rubella, and against human Papilloma virus. We provide detailed information on the genes that encode human leukocyte antigen, cytokines and their receptors, and transcription factors and regulators associated with the efficacy and safety of the Hepatitis B and Measles, Mumps and Rubella virus vaccines. We also investigate the future prospects of vaccinomics and adversomics of a COVID-19 vaccine, which might represent the fastest development of a vaccine ever.
Collapse
Affiliation(s)
| | - Nataša Karas Kuželički
- Faculty of Pharmacy, University of Ljubljana, Aškerčeva cesta 7, 1000 Ljubljana, Slovenia;
| |
Collapse
|
17
|
Nguyen LN, Parikh SU, Batliwala SY, Davis AS, Riaz KM. Temporal Profile and Treatment of Purpureocillium lilacinum Keratitis Secondary to Herpes Zoster Reactivation Following Influenza Vaccination. Int Med Case Rep J 2020; 13:455-459. [PMID: 33061667 PMCID: PMC7519859 DOI: 10.2147/imcrj.s265724] [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: 06/04/2020] [Accepted: 08/18/2020] [Indexed: 12/23/2022] Open
Abstract
Purpose To report a temporal profile of Purpureocillium lilacinum keratitis (PLK) secondary to immune dysfunction induced by the combination of reactivation of herpes zoster dermatitis and recent influenza vaccination that suggests a possible association, including successful medical management. Methods A 64-year-old contact lens wearer presented with left eye keratitis days after receiving an influenza vaccination and subsequent development of herpes zoster lesions on the flank. Patient was initially treated for bacterial keratitis with fortified antibiotics and oral valacyclovir for her concurrent zoster. Pharmacotherapy was changed to topical voriconazole after cultures were positive for Purpureocillium lilacinum. Topography and anterior segment OCT demonstrated scarring at multiple levels within the cornea with irregular astigmatism. A literature review was conducted to identify mechanisms that demonstrate a temporal link between influenza vaccination, herpes zoster reactivation, and fungal keratitis. Results After the conclusion of topical therapy, the central corneal infiltrate regressed and a partial light-blocking anterior stromal scar remained. Best corrected visual acuity improved from 20/400 to 20/25. Conclusion Transient systemic immune dysregulation, secondary to influenza vaccination and reactivation of systemic herpetic disease, compounded by contact lens wear, may create a favorable environment for opportunistic fungal keratitis. This case highlights the importance of adequately assessing and treating for existing comorbidities in the successful treatment of mycotic keratitis.
Collapse
Affiliation(s)
- Lindsey N Nguyen
- College of Medicine, University of Oklahoma, Oklahoma City, OK, USA
| | | | - Shehzad Y Batliwala
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma, Oklahoma City, OK, USA
| | - Alexander S Davis
- Division of Ophthalmology, University of New Mexico, Albuquerque, NM, USA
| | - Kamran M Riaz
- Dean McGee Eye Institute, Department of Ophthalmology, University of Oklahoma, Oklahoma City, OK, USA
| |
Collapse
|
18
|
Wang S, Gao S, Zhou D, Qian X, Luan J, Lv X. The role of the CD39-CD73-adenosine pathway in liver disease. J Cell Physiol 2020; 236:851-862. [PMID: 32648591 DOI: 10.1002/jcp.29932] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 02/06/2023]
Abstract
Extracellular adenosine triphosphate (ATP) is a danger signal released by dying and damaged cells, and it functions as an immunostimulatory signal that promotes inflammation. The ectonucleotidases CD39/ectonucleoside triphosphate diphosphohydrolase-1 and CD73/ecto-5'-nucleotidase are cell-surface enzymes that breakdown extracellular ATP into adenosine. This drives a shift from an ATP-driven proinflammatory environment to an anti-inflammatory milieu induced by adenosine. The CD39-CD73-adenosine pathway changes dynamically with the pathophysiological context in which it is embedded. Accumulating evidence suggests that CD39 and CD73 play important roles in liver disease as critical components of the extracellular adenosinergic pathway. Recent studies have shown that the modification of the CD39-CD73-adenosine pathway alters the liver's response to injury. Moreover, adenosine exerts different effects on the pathophysiology of the liver through different receptors. In this review, we aim to describe the role of the CD39-CD73-adenosine pathway and adenosine receptors in liver disease, highlighting potential therapeutic targets in this pathway, which will facilitate the development of therapeutic strategies for the treatment of liver disease.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China.,The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Institute for Liver Disease, Anhui Medical University, Hefei, Anhui, China
| | - Songsen Gao
- Department of Orthopedics (Spinal Surgery), The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Dexi Zhou
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Xueyi Qian
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Xiongwen Lv
- The Key Laboratory of Anti-Inflammatory and Immune Medicines, Ministry of Education, School of Pharmacy, Institute for Liver Disease, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
19
|
Characterization of potential biomarkers of reactogenicity of licensed antiviral vaccines: randomized controlled clinical trials conducted by the BIOVACSAFE consortium. Sci Rep 2019; 9:20362. [PMID: 31889148 PMCID: PMC6937244 DOI: 10.1038/s41598-019-56994-8] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 12/10/2019] [Indexed: 02/08/2023] Open
Abstract
Biomarkers predictive of inflammatory events post-vaccination could accelerate vaccine development. Within the BIOVACSAFE framework, we conducted three identically designed, placebo-controlled inpatient/outpatient clinical studies (NCT01765413/NCT01771354/NCT01771367). Six antiviral vaccination strategies were evaluated to generate training data-sets of pre-/post-vaccination vital signs, blood changes and whole-blood gene transcripts, and to identify putative biomarkers of early inflammation/reactogenicity that could guide the design of subsequent focused confirmatory studies. Healthy adults (N = 123; 20-21/group) received one immunization at Day (D)0. Alum-adjuvanted hepatitis B vaccine elicited vital signs and inflammatory (CRP/innate cells) responses that were similar between primed/naive vaccinees, and low-level gene responses. MF59-adjuvanted trivalent influenza vaccine (ATIV) induced distinct physiological (temperature/heart rate/reactogenicity) response-patterns not seen with non-adjuvanted TIV or with the other vaccines. ATIV also elicited robust early (D1) activation of IFN-related genes (associated with serum IP-10 levels) and innate-cell-related genes, and changes in monocyte/neutrophil/lymphocyte counts, while TIV elicited similar but lower responses. Due to viral replication kinetics, innate gene activation by live yellow-fever or varicella-zoster virus (YFV/VZV) vaccines was more suspended, with early IFN-associated responses in naïve YFV-vaccine recipients but not in primed VZV-vaccine recipients. Inflammatory responses (physiological/serum markers, innate-signaling transcripts) are therefore a function of the vaccine type/composition and presence/absence of immune memory. The data reported here have guided the design of confirmatory Phase IV trials using ATIV to provide tools to identify inflammatory or reactogenicity biomarkers.
Collapse
|
20
|
Schmidt ST, Pedersen GK, Christensen D. Rational Design and In Vivo Characterization of Vaccine Adjuvants. ILAR J 2019; 59:309-322. [PMID: 30624655 DOI: 10.1093/ilar/ily018] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Revised: 09/05/2018] [Indexed: 12/14/2022] Open
Abstract
Many different adjuvants are currently being developed for subunit vaccines against a number of pathogens and diseases. Rational design is increasingly used to develop novel vaccine adjuvants, which requires extensive knowledge of, for example, the desired immune responses, target antigen-presenting cell subsets, their localization, and expression of relevant pattern-recognition receptors. The adjuvant mechanism of action and efficacy are usually evaluated in animal models, where mice are by far the most used. In this review, we present methods for assessing adjuvant efficacy and function in animal models: (1) whole-body biodistribution evaluated by using fluorescently and radioactively labeled vaccine components; (2) association and activation of immune cell subsets at the injection site, in the draining lymph node, and the spleen; (4) adaptive immune responses, such as cytotoxic T-lymphocytes, various T-helper cell subsets, and antibody responses, which may be quantitatively evaluated using ELISA, ELISPOT, and immunoplex assays and qualitatively evaluated using flow cytometric and single cell sequencing assays; and (5) effector responses, for example, antigen-specific cytotoxic potential of CD8+ T cells and antibody neutralization assays. While the vaccine-induced immune responses in mice often correlate with the responses induced in humans, there are instances where immune responses detected in mice are not translated to the human situation. We discuss some examples of correlation and discrepancy between mouse and human immune responses and how to understand them.
Collapse
Affiliation(s)
- Signe Tandrup Schmidt
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| | - Gabriel Kristian Pedersen
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| | - Dennis Christensen
- Statens Serum Institut, Center for Vaccine Research, Department of Infectious Disease Immunology, Copenhagen S, Denmark
| |
Collapse
|
21
|
Messina NL, Zimmermann P, Curtis N. The impact of vaccines on heterologous adaptive immunity. Clin Microbiol Infect 2019; 25:1484-1493. [PMID: 30797062 DOI: 10.1016/j.cmi.2019.02.016] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Revised: 02/11/2019] [Accepted: 02/13/2019] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vaccines induce antigen-specific memory in adaptive immune cells that enables long-lived protection against the target pathogen. In addition to this, several vaccines have beneficial effects greater than protection against their target pathogen. These non-specific effects are proposed to be the result of vaccine-induced immunomodulation. In the case of bacille Calmette-Guérin (BCG) vaccine, this involves induction of innate immune memory, termed 'trained immunity', in monocytes and natural killer cells. OBJECTIVES This review discusses current evidence for vaccine-induced immunomodulation of adaptive immune cells and heterologous adaptive immune responses. CONTENT The three vaccines that have been associated with changes in all-cause infant mortality: BCG, diphtheria-tetanus-pertussis (DTP) and measles-containing vaccines (MCV) alter T-cell and B-cell immunity. The majority of studies that investigated non-specific effects of these vaccines on the adaptive immune system report changes in numbers or proportions of adaptive immune cell populations. However, there is also evidence for effects of these vaccines on adaptive immune cell function and responses to heterologous stimuli. There is some evidence that, in addition to BCG, DTP and MCV, other vaccines (that have not been associated with changes in all-cause mortality) may alter adaptive immune responses to unrelated stimuli. IMPLICATIONS This review concludes that vaccines alter adaptive immune cell populations and heterologous immune responses. The non-specific effects differ between various vaccines and their effects on heterologous adaptive immune responses may also involve bystander activation, cross-reactivity and other as yet undefined mechanisms. This has major implications for future vaccine design and vaccination scheduling.
Collapse
Affiliation(s)
- N L Messina
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia.
| | - P Zimmermann
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia; Department of Paediatrics, Fribourg Hospital HFR, Fribourg, Switzerland
| | - N Curtis
- Infectious Diseases Research Group, Murdoch Children's Research Institute, Parkville, VIC, Australia; Department of Paediatrics, The University of Melbourne, Parkville, VIC, Australia; Infectious Diseases Unit, The Royal Children's Hospital Melbourne, Parkville, VIC, Australia
| |
Collapse
|
22
|
Chissumba RM, Luciano A, Namalango E, Bauer A, Bhatt N, Wahren B, Nilsson C, Geldmacher C, Scarlatti G, Jani I, Kestens L. Regulatory T cell abundance and activation status before and after priming with HIVIS-DNA and boosting with MVA-HIV/rgp140/GLA-AF may impact the magnitude of the vaccine-induced immune responses. Immunobiology 2018; 223:792-801. [PMID: 30121146 DOI: 10.1016/j.imbio.2018.08.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 08/11/2018] [Indexed: 01/27/2023]
Abstract
BACKGROUND Little is known about regulatory CD4 T cells (Tregs) in the context of HIV vaccines. Tregs can be differentiated into resting (FoxP3+CD45RA+ - rTregs), activated (FoxP3HighCD45RA- - aTregs) and memory (FoxP3LowCD45RA- - mTregs). Tregs, as CD4 T cells, are also frequent targets for HIV infection. We studied how the abundance and phenotypes of Tregs in terms of activation status and expression of HIV-1 binding molecules would have changed during vaccination in healthy volunteers participating in a phase IIa HIV vaccine clinical trial. Subjects were primed three times with HIVIS-DNA and boosted twice with MVA-CMDR-HIV alone (n = 12) or MVA-CMDR combined with protein CN54rgp140 (n = 13). The proportions of β7 integrin in all CD4 T cells and in the Tregs subset decreased moderately after the final vaccination (p = 0.001 and p = 0.033, respectively) and the rTregs proportion within the total Tregs were also decreased after the final vaccination (p = 0.038). All these proportions returned to normal values within the three months after the final vaccination. The magnitude of HIV-Envelope-specific IFNγ + T cells after vaccination (r = 0.66; p = 0.021) correlated directly with the proportion of Tregs, and correlated inversely correlated with ratios of Th17/Tregs (r = -0.75; p = 0.0057) and Th17/mTregs (r = -0.78; p = 0.0065). Higher titers of IgG gp140 antibodies were observed in subjects with higher mTregs proportions (r = 0.52; p = 0.022). Interestingly, pre-vaccination levels of mTregs correlated with vaccine-induced Env-binding antibodies (r = 0.57; p = 0.01) and presence of neutralizing antibodies (r = 0.61; p = 0.01), while the pre-vaccination Th17/mTregs ratio correlated inversely with the magnitude of cellular IFN-γ ELISpot responses (r = -0.9; p = 0.002). Taken together, these results suggest that pre- and post-vaccination Tregs, their activation status, the Th17/Tregs ratio and other host factors affecting Treg abundance, have an impact on the magnitude of HIV vaccine-induced immune responses. Moreover, the DNA-HIVIS/MVA-HIV regimen, alone or in combination with CN54rgp140 induced moderate and temporary alterations of the Tregs activation status. We also show a decrease in expression of the HIV-1 ligand β7 integrin on Tregs and all CD4 T cells.
Collapse
Affiliation(s)
- Raquel Matavele Chissumba
- Instituto Nacional de Saúde, Ministry of Health, Maputo, Mozambique; Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium.
| | - Abílio Luciano
- Instituto de Ciências de Saúde, Ministry of Health, Maputo, Mozambique
| | | | - Asli Bauer
- National Institute for Medical Research, Mbeya Medical Research Center, Mbeya, Tanzania
| | - Nilesh Bhatt
- Instituto Nacional de Saúde, Ministry of Health, Maputo, Mozambique
| | - Britta Wahren
- Department of Microbiology Tumor and Cell Biology, Karolinska Institutet, 17177 Stockholm, Sweden
| | - Charlotta Nilsson
- Department of Microbiology, Public Health Agency of Sweden, Solna, Sweden; Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet, Huddinge, Sweden
| | - Christof Geldmacher
- Division of Infectious Diseases and Tropical Medicine, Klinikum of the University of Munich (LMU), Munich, Germany; German Center for Infection Research (DZIF), Partner Site Munich, Munich, Germany
| | - Gabriella Scarlatti
- Viral Evolution and Transmission Unit, Department of Immunology, Transplant and Infectious Diseases, San Raffaele Scientific Institute, Milan, Italy
| | - Ilesh Jani
- Instituto Nacional de Saúde, Ministry of Health, Maputo, Mozambique
| | - Luc Kestens
- Department of Biomedical Sciences, Institute of Tropical Medicine, Antwerp, Belgium; Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|