1
|
Reis LR, Silva-Moraes V, Teixeira-Carvalho A, Ross TM. B-cell dynamics underlying poor response upon split-inactivated influenza virus vaccination. Front Immunol 2024; 15:1481910. [PMID: 39635527 PMCID: PMC11614812 DOI: 10.3389/fimmu.2024.1481910] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 10/24/2024] [Indexed: 12/07/2024] Open
Abstract
This investigation elucidated the differences in humoral and H1N1 HA-specific memory B-cells response in participants exhibiting distinct immune response patterns prior to and after vaccination with Fluzone, the quadrivalent split-inactivated seasonal influenza virus vaccine. Participants were categorized into persistent non-responders and persistent responders based on their hemagglutination-inhibition (HAI) antibody titers to the H1N1 component from each vaccine administered between the 2019-2020 to 2023-2024 seasons. Persistent responders had higher fold change in H1N1 HA-specific CD21 expressing B-cells, plasmablasts, and plasma cells. A significant increase in H1N1 HA-specific transitional B-cells in persistent non-responders was observed. The frequency and fold change of H1N1-specific IgM-expressing memory B-cells was higher in persistent non-responders. Dimensionality reduction analysis also demonstrated higher IgM expression for persistent non-responders than persistent responders. Furthermore, persistent non-responders had a significant fold change increase in IgA tissue-like memory, IgG exhausted tissue-like memory, and double negative (DN) activated memory cells. In contrast, persistent responders had increased frequency of IgG-activated memory B-cells, IgG resting B-cells and DN resting B-cells. Correlation analysis revealed a positive correlation between HAI titers and DN memory B-cells and a negative correlation between HAI titers and IgG-expressing memory B-cells in persistent non-responders. Conversely, persistent responders had a positive correlation between HAI titers and IgA resting memory B-cells and a negative correlation between IgG memory B-cells and DN memory B-cells. Overall, this study provided valuable insights into the differential immune memory B-cell responses following influenza virus vaccination and paves the way for future research to further unravel the complexities of vaccine-induced memory B-cells and ultimately improve vaccination strategies against influenza virus infection.
Collapse
Affiliation(s)
- Laise Rodrigues Reis
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, United States
| | - Vanessa Silva-Moraes
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, United States
| | | | - Ted M. Ross
- Florida Research and Innovation Center, Cleveland Clinic, Port Saint Lucie, FL, United States
- Department of Infection Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, United States
- Department of Infectious Diseases, University of Georgia, Athens, GA, United States
- Center for Vaccines and Immunology, University of Georgia, Athens, GA, United States
| |
Collapse
|
2
|
Manfroi B, Cuc BT, Sokal A, Vandenberghe A, Temmam S, Attia M, El Behi M, Camaglia F, Nguyen NT, Pohar J, Salem-Wehbe L, Pottez-Jouatte V, Borzakian S, Elenga N, Galeotti C, Morelle G, de Truchis de Lays C, Semeraro M, Romain AS, Aubart M, Ouldali N, Mahuteau-Betzer F, Beauvineau C, Amouyal E, Berthaud R, Crétolle C, Arnould MD, Faye A, Lorrot M, Benoist G, Briand N, Courbebaisse M, Martin R, Van Endert P, Hulot JS, Blanchard A, Tartour E, Leite-de-Moraes M, Lezmi G, Ménager M, Luka M, Reynaud CA, Weill JC, Languille L, Michel M, Chappert P, Mora T, Walczak AM, Eloit M, Bacher P, Scheffold A, Mahévas M, Sermet-Gaudelus I, Fillatreau S. Preschool-age children maintain a distinct memory CD4 + T cell and memory B cell response after SARS-CoV-2 infection. Sci Transl Med 2024; 16:eadl1997. [PMID: 39292802 DOI: 10.1126/scitranslmed.adl1997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Accepted: 07/19/2024] [Indexed: 09/20/2024]
Abstract
The development of the human immune system lasts for several years after birth. The impact of this maturation phase on the quality of adaptive immunity and the acquisition of immunological memory after infection at a young age remains incompletely defined. Here, using an antigen-reactive T cell (ARTE) assay and multidimensional flow cytometry, we profiled circulating severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-reactive CD3+CD4+CD154+ T cells in children and adults before infection, during infection, and 11 months after infection, stratifying children into separate age groups and adults according to disease severity. During SARS-CoV-2 infection, children younger than 5 years old displayed a lower antiviral CD4+ T cell response, whereas children older than 5 years and adults with mild disease had, quantitatively and phenotypically, comparable virus-reactive CD4+ T cell responses. Adults with severe disease mounted a response characterized by higher frequencies of virus-reactive proinflammatory and cytotoxic T cells. After SARS-CoV-2 infection, preschool-age children not only maintained neutralizing SARS-CoV-2-reactive antibodies postinfection comparable to adults but also had phenotypically distinct memory T cells displaying high inflammatory features and properties associated with migration toward inflamed sites. Moreover, preschool-age children had markedly fewer circulating virus-reactive memory B cells compared with the other cohorts. Collectively, our results reveal unique facets of antiviral immunity in humans at a young age and indicate that the maturation of adaptive responses against SARS-CoV-2 toward an adult-like profile occurs in a progressive manner.
Collapse
Affiliation(s)
- Benoît Manfroi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Bui Thi Cuc
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Aurélien Sokal
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine interne, Hôpital Beaujon, Assistance Publique-Hôpitaux de Paris (AP-HP), 92110 Clichy, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Alexis Vandenberghe
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Sarah Temmam
- Pathogen Discovery Laboratory, Institut Pasteur, Université Paris Cité, and Institut Pasteur, the WOAH Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Université Paris Cité, 75015 Paris, France
| | - Mikaël Attia
- Molecular Genetics of RNA Viruses, Department of Virology, Institut Pasteur, Université Paris-Cité, CNRS UMR 3569, 75015 Paris, France
| | - Mohamed El Behi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Francesco Camaglia
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Ngan Thu Nguyen
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Jelka Pohar
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Immunology and Cellular Immunotherapy (ICI) Group, Department of Genetic Toxicology and Cancer Biology, National Institute of Biology, 1000 Ljubljana, Slovenia
| | - Layale Salem-Wehbe
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Valentine Pottez-Jouatte
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Sibyline Borzakian
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Narcisse Elenga
- Service de Pédiatrie, Centre Hospitalier de Cayenne, 97300 French Guiana
| | - Caroline Galeotti
- Department of Pediatric Rheumatology, Bicêtre Hospital, AP-HP, Paris-Saclay University, 94275 Le Kremlin-Bicêtre, France
| | - Guillaume Morelle
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris Saclay, 94275 Le Kremlin-Bicêtre, France
| | - Camille de Truchis de Lays
- Service de Pédiatrie. Hôpital Jean-Verdier, AP-HP, Hôpitaux Universitaires Paris Seine-Saint-Denis, 93140 Bondy, France
| | - Michaela Semeraro
- University of Paris Cité, and Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Anne-Sophie Romain
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, General Paediatrics Department, 75012 Paris, France
| | - Mélodie Aubart
- INSERM U1163, Genetic Predisposition to Infectious Diseases, Imagine Institute, Université Paris Cité, Paris F-75015, France
- Pediatric Neurology Department, Necker-Enfants Malades Universitary Hospital, AP-HP, Paris-Cité University, 75015 Paris, France
| | - Naim Ouldali
- Department of General Pediatrics, Pediatric Infectious Disease and Internal Medicine, Robert Debré University Hospital, Assistance Publique-Hôpitaux de Paris, 75019 Paris, France
- Paris Cité University, INSERM UMR 1137, Infection, Antimicrobials, Modelling, Evolution (IAME), 75018 Paris, France
| | - Florence Mahuteau-Betzer
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Claire Beauvineau
- CNRS UMR 9187, INSERM U1196, Chemistry and Modeling for the Biological of Cancer, Institut Curie, PSL Research University, 91405 Orsay, France
- Université Paris-Saclay, 91405 Orsay, France
| | - Elsa Amouyal
- SIREDO Pediatric Oncology Center, Institut Curie, Paris-Science Lettres University, 75005 Paris, France
| | - Romain Berthaud
- Pediatric Nephrology, Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA) Reference Center, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
| | - Célia Crétolle
- Département de Pédiatrie, Service de Chirurgie viscérale pédiatrique, Hôpital Universitaire Necker-Enfants Malades, GH Paris Centre, 75015 Paris, France
| | - Marc Duval Arnould
- Department of General Paediatrics, Hôpital Bicêtre, AP-HP, University of Paris Saclay, 94275 Le Kremlin-Bicêtre, France
| | - Albert Faye
- Pediatric Neurology Department, Necker-Enfants Malades Universitary Hospital, AP-HP, Paris-Cité University, 75015 Paris, France
| | - Mathie Lorrot
- Sorbonne Université, Assistance Publique-Hôpitaux de Paris, Trousseau Hospital, General Paediatrics Department, 75012 Paris, France
| | - Grégoire Benoist
- Service de pédiatrie générale et hôpital de jour allergologie, CHU Ambroise-Paré, AP-HP, 92100 Boulogne-Billancourt, France
| | - Nelly Briand
- University of Paris Cité, and Clinical Investigation Center, Clinical Research Unit, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
| | - Marie Courbebaisse
- Faculté de Médecine, Université Paris Cité, 75015 Paris, France
- Explorations fonctionnelles rénales, Physiologie, Hôpital européen Georges-Pompidou, Assistance Publique-Hôpitaux de Paris, 75908 Paris Cedex 15, France
| | - Roland Martin
- Institute of Experimental Immunology, University of Zurich, Winterthurerstrasse 190, 8057 Zurich, Switzerland
- Therapeutic Immune Design, Center for Molecular Medicine, Department of Clinical Neuroscience, Karolinska Institute, 171 76 Stockholm, Sweden
| | - Peter Van Endert
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| | - Jean-Sébastien Hulot
- PARCC, INSERM, Université Paris Cité, 75015 Paris, France
- Centre d'Investigation Clinique, AP-HP, INSERM CIC-1418, Européen Georges Pompidou Hospital, 75015 Paris, France
| | - Anne Blanchard
- Centre d'Investigation Clinique, AP-HP, INSERM CIC-1418, Européen Georges Pompidou Hospital, 75015 Paris, France
- Sorbonne Paris Cité, Paris Descartes University, 75015 Paris, France
| | - Eric Tartour
- Pediatric Nephrology, Maladies Rénales Héréditaires de l'Enfant et de l'Adulte (MARHEA) Reference Center, Necker-Children's Hospital, Assistance Publique-Hôpitaux de Paris, 75015 Paris, France
- PARCC, INSERM, Université Paris Cité, 75015 Paris, France
- Department of Immunology, Hôpital Européen Georges-Pompidou, AP-HP, CEDEX 15, 75908 Paris, France
| | - Maria Leite-de-Moraes
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
| | - Guillaume Lezmi
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- AP-HP, Hôpital Necker-Enfants Malades, Service de Pneumologie et Allergologie Pédiatriques, 75015 Paris, France
| | - Mickael Ménager
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Université Paris Cité, Imagine Institute, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, 75015 Paris, France
| | - Marine Luka
- Laboratory of Inflammatory Responses and Transcriptomic Networks in Diseases, Atip-Avenir Team, Université Paris Cité, Imagine Institute, 75015 Paris, France
- Labtech Single-Cell@Imagine, Imagine Institute, 75015 Paris, France
| | - Claude-Agnès Reynaud
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
| | - Jean-Claude Weill
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
| | - Laetitia Languille
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Pascal Chappert
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Thierry Mora
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Aleksandra M Walczak
- Laboratoire de physique de l'École normale supérieure, CNRS, Paris Sciences et Lettres (PSL) University, Sorbonne Université, and Université de Paris, 75005 Paris, France
| | - Marc Eloit
- Pathogen Discovery Laboratory, Institut Pasteur, Université Paris Cité, and Institut Pasteur, the WOAH Collaborating Center for the Detection and Identification in Humans of Emerging Animal Pathogens, Université Paris Cité, 75015 Paris, France
- Ecole Nationale Vétérinaire d'Alfort, University of Paris-Est, 94700 Maisons-Alfort, France
| | - Petra Bacher
- Institute of Immunology, Christian-Albrecht Universität zu Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
- Institute of Clinical Molecular Biology, Christian-Albrecht University of Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrecht Universität zu Kiel and UKSH Schleswig-Holstein, 24105 Kiel, Germany
| | - Matthieu Mahévas
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Action thématique incitative sur programme-Avenir Team, Auto-Immune and Immune B cells, F-75015 Paris, France
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
- INSERM U955, équipe 2. Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), 94000 Créteil, France
| | - Isabelle Sermet-Gaudelus
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Reference Center for Rare Diseases: Cystic Fibrosis and Other Epithelial Respiratory Protein Misfolding Diseases, Hôpital Necker-Enfants Malades, AP-HP Centre Université Paris Cité, 75015 Paris, France
| | - Simon Fillatreau
- Université Paris Cité, INSERM U1151, CNRS UMR8253, Institut Necker Enfants Malades-INEM, F-75015 Paris, France
- Necker-Enfants Malades Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), 75015 Paris, France
- Faculté de Médecine, Université Paris Cité, 75015 Paris, France
- Service Immunologie Biologique, AP-HP, Hôpital Universitaire Necker-Enfants Malades, F-75015 Paris, France
| |
Collapse
|
3
|
Seong H, Yoon JG, Nham E, Choi YJ, Noh JY, Cheong HJ, Kim WJ, Kim EH, Kim C, Han YH, Lim S, Song JY. The gut microbiota modifies antibody durability and booster responses after SARS-CoV-2 vaccination. J Transl Med 2024; 22:827. [PMID: 39242525 PMCID: PMC11380214 DOI: 10.1186/s12967-024-05637-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 08/26/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) vaccines are pivotal in combating coronavirus disease 2019 (COVID-19); however, the declining antibody titers postvaccination pose challenges for sustained protection and herd immunity. Although gut microbiome is reported to affect the early antibody response after vaccination, its impact on the longevity of vaccine-induced antibodies remains unexplored. METHODS A prospective cohort study was conducted involving 44 healthy adults who received two doses of either the BNT162b2 or ChAdOx1 vaccine, followed by a BNT162b2 booster at six months. The gut microbiome was serially analyzed using 16S rRNA and shotgun sequencing, while humoral immune response was assessed using a SARS-CoV-2 spike protein immunoassay. RESULTS Faecalibacterium prausnitzii was associated with robust and persistent antibody responses post-BNT162b2 vaccination. In comparison, Escherichia coli was associated with a slower antibody decay following ChAdOx1 vaccination. The booster immune response was correlated with metabolic pathways involving cellular functions and aromatic amino acid synthesis. CONCLUSIONS The findings of this study underscored the potential interaction between the gut microbiome and the longevity/boosting effect of antibodies following vaccination against SARS-CoV-2. The identification of specific microbial associations suggests the prospect of microbiome-based strategies for enhancing vaccine efficacy.
Collapse
Affiliation(s)
- Hye Seong
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Jin Gu Yoon
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Eliel Nham
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Yu Jung Choi
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Ji Yun Noh
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Hee Jin Cheong
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Woo Joo Kim
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
| | - Eui Ho Kim
- Viral Immunology Laboratory, Institut Pasteur Korea, Seongnam, Republic of Korea
| | - Chulwoo Kim
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea
- Department of Microbiology, Korea University College of Medicine, Seoul, Republic of Korea
| | - Young-Hee Han
- Department of Food and Nutrition, Chungbuk National University, Cheongju, Republic of Korea
| | - Sooyeon Lim
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea.
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea.
| | - Joon Young Song
- Department of Internal Medicine, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea.
- Asia Pacific Influenza Institute, Guro Hospital, Korea University College of Medicine, Gurodong-Ro 148, Guro-Gu, Seoul, 08308, Republic of Korea.
- Vaccine Innovation Center-KU Medicine, Seoul, Republic of Korea.
| |
Collapse
|
4
|
van der Straten K, Guerra D, Kerster G, Claireaux M, Grobben M, Schriek AI, Boyd A, van Rijswijk J, Tejjani K, Eggink D, Beaumont T, de Taeye SW, de Bree GJ, Sanders RW, van Gils MJ. Primary SARS-CoV-2 variant of concern infections elicit broad antibody Fc-mediated effector functions and memory B cell responses. PLoS Pathog 2024; 20:e1012453. [PMID: 39146376 PMCID: PMC11349224 DOI: 10.1371/journal.ppat.1012453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 08/27/2024] [Accepted: 07/26/2024] [Indexed: 08/17/2024] Open
Abstract
Neutralization of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) by human sera is a strong correlate of protection against symptomatic and severe Coronavirus Disease 2019 (COVID-19). The emergence of antigenically distinct SARS-CoV-2 variants of concern (VOCs) and the relatively rapid waning of serum antibody titers, however, raises questions about the sustainability of serum protection. In addition to serum neutralization, other antibody functionalities and the memory B cell (MBC) response are suggested to help maintaining this protection. In this study, we investigate the breadth of spike (S) protein-specific serum antibodies that mediate effector functions by interacting with Fc-gamma receptor IIa (FcγRIIa) and FcγRIIIa, and of the receptor binding domain (RBD)-specific MBCs, following a primary SARS-CoV-2 infection with the D614G, Alpha, Beta, Gamma, Delta, Omicron BA.1 or BA.2 variant. Irrespectively of the variant causing the infection, the breadth of S protein-specific serum antibodies that interact with FcγRIIa and FcγRIIIa and the RBD-specific MBC responses exceeded the breadth of serum neutralization, although the Alpha-induced B cell response seemed more strain-specific. Between VOC groups, both quantitative and qualitative differences in the immune responses were observed, suggesting differences in immunogenicity. Overall, this study contributes to the understanding of protective humoral and B cell responses in the light of emerging antigenically distinct VOCs, and highlights the need to study the immune system beyond serum neutralization to gain a better understanding of the protection against emerging variants.
Collapse
Affiliation(s)
- Karlijn van der Straten
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Denise Guerra
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Gius Kerster
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Mathieu Claireaux
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Marloes Grobben
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Angela I. Schriek
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Anders Boyd
- Department of Infectious Diseases, Public Health Service of Amsterdam, Amsterdam, the Netherlands
- Stichting HIV monitoring, Amsterdam, the Netherlands
| | - Jacqueline van Rijswijk
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Khadija Tejjani
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Dirk Eggink
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, The Netherlands
| | - Tim Beaumont
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Steven W. de Taeye
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| | - Godelieve J. de Bree
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Amsterdam UMC, location Academic Medical Center, Department of Internal Medicine, Amsterdam, The Netherlands
| | - Rogier W. Sanders
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
- Department of Microbiology and Immunology, Weill Medical College of Cornell University, New York, New York, United States of America
| | - Marit J. van Gils
- Amsterdam UMC, location Academic Medical Center, Department of Medical Microbiology and Infection Prevention, Laboratory of Experimental Virology, Amsterdam, The Netherlands
- Amsterdam Institute for Immunology and Infectious Diseases, Amsterdam, The Netherlands
| |
Collapse
|
5
|
Rival C, Mandal M, Cramton K, Qiao H, Arish M, Sun J, McCann JV, Dudley AC, Solga MD, Erdbrügger U, Erickson LD. B cells secrete functional antigen-specific IgG antibodies on extracellular vesicles. Sci Rep 2024; 14:16970. [PMID: 39043800 PMCID: PMC11266516 DOI: 10.1038/s41598-024-67912-y] [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: 01/24/2024] [Accepted: 07/17/2024] [Indexed: 07/25/2024] Open
Abstract
B cells and the antibodies they produce are critical in host defense against pathogens and contribute to various immune-mediated diseases. B cells responding to activating signals in vitro release extracellular vesicles (EV) that carry surface antibodies, yet B cell production of EVs that express antibodies and their function in vivo is incompletely understood. Using transgenic mice expressing the Cre recombinase in B cells switching to IgG1 to induce expression of fusion proteins between emerald green fluorescent protein (emGFP) and the EV tetraspanin CD63 as a model, we identify emGFP expression in B cells responding to foreign antigen in vivo and characterize the emGFP+ EVs they release. Our data suggests that emGFP+ germinal center B cells undergoing immunoglobulin class switching to express IgG and their progeny memory B cells and plasma cells, also emGFP+, are sources of circulating antigen-specific IgG+ EVs. Furthermore, using a mouse model of influenza virus infection, we find that IgG+ EVs specific for the influenza hemagglutinin antigen protect against virus infection. In addition, crossing the B cell Cre driver EV reporter mice onto the Nba2 lupus-prone strain revealed increased circulating emGFP+ EVs that expressed surface IgG against nuclear antigens linked to autoimmunity. These data identify EVs loaded with antibodies as a novel route for antibody secretion in B cells that contribute to adaptive immune responses, with important implications for different functions of IgG+ EVs in infection and autoimmunity.
Collapse
Affiliation(s)
- Claudia Rival
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mahua Mandal
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Kayla Cramton
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Hui Qiao
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mohd Arish
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Jie Sun
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA
- Division of Infectious Diseases and International Health, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - James V McCann
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Andrew C Dudley
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA
- Emily Couric Cancer Center, University of Virginia, Charlottesville, VA, 22908, USA
| | - Michael D Solga
- Flow Cytometry Core, University of Virginia, Charlottesville, VA, 22908, USA
| | - Uta Erdbrügger
- Division of Nephrology, Department of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Loren D Erickson
- Beirne Carter Center for Immunology Research, University of Virginia, PO Box 801386, Charlottesville, VA, 22908, USA.
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, VA, 22908, USA.
| |
Collapse
|
6
|
Eslami M, Schuepbach-Mallepell S, Diana D, Willen L, Kowalczyk-Quintas C, Desponds C, Peter B, Vigolo M, Renevey F, Donzé O, Luther SA, Yalkinoglu Ö, Alouche N, Schneider P. Unique and redundant roles of mouse BCMA, TACI, BAFF, APRIL, and IL-6 in supporting antibody-producing cells in different tissues. Proc Natl Acad Sci U S A 2024; 121:e2404309121. [PMID: 38990948 PMCID: PMC11260164 DOI: 10.1073/pnas.2404309121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Accepted: 06/03/2024] [Indexed: 07/13/2024] Open
Abstract
Antibody-producing plasma cells fuel humoral immune responses. They also contribute to autoimmune diseases such as systemic lupus erythematosus or IgA nephropathy. Interleukin-6 and the tumor necrosis factor (TNF) family ligands BAFF (B cell-activating factor) and APRIL (a proliferation-inducing ligand) participate in plasma cell survival. BAFF binds to three receptors, BAFFR (BAFF receptor), TACI (transmembrane activator and CAML interactor), and BCMA (B cell maturation antigen), while APRIL binds to TACI, BCMA, and proteoglycans. However, which ligand-receptor pair(s) are required to maintain plasma cells in different body locations remains unknown. Here, by combining mouse genetic and pharmacological approaches, we found that plasma cells required BCMA and/or TACI but not BAFFR. BCMA responded exclusively to APRIL, while TACI responded to both BAFF and APRIL, identifying three self-sufficient ligand-receptor pairs for plasma cell maintenance: BAFF-TACI, APRIL-TACI, and APRIL-BCMA. Together, these actors accounted for 90% of circulating antibodies. In BAFF-ko mice, the reduction of plasma cells upon APRIL inhibition indicated that APRIL could function in the absence of BAFF-APRIL heteromers. No evidence was found that in the absence of BCMA and TACI, binding of APRIL to proteoglycans would help maintain plasma cells. IL-6, alone or together with BAFF and APRIL, supported mainly splenic plasmablasts and plasma cells and contributed to circulating IgG but not IgA levels. In conclusion, survival factors for plasma cells can vary with body location and with the antibody isotype that plasma cells produce. To efficiently target plasma cells, in particular IgA-producing ones, dual inhibition of BAFF and APRIL is required.
Collapse
Affiliation(s)
- Mahya Eslami
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | | | - Daniela Diana
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Laure Willen
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | | | - Chantal Desponds
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Benjamin Peter
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Michele Vigolo
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - François Renevey
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | | | - Sanjiv A. Luther
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Özkan Yalkinoglu
- Clinical Pharmacology, Global Early Development, The Healthcare Business of Merck KGaA, Darmstadt64293, Germany
| | - Nagham Alouche
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| | - Pascal Schneider
- Department of Immunobiology, University of Lausanne, Epalinges1066, Switzerland
| |
Collapse
|
7
|
Park JE, Kim DH. Advanced Immunomodulatory Biomaterials for Therapeutic Applications. Adv Healthc Mater 2024:e2304496. [PMID: 38716543 DOI: 10.1002/adhm.202304496] [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: 12/16/2023] [Revised: 04/15/2024] [Indexed: 05/22/2024]
Abstract
The multifaceted biological defense system modulating complex immune responses against pathogens and foreign materials plays a critical role in tissue homeostasis and disease progression. Recently developed biomaterials that can specifically regulate immune responses, nanoparticles, graphene, and functional hydrogels have contributed to the advancement of tissue engineering as well as disease treatment. The interaction between innate and adaptive immunity, collectively determining immune responses, can be regulated by mechanobiological recognition and adaptation of immune cells to the extracellular microenvironment. Therefore, applying immunomodulation to tissue regeneration and cancer therapy involves manipulating the properties of biomaterials by tailoring their composition in the context of the immune system. This review provides a comprehensive overview of how the physicochemical attributes of biomaterials determine immune responses, focusing on the physical properties that influence innate and adaptive immunity. This review also underscores the critical aspect of biomaterial-based immune engineering for the development of novel therapeutics and emphasizes the importance of understanding the biomaterials-mediated immunological mechanisms and their role in modulating the immune system.
Collapse
Affiliation(s)
- Ji-Eun Park
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
- Department of Integrative Energy Engineering, College of Engineering, Korea University, Seoul, 02841, Republic of Korea
- Biomedical Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| |
Collapse
|
8
|
Airola C, Andaloro S, Gasbarrini A, Ponziani FR. Vaccine Responses in Patients with Liver Cirrhosis: From the Immune System to the Gut Microbiota. Vaccines (Basel) 2024; 12:349. [PMID: 38675732 PMCID: PMC11054513 DOI: 10.3390/vaccines12040349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 04/28/2024] Open
Abstract
Vaccines prevent a significant number of deaths annually. However, certain populations do not respond adequately to vaccination due to impaired immune systems. Cirrhosis, a condition marked by a profound disruption of immunity, impairs the normal immunization process. Critical vaccines for cirrhotic patients, such as the hepatitis A virus (HAV), hepatitis B virus (HBV), influenza, pneumococcal, and coronavirus disease 19 (COVID-19), often elicit suboptimal responses in these individuals. The humoral response, essential for immunization, is less effective in cirrhosis due to a decline in B memory cells and an increase in plasma blasts, which interfere with the creation of a long-lasting response to antigen vaccination. Additionally, some T cell subtypes exhibit reduced activation in cirrhosis. Nonetheless, the persistence of memory T cell activity, while not preventing infections, may help to attenuate the severity of diseases in these patients. Alongside that, the impairment of innate immunity, particularly in dendritic cells (DCs), prevents the normal priming of adaptive immunity, interrupting the immunization process at its onset. Furthermore, cirrhosis disrupts the gut-liver axis balance, causing dysbiosis, reduced production of short-chain fatty acids (SCFAs), increased intestinal permeability, and bacterial translocation. Undermining the physiological activity of the immune system, these alterations could impact the vaccine response. Enhancing the understanding of the molecular and cellular factors contributing to impaired vaccination responses in cirrhotic patients is crucial for improving vaccine efficacy in this population and developing better prevention strategies.
Collapse
Affiliation(s)
- Carlo Airola
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
| | - Silvia Andaloro
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
| | - Antonio Gasbarrini
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| | - Francesca Romana Ponziani
- Liver Unit, CEMAD Centro Malattie dell’Apparato Digerente, Medicina Interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (C.A.); (S.A.); (A.G.)
- Department of Translational Medicine and Surgery, Catholic University, 00168 Rome, Italy
| |
Collapse
|
9
|
Ahmed N, Athavale A, Tripathi AH, Subramaniam A, Upadhyay SK, Pandey AK, Rai RC, Awasthi A. To be remembered: B cell memory response against SARS-CoV-2 and its variants in vaccinated and unvaccinated individuals. Scand J Immunol 2024; 99:e13345. [PMID: 38441373 DOI: 10.1111/sji.13345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 10/20/2023] [Accepted: 11/13/2023] [Indexed: 03/07/2024]
Abstract
COVID-19 disease has plagued the world economy and affected the overall well-being and life of most of the people. Natural infection as well as vaccination leads to the development of an immune response against the pathogen. This involves the production of antibodies, which can neutralize the virus during future challenges. In addition, the development of cellular immune memory with memory B and T cells provides long-lasting protection. The longevity of the immune response has been a subject of intensive research in this field. The extent of immunity conferred by different forms of vaccination or natural infections remained debatable for long. Hence, understanding the effectiveness of these responses among different groups of people can assist government organizations in making informed policy decisions. In this article, based on the publicly available data, we have reviewed the memory response generated by some of the vaccines against SARS-CoV-2 and its variants, particularly B cell memory in different groups of individuals.
Collapse
Affiliation(s)
- Nafees Ahmed
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Atharv Athavale
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Ankita H Tripathi
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | - Adarsh Subramaniam
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Santosh K Upadhyay
- Department of Biotechnology, Kumaun University, Nainital, Uttarakhand, India
| | | | - Ramesh Chandra Rai
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| | - Amit Awasthi
- Translational Health Science and Technology Institute, Faridabad, Haryana, India
| |
Collapse
|
10
|
Wu CY, Tseng YC, Kao SE, Wu LY, Hou JT, Yang YC, Hsiao PW, Chen JR. Monoglycosylated SARS-CoV-2 receptor binding domain fused with HA stem-scaffolded protein vaccine confers broad protective immunity against SARS-CoV-2 and influenza viruses. Antiviral Res 2023; 220:105759. [PMID: 37984568 DOI: 10.1016/j.antiviral.2023.105759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 11/13/2023] [Accepted: 11/15/2023] [Indexed: 11/22/2023]
Abstract
The SARS-CoV-2 and influenza pandemics have posed a devastating threat to global public health. The best strategy for preventing the further spread of these respiratory viruses worldwide is to administer a vaccine capable of targeting both viruses. Here, we show that a novel monoglycosylated vaccine designed based on the influenza virus HAstem conserved domain fused with the SARS-CoV-2 spike-RBD domain (HSSRmg) can present proper antigenicity that elicits sufficient neutralization efficacy against various SARS-CoV-2 variants while simultaneously providing broad protection against H1N1 viruses in mice. Compared with the fully glycosylated HSSR (HSSRfg), HSSRmg induced higher ELISA titers targeting HAstem and spike-RBD and exhibited significantly enhanced neutralization activity against the Wuhan pseudovirus. The enhanced immune responses raised by JR300-adjuvanted HSSRmg compared to HSSRmg alone include more anti-HAstem and anti-spike-RBD antibodies that provide cross-protection against H1N1 challenges and cross-neutralization of SARS-CoV-2 pseudoviruses. Furthermore, the enhanced immune response raised by JR300-adjuvanted-HSSRmg skews toward a more balanced Th1/Th2 response than that raised by HSSRmg alone. Notably, HSSRmg elicited more plasma B cells and memory B cells, and higher IL-4 and IFN-γ cytokine immune responses than spike (S-2P) in mice with preexisting influenza-specific immunity, suggesting that B-cell activation most likely occurs through CD4+ T-cell stimulation. This study demonstrated that HSSRmg produced using a monoglycosylation process and combined with the JR300 adjuvant elicits superior cross-strain immune responses against SARS-CoV-2 and influenza viruses in mice compared with S-2P. JR300-adjuvanted HSSRmg has great potential as a coronavirus-influenza vaccine that provides dual protection against SARS-CoV-2 and influenza infections.
Collapse
Affiliation(s)
| | | | - Shao-En Kao
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Li-Yang Wu
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Jen-Tzu Hou
- RuenHuei Biopharmaceuticals Inc. Taipei, Taiwan
| | - Yu-Chih Yang
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | - Pei-Wen Hsiao
- Agricultural Biotechnology Research Center, Academia Sinica, Taipei, Taiwan
| | | |
Collapse
|
11
|
Toapanta FR, Hu J, Meron-Sudai S, Mulard LA, Phalipon A, Cohen D, Sztein MB. Further characterization of Shigella-specific (memory) B cells induced in healthy volunteer recipients of SF2a-TT15, a Shigella flexneri 2a synthetic glycan-based vaccine candidate. Front Immunol 2023; 14:1291664. [PMID: 38022674 PMCID: PMC10653583 DOI: 10.3389/fimmu.2023.1291664] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 10/18/2023] [Indexed: 12/01/2023] Open
Abstract
Shigellosis is common worldwide, and it causes significant morbidity and mortality mainly in young children in low- and middle- income countries. To date, there are not broadly available licensed Shigella vaccines. A novel type of conjugate vaccine candidate, SF2a-TT15, was developed against S. flexneri serotype 2a (SF2a). SF2a-TT15 is composed of a synthetic 15mer oligosaccharide, designed to act as a functional mimic of the SF2a O-antigen and covalently linked to tetanus toxoid (TT). SF2a-TT15 was recently shown to be safe and immunogenic in a Phase 1 clinical trial, inducing specific memory B cells and sustained antibody response up to three years after the last injection. In this manuscript, we advance the study of B cell responses to parenteral administration of SF2a-TT15 to identify SF2a LPS-specific B cells (SF2a+ B cells) using fluorescently labeled bacteria. SF2a+ B cells were identified mainly within class-switched B cells (SwB cells) in volunteers vaccinated with SF2a-TT15 adjuvanted or not with aluminium hydroxide (alum), but not in placebo recipients. These cells expressed high levels of CXCR3 and low levels of CD21 suggesting an activated phenotype likely to represent the recently described effector memory B cells. IgG SF2a+ SwB cells were more abundant than IgA SF2a + SwB cells. SF2a+ B cells were also identified in polyclonally stimulated B cells (antibody secreting cells (ASC)-transformed). SF2a+ ASC-SwB cells largely maintained the activated phenotype (CXCR3 high, CD21 low). They expressed high levels of CD71 and integrin α4β7, suggesting a high proliferation rate and ability to migrate to gut associated lymphoid tissues. Finally, ELISpot analysis showed that ASC produced anti-SF2a LPS IgG and IgA antibodies. In summary, this methodology confirms the ability of SF2a-TT15 to induce long-lived memory B cells, initially identified by ELISpots, which remain identifiable in blood up to 140 days following vaccination. Our findings expand and complement the memory B cell data previously reported in the Phase 1 trial and provide detailed information on the immunophenotypic characteristics of these cells. Moreover, this methodology opens the door to future studies at the single-cell level to better characterize the development of B cell immunity to Shigella.
Collapse
Affiliation(s)
- Franklin R. Toapanta
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Jingping Hu
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Shiri Meron-Sudai
- School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Laurence A. Mulard
- Institut Pasteur, Université Paris Cité, CNRS UMR3523, Unité Chimie des Biomolécules, Paris, France
| | - Armelle Phalipon
- Institut Pasteur, Université Paris Cité, Laboratoire Innovation: Vaccins, Paris, France
| | - Dani Cohen
- School of Public Health, Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Marcelo B. Sztein
- Department of Medicine and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
- Department of Pediatrics and Center for Vaccine Development and Global Health, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
12
|
Doherty J, O'Morain N, Stack R, Tosetto M, Inzitiari R, O'Reilly S, Gu L, Sheridan J, Cullen G, Mc Dermott E, Buckley M, Horgan G, Mulcahy H, Walshe M, Ryan EJ, Gautier V, Prostko J, Frias E, Daghfal D, Doran P, O'Morain C, Doherty GA. Reduced Serological Response to COVID-19 Booster Vaccine is Associated with Reduced B Cell Memory in Patients With Inflammatory Bowel Disease; VARIATION [VAriability in Response in IBD AgainsT SARS-COV-2 ImmunisatiON]. J Crohns Colitis 2023; 17:1445-1456. [PMID: 37018462 DOI: 10.1093/ecco-jcc/jjad065] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Indexed: 04/07/2023]
Abstract
BACKGROUND AND AIMS Patients with inflammatory bowel disease [IBD] have an attenuated response to initial COVID-19 vaccination. We sought to characterize the impact of IBD and its treatment on responses after the third vaccine against SARS-CoV-2. METHODS This was a prospective multicentre observational study of patients with IBD [n = 202] and healthy controls [HC, n = 92]. Serological response to vaccination was assessed by quantification of anti-spike protein [SP] immunoglobulin [Ig]G levels [anti-SPIgG] and in vitro neutralization of binding to angiotensin-converting enzyme 2 [ACE2]. Peripheral blood B-cell phenotype populations were assessed by flow cytometry. SARS-CoV-2 antigen-specific B-cell responses were assessed in ex vivo culture. RESULTS Median anti-SP IgG post-third vaccination in our IBD cohort was significantly lower than HCs [7862 vs 19 622 AU/mL, p < 0.001] as was ACE2 binding inhibition [p < 0.001]. IBD patients previously infected with COVID-19 [30%] had similar quantitative antibody response as HCs previously infected with COVID-19 [p = 0.12]. Lowest anti-SP IgG titres and neutralization were seen in IBD patients on anti-tumour necrosis factor [anti-TNF] agents, without prior COVID-19 infection, but all IBD patients show an attenuated vaccine response compared to HCs. Patients with IBD have reduced memory B-cell populations and attenuated B-cell responses to SARS-CoV-2 antigens if not previously infected with COVID-19 [p = 0.01]. Higher anti-TNF drug levels and zinc levels <65 ng/ml were associated with significantly lower serological responses. CONCLUSIONS Patients with IBD have an attenuated response to three doses of SARS-CoV-2 vaccine. Physicians should consider patients with higher anti-TNF drug levels and/or zinc deficiency as potentially at higher risk of attenuated response to vaccination.
Collapse
Affiliation(s)
- Jayne Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Neil O'Morain
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Roisin Stack
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Miriam Tosetto
- School of Medicine, University College Dublin, Dublin, Ireland
| | | | - Sophie O'Reilly
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - Lili Gu
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - Juliette Sheridan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Garret Cullen
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Edel Mc Dermott
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
| | - Maire Buckley
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- St Michaels Hospital, Dun Laoghaire, Co Dublin, Ireland
| | - Gareth Horgan
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- St Columcille's Hospital, Loughlinstown, Co Dublin, Ireland
| | - Hugh Mulcahy
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Margaret Walshe
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Elizabeth J Ryan
- Department of Biological Sciences, Health Research Institute, University of Limerick, Limerick, Ireland
| | - Virginie Gautier
- Centre for Experimental Pathogen Host Research, School of Medicine, University College Dublin, Ireland
| | - John Prostko
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - Edwin Frias
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - David Daghfal
- Abbott Laboratories, Abbott Diagnostics, Lake Forest, IL 60045, USA
| | - Peter Doran
- School of Medicine, University College Dublin, Dublin, Ireland
| | - Colm O'Morain
- Beacon Hospital, Sandyford, Co. Dublin and Trinity College Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| | - Glen A Doherty
- Centre for Colorectal Disease, St Vincent's University Hospital, Dublin, Ireland
- School of Medicine, University College Dublin, Dublin, Ireland
- INITIative IBD Research Network (www.initiativeibd.ie)
| |
Collapse
|
13
|
Xipell M, Lledó GM, Egan AC, Tamirou F, Del Castillo CS, Rovira J, Gómez-Puerta JA, García-Herrera A, Cervera R, Kronbichler A, Jayne DRW, Anders HJ, Houssiau F, Espinosa G, Quintana LF. From systemic lupus erythematosus to lupus nephritis: The evolving road to targeted therapies. Autoimmun Rev 2023; 22:103404. [PMID: 37543287 DOI: 10.1016/j.autrev.2023.103404] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 08/02/2023] [Indexed: 08/07/2023]
Abstract
Systemic lupus erythematosus is a chronic autoimmune disease characterized by loss of tolerance against nuclear and cytoplasmic self-antigens, induction of immunity and tissue inflammation. Lupus nephritis (LN), the most important predictor of morbidity in SLE, develops in almost 30% of SLE patients at disease onset and in up to 50-60% within the first 10 years. Firstly, in this review, we put the pathogenic mechanisms of the disease into a conceptual frame, giving emphasis to the role of the innate immune system in this loss of self-tolerance and the induction of the adaptive immune response. In this aspect, many mechanisms have been described such as dysregulation and acceleration of cell-death pathways, an aberrant clearance and overload of immunogenic acid-nucleic-containing debris and IC, and the involvement of antigen-presenting cells and other innate immune cells in the induction of this adaptive immune response. This result in a clonal expansion of autoreactive lymphocytes with generation of effector T-cells, memory B-cells and plasma cells that produce autoantibodies that will cause kidney damage. Secondly, we review the immunological pathways of damage in the kidney parenchyma, initiated by autoantibody binding and immune complex deposition, and followed by complement-mediated microvascular injury, activation of kidney stromal cells and the recruitment of leukocytes. Finally, we summarize the rationale for the treatment of LN, from conventional to new targeted therapies, focusing on their systemic immunologic effects and the minimization of podocytary damage.
Collapse
Affiliation(s)
- Marc Xipell
- Department of Nephrology and Renal Transplantation, Clinic Barcelona, Spain; Reference Center for Complex Glomerular Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Gema M Lledó
- Department of Autoimmune Diseases, Clínic Barcelona, Spain; Reference Center for Systemic Autoimmune Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Allyson C Egan
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, United Kingdom
| | - Farah Tamirou
- Rheumatology Department, Cliniques Universitaires Saint-Luc, Bruxelles, Belgium; Pôle de Pathologies Rhumatismales Inflammatoires et Systémiques, Institut de Recherche Expérimentale et Clinique, Université catholique de Louvain, Belgium
| | | | - Jordi Rovira
- Laboratori Experimental de Nefrologia i Trasplantament (LENIT), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - José A Gómez-Puerta
- Department of Rheumatology, Clínic Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Catalonia, Spain
| | - Adriana García-Herrera
- Department of Pathology, Clínic Barcelona, Spain; Reference Center for Complex Glomerular Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain
| | - Ricard Cervera
- Department of Autoimmune Diseases, Clínic Barcelona, Spain
| | - Andreas Kronbichler
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - David R W Jayne
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom
| | - Hans-Joachim Anders
- Division of Nephrology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Frédéric Houssiau
- Vasculitis and Lupus Service, Addenbrooke's Hospital, Cambridge, United Kingdom; Department of Medicine, University of Cambridge, United Kingdom
| | - Gerard Espinosa
- Department of Autoimmune Diseases, Clínic Barcelona, Spain; Reference Center for Systemic Autoimmune Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain.
| | - Luis F Quintana
- Department of Nephrology and Renal Transplantation, Clinic Barcelona, Spain; Reference Center for Complex Glomerular Diseases of the Spanish Health System (CSUR), Department of Medicine, University of Barcelona, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain.
| |
Collapse
|
14
|
Schulz AR, Fiebig L, Hirseland H, Diekmann LM, Reinke S, Hardt S, Niedobitek A, Mei HE. SARS-CoV-2 specific plasma cells acquire long-lived phenotypes in human bone marrow. EBioMedicine 2023; 95:104735. [PMID: 37556944 PMCID: PMC10432952 DOI: 10.1016/j.ebiom.2023.104735] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 07/08/2023] [Accepted: 07/15/2023] [Indexed: 08/11/2023] Open
Abstract
BACKGROUND SARS-CoV-2 specific antibody-secreting plasma cells (PC) mediating specific humoral immunity have been identified in the human bone marrow (BM) after COVID-19 or vaccination against SARS-CoV-2. However, it remained unclear whether or not they acquire phenotypes of human memory plasma cells. METHODS SARS-CoV-2-specific human bone marrow plasma cells (BMPC) were characterised by tetramer-based, antigen-specific flow cytometry and FluoroSpot assay. FINDINGS SARS-CoV-2 spike-S1-specific PC were detectable in all tested BM samples of previously vaccinated individuals, representing 0.22% of total BMPC. The majority of SARS-CoV-2-specific BMPC expressed IgG and their specificity for the spike S1 protein indicated emergence from a systemic vaccination response. Of note, one-fifth of SARS-CoV-2-specific BMPC showed the phenotype of memory plasma cells, i.e., downregulated CD19 and present or absent CD45 expression. INTERPRETATION Our data indicate the establishment of phenotypically diverse SARS-CoV-2-specific PC in the human BM after basic mRNA immunization, including the formation of memory phenotypes. These results suggest the induction of durable humoral immunity after basic mRNA vaccination against SARS-CoV-2. FUNDING The study was supported by funding by the DFG grants TRR130 TP24, ME 3644/8-1, and the Berlin Senate. SR received funding from DFGSFB-1444 C01 Central Service Project.
Collapse
Affiliation(s)
- Axel R Schulz
- Deutsches Rheuma-Forschungszentrum Berlin, A Leibniz Institute, Berlin, Germany
| | - Leonard Fiebig
- Deutsches Rheuma-Forschungszentrum Berlin, A Leibniz Institute, Berlin, Germany
| | - Heike Hirseland
- Deutsches Rheuma-Forschungszentrum Berlin, A Leibniz Institute, Berlin, Germany
| | - Lisa-Marie Diekmann
- Deutsches Rheuma-Forschungszentrum Berlin, A Leibniz Institute, Berlin, Germany
| | - Simon Reinke
- Cell Harvesting Core, Berlin Institute of Health, Berlin, Germany
| | - Sebastian Hardt
- Center for Musculoskeletal Surgery, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Antonia Niedobitek
- Deutsches Rheuma-Forschungszentrum Berlin, A Leibniz Institute, Berlin, Germany
| | - Henrik E Mei
- Deutsches Rheuma-Forschungszentrum Berlin, A Leibniz Institute, Berlin, Germany.
| |
Collapse
|
15
|
Abstract
Bone marrow is known as the site of hematopoiesis. What is not being described in textbooks of immunology is the fact that bone marrow is not only a generative, but also an antigen-responsive, immune organ. It is also a major storage site for antigen-specific memory B and T cells. That bone marrow is a priming site for T cell responses to blood borne antigens was discovered exactly 20 years ago. This review celebrates this important discovery. The review provides a number of examples of medical relevance of bone marrow as a central immune system, including cancer, microbial infections, autoimmune reactions, and bone marrow transplantation. Bone marrow mesenchymal stem cell-derived stromal cells provide distinct bone marrow niches for stem cells and immune cells. By transmitting anti-inflammatory dampening effects, facilitating wound healing and tissue regeneration mesenchymal stem cells contribute to homeostasis of bone and other tissues. Based on the evidence presented, the review proposes that bone marrow is a multifunctional and protective immune system. In an analogy to the central nervous system, it is suggested that bone marrow be designated as the central immune system.
Collapse
|
16
|
Wang F, Liang J, Zhu D, Xiang P, Zhou L, Yang C. Characteristic gene prognostic model of type 1 diabetes mellitus via machine learning strategy. Endocr J 2023; 70:281-294. [PMID: 36477008 DOI: 10.1507/endocrj.ej22-0178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The present study was designed to detect possible biomarkers associated with Type 1 diabetes mellitus (T1DM) incidence in an effort to develop novel treatments for this condition. Three mRNA expression datasets of peripheral blood mononuclear cells (PBMCs) were obtained from the GEO database. Differentially expressed genes (DEGs) between T1DM patients and healthy controls were identified by Limma package in R, and using the DEGs to conduct GO and DO pathway enrichment. The LASSO-SVM were used to screen the hub genes. We performed immune correlation analysis of hub genes and established a T1DM prognosis model. CIBERSORT algorithm was used to identify the different immune cells in distribution between T1DM and normal samples. The correlation of the hub genes and immune cells was analyzed by Spearman. ROC curves were used to assess the diagnostic value of genes in T1DM. A total of 60 immune related DEGs were obtained from the T1DM and normal samples. Then, DEGs were further screened to obtain 3 hub genes, ANP32A-IT1, ESCO2 and NBPF1. CIBERSORT analysis revealed the percentage of immune cells in each sample, indicating that there was significant difference in monocytes, T cells CD8+, gamma delta T cells, naive CD4+ T cells and activated memory CD4+ T cells between T1DM and normal samples. The area under curve (AUC) of ESCO2, ANP32A-IT1 and NBPF1 were all greater than 0.8, indicating that these three genes have high diagnostic value for T1DM. Together, the findings of these bioinformatics analyses thus identified key hub genes associated with T1DM development.
Collapse
Affiliation(s)
- Fenglin Wang
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
- Hebei North University, Zhangjiakou 075000, China
| | - Jiemei Liang
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
- Hebei North University, Zhangjiakou 075000, China
| | - Di Zhu
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
| | - Pengan Xiang
- Hospital of 94498 Troops, People's Liberation Army, Nanyang 474300, China
| | - Luyao Zhou
- Hebei North University, Zhangjiakou 075000, China
| | - Caizhe Yang
- Department of Endocrinology of the Air Force Medical Center, People's Liberation Army, Beijing 100142, China
| |
Collapse
|
17
|
Sollid LM, Iversen R. Tango of B cells with T cells in the making of secretory antibodies to gut bacteria. Nat Rev Gastroenterol Hepatol 2023; 20:120-128. [PMID: 36056203 DOI: 10.1038/s41575-022-00674-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 02/03/2023]
Abstract
Polymeric IgA and IgM are transported across the epithelial barrier from plasma cells in the lamina propria to exert a function in the gut lumen as secretory antibodies. Many secretory antibodies are reactive with the gut bacteria, and mounting evidence suggests that these antibodies are important for the host to control gut bacterial communities. However, we have incomplete knowledge of how bacteria-reactive secretory antibodies are formed. Antibodies from gut plasma cells often show bacterial cross-species reactivity, putting the degree of specificity behind anti-bacterial antibody responses into question. Such cross-species reactive antibodies frequently recognize non-genome-encoded membrane glycan structures. On the other hand, the T cell epitopes are peptides encoded in the bacterial genomes, thereby allowing a higher degree of predictable specificity on the T cell side of anti-bacterial immune responses. In this Perspective, we argue that the production of bacteria-reactive secretory antibodies is mainly controlled by the antigen specificity of T cells, which provide help to B cells. To be able to harness this system (for instance, for manipulation with vaccines), we need to obtain insight into the bacterial epitopes recognized by T cells in addition to characterizing the reactivity of the antibodies.
Collapse
Affiliation(s)
- Ludvig M Sollid
- K.G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| | - Rasmus Iversen
- K.G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway. .,Department of Immunology, Oslo University Hospital - Rikshospitalet, Oslo, Norway.
| |
Collapse
|
18
|
Assessing the impact of hepatitis B immune globulin (HBIG) on responses to hepatitis B vaccine during co-administration. Vaccine 2023; 41:955-964. [PMID: 36586740 DOI: 10.1016/j.vaccine.2022.12.055] [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: 06/07/2022] [Revised: 12/20/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022]
Abstract
INTRODUCTION A hepatitis B vaccination (HepB) series with an initial dose of hepatitis B immune globulin (HBIG) is the recommended prophylaxis for infants born to mothers with chronic hepatitis B virus (HBV) infection and for HBV-exposed persons without known protection. The HepB and HBIG are administered at different sites (limbs). Instances of HepB and HBIG administered at the same site are documented but the impact on immune responses to HepB remains unanswered. METHODS Newborn and adult BALB/c mice received one dose of HepB at time zero alone or with HBIG in the same or different sites, followed by 2 additional doses of HepB at 3 and 10 weeks (newborn mice) or 4 and 16 weeks (adult mice). To study memory responses mice were given a 4th, booster, dose of HepB at 26 weeks and B cells analyzed. RESULTS Administration of HepB with HBIG resulted in reduced responses to HepB following the first 2 doses, regardless of site, compared to mice that received HepB only. Lower levels of antibody to HBV surface antigen (anti-HBs) were observed at the end of the 3-dose series (p < 0.0001) in all groups of newborn mice that received HepB and HBIG. In adult mice, this difference was only seen when HepB and HBIG were delivered at the same site. However, following a HepB booster at 26 weeks, HBsAg-specific B-cell expansion and memory phenotype were not impacted by initial HBIG administration CONCLUSION: Administration of HBIG with HepB can delay and reduce responses to HepB in mice. Our findings suggest that the initial circulating levels of HBIG could prevent infection despite an impaired response to vaccine and support the current recommendation of assessing seroprotection after series completion for infants born to HBV carrier mothers, including in cases where vaccine and HBIG are administered incorrectly at the same site.
Collapse
|
19
|
Besbassi H, Garcia-Fogeda I, Quinlivan M, Breuer J, Abrams S, Hens N, Ogunjimi B, Beutels P. Modeling antibody dynamics following herpes zoster indicates that higher varicella-zoster virus viremia generates more VZV-specific antibodies. Front Immunol 2023; 14:1104605. [PMID: 36875105 PMCID: PMC9978810 DOI: 10.3389/fimmu.2023.1104605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Accepted: 01/27/2023] [Indexed: 02/18/2023] Open
Abstract
Introduction Studying antibody dynamics following re-exposure to infection and/or vaccination is crucial for a better understanding of fundamental immunological processes, vaccine development, and health policy research. Methods We adopted a nonlinear mixed modeling approach based on ordinary differential equations (ODE) to characterize varicella-zoster virus specific antibody dynamics during and after clinical herpes zoster. Our ODEs models convert underlying immunological processes into mathematical formulations, allowing for testable data analysis. In order to cope with inter- and intra-individual variability, mixed models include population-averaged parameters (fixed effects) and individual-specific parameters (random effects). We explored the use of various ODE-based nonlinear mixed models to describe longitudinally collected markers of immunological response in 61 herpes zoster patients. Results Starting from a general formulation of such models, we study different plausible processes underlying observed antibody titer concentrations over time, including various individual-specific parameters. Among the converged models, the best fitting and most parsimonious model implies that once Varicella-zoster virus (VZV) reactivation is clinically apparent (i.e., Herpes-zoster (HZ) can be diagnosed), short-living and long-living antibody secreting cells (SASC and LASC, respectively) will not expand anymore. Additionally, we investigated the relationship between age and viral load on SASC using a covariate model to gain a deeper understanding of the population's characteristics. Conclusion The results of this study provide crucial and unique insights that can aid in improving our understanding of VZV antibody dynamics and in making more accurate projections regarding the potential impact of vaccines.
Collapse
Affiliation(s)
- Hajar Besbassi
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium.,Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Irene Garcia-Fogeda
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| | - Mark Quinlivan
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Judy Breuer
- Division of Infection and Immunity, University College London, London, United Kingdom
| | - Steven Abrams
- Global Health Institute (GHI), Family Medicine and Population Health (FAMPOP), University of Antwerp, Antwerp, Belgium.,Data Science Institute (DSI), Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), UHasselt, Hasselt, Belgium
| | - Niel Hens
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Data Science Institute (DSI), Interuniversity Institute for Biostatistics and Statistical Bioinformatics (I-BioStat), UHasselt, Hasselt, Belgium
| | - Benson Ogunjimi
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Antwerp Unit for Data Analysis and Computation in Immunology and Sequencing (AUDACIS), University of Antwerp, Antwerp, Belgium.,Antwerp Center for Translational Immunology and Virology (ACTIV), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium.,Department of Paediatrics, Antwerp University Hospital, Edegem, Belgium
| | - Philippe Beutels
- Centre for Health Economics Research and Modeling Infectious Diseases (CHERMID), Vaccine and Infectious Disease Institute (VAXINFECTIO), University of Antwerp, Antwerp, Belgium
| |
Collapse
|
20
|
Pan G, Zhang P, Yang J, Wu Y. The regulatory effect of specialized pro-resolving mediators on immune cells. Biomed Pharmacother 2022; 156:113980. [DOI: 10.1016/j.biopha.2022.113980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Revised: 10/22/2022] [Accepted: 11/04/2022] [Indexed: 11/08/2022] Open
|
21
|
Garcia-Dominguez D, Henry C, Ma L, Jani H, Amato NJ, Manning T, Freyn A, Davis H, Hsiao CJ, Li M, Koch H, Elbashir S, DiPiazza A, Carfi A, Edwards D, Bahl K. Altering the mRNA-1273 dosing interval impacts the kinetics, quality, and magnitude of immune responses in mice. Front Immunol 2022; 13:948335. [PMID: 36426367 PMCID: PMC9679967 DOI: 10.3389/fimmu.2022.948335] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/13/2022] [Indexed: 12/17/2024] Open
Abstract
For a vaccine to achieve durable immunity and optimal efficacy, many require a multi-dose primary vaccination schedule that acts to first "prime" naive immune systems and then "boost" initial immune responses by repeated immunizations (ie, prime-boost regimens). In the context of the global coronavirus disease 2019 (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), 2-dose primary vaccination regimens were often selected with short intervals between doses to provide rapid protection while still inducing robust immunity. However, emerging post-authorization evidence has suggested that longer intervals between doses 1 and 2 for SARS-CoV-2 vaccines may positively impact robustness and durability of immune responses. Here, the dosing interval for mRNA-1273, a messenger RNA based SARS-CoV-2 vaccine administered on a 2-dose primary schedule with 4 weeks between doses, was evaluated in mice by varying the dose interval between 1 and 8 weeks and examining immune responses through 24 weeks after dose 2. A dosing interval of 6 to 8 weeks generated the highest level of antigen-specific serum immunoglobulin G binding antibody titers. Differences in binding antibody titers between mRNA-1273 1 µg and 10 µg decreased over time for dosing intervals of ≥4 weeks, suggesting a potential dose-sparing effect. Longer intervals (≥4 weeks) also increased antibody-dependent cellular cytotoxicity activity and numbers of antibody-secreting cells (including long-lived plasma cells) after the second dose. An interval of 6 to 8 weeks elicited the strongest CD8+ T-cell responses, while an interval of 3 weeks elicited the strongest CD4+ T-cell response. Overall, these results suggest that in a non-pandemic setting, a longer interval (≥6 weeks) between the doses of the primary series for mRNA-1273 may induce more durable immune responses.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kapil Bahl
- Moderna, Inc., Cambridge, MA, United States
| |
Collapse
|
22
|
Ott JA, Haakenson JK, Kelly AR, Christian C, Criscitiello MF, Smider VV. Evolution of surrogate light chain in tetrapods and the relationship between lengths of CDR H3 and VpreB tails. Front Immunol 2022; 13:1001134. [PMID: 36311706 PMCID: PMC9614664 DOI: 10.3389/fimmu.2022.1001134] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 09/20/2022] [Indexed: 12/05/2022] Open
Abstract
In the mammalian immune system, the surrogate light chain (SLC) shapes the antibody repertoire during B cell development by serving as a checkpoint for production of functional heavy chains (HC). Structural studies indicate that tail regions of VpreB contact and cover the third complementarity-determining region of the HC (CDR H3). However, some species, particularly bovines, have CDR H3 regions that may not be compatible with this HC-SLC interaction model. With immense structural and genetic diversity in antibody repertoires across species, we evaluated the genetic origins and sequence features of surrogate light chain components. We examined tetrapod genomes for evidence of conserved gene synteny to determine the evolutionary origin of VpreB1, VpreB2, and IGLL1, as well as VpreB3 and pre-T cell receptor alpha (PTCRA) genes. We found the genes for the SLC components (VpreB1, VpreB2, and IGLL1) only in eutherian mammals. However, genes for PTCRA occurred in all amniote groups and genes for VpreB3 occurred in all tetrapod groups, and these genes were highly conserved. Additionally, we found evidence of a new VpreB gene in non-mammalian tetrapods that is similar to the VpreB2 gene of eutherian mammals, suggesting VpreB2 may have appeared earlier in tetrapod evolution and may be a precursor to traditional VpreB2 genes in higher vertebrates. Among eutherian mammals, sequence conservation between VpreB1 and VpreB2 was low for all groups except rabbits and rodents, where VpreB2 was nearly identical to VpreB1 and did not share conserved synteny with VpreB2 of other species. VpreB2 of rabbits and rodents likely represents a duplicated variant of VpreB1 and is distinct from the VpreB2 of other mammals. Thus, rabbits and rodents have two variants of VpreB1 (VpreB1-1 and VpreB1-2) but no VpreB2. Sequence analysis of VpreB tail regions indicated differences in sequence content, charge, and length; where repertoire data was available, we observed a significant relationship between VpreB2 tail length and maximum DH length. We posit that SLC components co-evolved with immunoglobulin HC to accommodate the repertoire - particularly CDR H3 length and structure, and perhaps highly unusual HC (like ultralong HC of cattle) may bypass this developmental checkpoint altogether.
Collapse
Affiliation(s)
- Jeannine A. Ott
- Comparative Immunogenetics Lab, Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Jeremy K. Haakenson
- Applied Biomedical Science Institute, San Diego, CA, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| | - Abigail R. Kelly
- Applied Biomedical Science Institute, San Diego, CA, United States
| | - Claire Christian
- Comparative Immunogenetics Lab, Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Michael F. Criscitiello
- Comparative Immunogenetics Lab, Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, United States
| | - Vaughn V. Smider
- Applied Biomedical Science Institute, San Diego, CA, United States
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, United States
| |
Collapse
|
23
|
Ruiz MJ, Siracusano G, Cottignies-Calamarte A, Tudor D, Real F, Zhu A, Pastori C, Capron C, Rosenberg AR, Temperton N, Cantoni D, Liao H, Ternette N, Moine P, Godement M, Geri G, Chiche JD, Annane D, Cramer Bordé E, Lopalco L, Bomsel M. Persistent but dysfunctional mucosal SARS-CoV-2-specific IgA and low lung IL-1β associate with COVID-19 fatal outcome: A cross-sectional analysis. Front Immunol 2022; 13:842468. [PMID: 36248831 PMCID: PMC9560774 DOI: 10.3389/fimmu.2022.842468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Accepted: 07/19/2022] [Indexed: 11/17/2022] Open
Abstract
The role of the mucosal pulmonary antibody response in coronavirus disease 2019 (COVID-19) outcome remains unclear. Here, we found that in bronchoalveolar lavage (BAL) samples from 48 patients with severe COVID-19-infected with the ancestral Wuhan virus, mucosal IgG and IgA specific for S1, receptor-binding domain (RBD), S2, and nucleocapsid protein (NP) emerged in BAL containing viruses early in infection and persist after virus elimination, with more IgA than IgG for all antigens tested. Furthermore, spike-IgA and spike-IgG immune complexes were detected in BAL, especially when the lung virus has been cleared. BAL IgG and IgA recognized the four main RBD variants. BAL neutralizing titers were higher early in COVID-19 when virus replicates in the lung than later in infection after viral clearance. Patients with fatal COVID-19, in contrast to survivors, developed higher levels of mucosal spike-specific IgA than IgG but lost neutralizing activities over time and had reduced IL-1β in the lung. Altogether, mucosal spike and NP-specific IgG and S1-specific IgA persisting after lung severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) clearance and low pulmonary IL-1β correlate with COVID-19 fatal outcome. Thus, mucosal SARS-CoV-2-specific antibodies may have adverse functions in addition to protective neutralization.
Collapse
Affiliation(s)
- Maria Julia Ruiz
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Gabriel Siracusano
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Andréa Cottignies-Calamarte
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Daniela Tudor
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Fernando Real
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Aiwei Zhu
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
| | - Claudia Pastori
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Claude Capron
- AP-HP, Hôpital Ambroise Paré, Service d'Hématologie, Boulogne-Billancourt, France
| | - Arielle R. Rosenberg
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- AP-HP, Hôpital Cochin, Service de Virologie, Paris, France
| | - Nigel Temperton
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Diego Cantoni
- Viral Pseudotype Unit, Medway School of Pharmacy, The Universities of Kent and Greenwich at Medway, Chatham, United Kingdom
| | - Hanqing Liao
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Nicola Ternette
- Centre for Cellular and Molecular Physiology, Nuffield Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Pierre Moine
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Mathieu Godement
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | - Guillaume Geri
- AP-HP, Hôpital Ambroise Paré, Service de Réanimation, Boulogne-Billancourt, France
- Université de Versailles-St Quentin en Yvelines, Versailles, France
| | | | - Djillali Annane
- FHU SEPSIS (Saclay and Paris Seine Nord Endeavour to PerSonalize Interventions for Sepsis), RHU RECORDS (Rapid rEcognition of CORticosteroiD resistant or sensitive Sepsis), Department of Intensive Care, Hôpital Raymond Poincaré (APHP), Laboratory of Infection and Inflammation – U1173, School of Medicine Simone Veil, University Versailles Saint Quentin – University Paris Saclay, INSERM, Garches, France
| | | | - Lucia Lopalco
- Immunobiology of HIV Unit, San Raffaele Scientific Institute, Milan, Italy
| | - Morgane Bomsel
- Mucosal Entry of HIV and Mucosal Immunity, Institut Cochin, Paris-Descartes University, Paris, France
- INSERM U1016, Paris, France
- CNRS UMR8104, Paris, France
- *Correspondence: Morgane Bomsel,
| |
Collapse
|
24
|
Skeletal muscle provides the immunological micro-milieu for specific plasma cells in anti-synthetase syndrome-associated myositis. Acta Neuropathol 2022; 144:353-372. [PMID: 35612662 PMCID: PMC9288384 DOI: 10.1007/s00401-022-02438-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 04/08/2022] [Accepted: 05/13/2022] [Indexed: 11/17/2022]
Abstract
Anti-synthetase syndrome (ASyS)-associated myositis is a major subgroup of the idiopathic inflammatory myopathies (IIM) and is characterized by disease chronicity with musculoskeletal, dermatological and pulmonary manifestations. One of eight autoantibodies against the aminoacyl-transferase RNA synthetases (ARS) is detectable in the serum of affected patients. However, disease-specific therapeutic approaches have not yet been established.To obtain a deeper understanding of the underlying pathogenesis and to identify putative therapeutic targets, we comparatively investigated the most common forms of ASyS associated with anti-PL-7, anti-PL-12 and anti-Jo-1. Our cohort consisted of 80 ASyS patients as well as healthy controls (n = 40), diseased controls (n = 40) and non-diseased controls (n = 20). We detected a reduced extent of necrosis and regeneration in muscle biopsies from PL-12+ patients compared to Jo-1+ patients, while PL-7+ patients had higher capillary dropout in biopsies of skeletal muscle. Aside from these subtle alterations, no significant differences between ASyS subgroups were observed. Interestingly, a tissue-specific subpopulation of CD138+ plasma cells and CXCL12+/CXCL13+CD20+ B cells common to ASyS myositis were identified. These cells were localized in the endomysium associated with alkaline phosphatase+ activated mesenchymal fibroblasts and CD68+MHC-II+CD169+ macrophages. An MHC-I+ and MHC-II+ MxA negative type II interferon-driven milieu of myofiber activation, topographically restricted to the perifascicular area and the adjacent perimysium, as well as perimysial clusters of T follicular helper cells defined an extra-medullary immunological niche for plasma cells and activated B cells. Consistent with this, proteomic analyses of muscle tissues from ASyS patients demonstrated alterations in antigen processing and presentation. In-depth immunological analyses of peripheral blood supported a B-cell/plasma-cell-driven pathology with a shift towards immature B cells, an increase of B-cell-related cytokines and chemokines, and activation of the complement system. We hypothesize that a B-cell-driven pathology with the presence and persistence of a specific subtype of plasma cells in the skeletal muscle is crucially involved in the self-perpetuating chronicity of ASyS myositis. This work provides the conceptual framework for the application of plasma-cell-targeting therapies in ASyS myositis.
Collapse
|
25
|
Neutrophils recruited to immunization sites initiating vaccine-induced antibody responses by locally expressing BAFF. iScience 2022; 25:104453. [PMID: 35874922 PMCID: PMC9301880 DOI: 10.1016/j.isci.2022.104453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/23/2022] [Accepted: 05/17/2022] [Indexed: 11/21/2022] Open
Abstract
Neutrophils played a key role in the innate immune responses. Less is known about whether and how the neutrophils recruited in the immunization sites affecting the vaccine-induced antibody responses. In the process of evaluating the efficacy of an oil-in-water emulsion-formulated vaccine in mice, we found that neutrophils were rapidly and massively recruited to immunization sites but were barely detected in the draining lymph nodes. Interestingly, B cell-activating factor (BAFF) was abundantly expressed in the recruiting neutrophils at a very early stage. The initial neutrophil-derived BAFF firstly brought about the B cell responses in the local part, then subsequently in lymphoid organs. Activated B cells produced more BAFF through TLR9-IRF5 signaling pathway, thereby amplifying the vaccine-induced antibody responses. Suppressing BAFF in the neutrophils could weaken the B cell activation and reduce the antibody production. The data indicate that vaccines endow neutrophils with the potential to orchestrate antibody responses at immunization sites. Neutrophils at immunization sites influencing subsequent immune responses Neutrophil-driven BAFF at immunization sites assisting B cell responses to vaccines Activated B cells produce more BAFF through TLR9-IRF5 signaling pathway BAFF-producing neutrophils orchestrate antibody responses at immunization sites
Collapse
|
26
|
Zhang Y, Garcia-Ibanez L, Ulbricht C, Lok LSC, Pike JA, Mueller-Winkler J, Dennison TW, Ferdinand JR, Burnett CJM, Yam-Puc JC, Zhang L, Alfaro RM, Takahama Y, Ohigashi I, Brown G, Kurosaki T, Tybulewicz VLJ, Rot A, Hauser AE, Clatworthy MR, Toellner KM. Recycling of memory B cells between germinal center and lymph node subcapsular sinus supports affinity maturation to antigenic drift. Nat Commun 2022; 13:2460. [PMID: 35513371 PMCID: PMC9072412 DOI: 10.1038/s41467-022-29978-y] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 03/31/2022] [Indexed: 02/04/2023] Open
Abstract
Infection or vaccination leads to the development of germinal centers (GC) where B cells evolve high affinity antigen receptors, eventually producing antibody-forming plasma cells or memory B cells. Here we follow the migratory pathways of B cells emerging from germinal centers (BEM) and find that many BEM cells migrate into the lymph node subcapsular sinus (SCS) guided by sphingosine-1-phosphate (S1P). From the SCS, BEM cells may exit the lymph node to enter distant tissues, while some BEM cells interact with and take up antigen from SCS macrophages, followed by CCL21-guided return towards the GC. Disruption of local CCL21 gradients inhibits the recycling of BEM cells and results in less efficient adaption to antigenic variation. Our findings thus suggest that the recycling of antigen variant-specific BEM cells and transport of antigen back to GC may support affinity maturation to antigenic drift.
Collapse
Affiliation(s)
- Yang Zhang
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Laura Garcia-Ibanez
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Carolin Ulbricht
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Laurence S C Lok
- University of Cambridge Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Jeremy A Pike
- Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, Birmingham, UK
- Institute of Cardiovascular Sciences, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | | | - Thomas W Dennison
- University of Cambridge Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - John R Ferdinand
- University of Cambridge Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Cameron J M Burnett
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Juan C Yam-Puc
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Lingling Zhang
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- The Francis Crick Institute, London, UK
| | - Raul Maqueda Alfaro
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Department of Cell Biology, Center for Research and Advanced Studies, The National Polytechnic Institute, Cinvestav-IPN, Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Mexico City, Mexico
| | - Yousuke Takahama
- Thymus Biology Section, Experimental Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Izumi Ohigashi
- Division of Experimental Immunology, Institute of Advanced Medical Sciences, University of Tokushima, Tokushima, 770-8503, Japan
| | - Geoffrey Brown
- Department of Cell Biology, Center for Research and Advanced Studies, The National Polytechnic Institute, Cinvestav-IPN, Av. IPN 2508, San Pedro Zacatenco, Gustavo A. Madero, 07360, Mexico City, Mexico
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, 565-0871, Japan
- Laboratory of Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences (IMS), Yokohama, Kanagawa, 230-0045, Japan
| | | | - Antal Rot
- Centre for Microvascular Research, The William Harvey Research Institute, Queen Mary University London, EC1M 6BQ, London, UK
- Centre for Inflammation and Therapeutic Innovation, Queen Mary University London, EC1M 6BQ, London, UK
- Institute for Cardiovascular Prevention, Ludwig-Maximilians University, 80336, Munich, Germany
| | - Anja E Hauser
- Department of Rheumatology and Clinical Immunology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, 10117, Berlin, Germany
- Deutsches Rheuma-Forschungszentrum (DRFZ), a Leibniz Institute, Charitéplatz 1, 10117, Berlin, Germany
| | - Menna R Clatworthy
- University of Cambridge Molecular Immunity Unit, MRC Laboratory of Molecular Biology, Cambridge Biomedical Campus, Cambridge, UK
| | - Kai-Michael Toellner
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK.
| |
Collapse
|
27
|
Villanueva-Hernández S, Adib Razavi M, van Dongen KA, Stadler M, de Luca K, Beyersdorf N, Saalmüller A, Gerner W, Mair KH. Co-Expression of the B-Cell Key Transcription Factors Blimp-1 and IRF4 Identifies Plasma Cells in the Pig. Front Immunol 2022; 13:854257. [PMID: 35464468 PMCID: PMC9024106 DOI: 10.3389/fimmu.2022.854257] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/08/2022] [Indexed: 11/18/2022] Open
Abstract
Antibody-secreting plasma cells (PCs) have remained largely uncharacterized for years in the field of porcine immunology. For an in-depth study of porcine PCs, we identified cross-reactive antibodies against three key transcription factors: PR domain zinc finger protein-1 (Blimp-1), interferon regulatory factor 4 (IRF4), and paired box 5 (Pax5). A distinct Blimp-1+IRF4+ cell population was found in cells isolated from blood, spleen, lymph nodes, bone marrow, and lung of healthy pigs. These cells showed a downregulation of Pax5 compared to other B cells. Within Blimp-1+IRF4+ B cells, IgM-, IgG-, and IgA-expressing cells were identified and immunoglobulin-class distribution was clearly different between the anatomical locations, with IgA+ PCs dominating in lung tissue and IgM+ PCs dominating in the spleen. Expression patterns of Ki-67, MHC-II, CD9, and CD28 were investigated in the different organs. A high expression of Ki-67 was observed in blood, suggesting a plasmablast stage. Blimp-1+IRF4+ cells showed an overall lower expression of MHC-II compared to regular B cells, confirming a progressive loss in B-cell differentiation toward the PC stage. CD28 showed slightly elevated expression levels in Blimp-1+IRF4+ cells in most organs, a phenotype that is also described for PCs in mice and humans. This was not seen for CD9. We further developed a FACS-sorting strategy for live porcine PCs for functional assays. CD3-CD16-CD172a– sorted cells with a CD49dhighFSC-Ahigh phenotype contained Blimp-1+IRF4+ cells and were capable of spontaneous IgG production, thus confirming PC identity. These results reveal fundamental phenotypes of porcine PCs and will facilitate the study of this specific B-cell subset in the future.
Collapse
Affiliation(s)
- Sonia Villanueva-Hernández
- Christian Doppler (CD) Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Mahsa Adib Razavi
- Christian Doppler (CD) Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Katinka A. van Dongen
- Christian Doppler (CD) Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Maria Stadler
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Karelle de Luca
- Laboratory of Veterinary Immunology, Global Innovation, Boehringer Ingelheim Animal Health, Lyon, France
| | - Niklas Beyersdorf
- Institute for Virology and Immunobiology, Julius-Maximilians-University, Würzburg, Germany
| | - Armin Saalmüller
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Wilhelm Gerner
- Christian Doppler (CD) Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - Kerstin H. Mair
- Christian Doppler (CD) Laboratory for Optimized Prediction of Vaccination Success in Pigs, Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Institute of Immunology, Department of Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- *Correspondence: Kerstin H. Mair,
| |
Collapse
|
28
|
Vitiello L, Gatta L, Ilari S, Bonassi S, Cristina M, Ciatti F, Fini M, Proietti S, Russo P, Tomino C, Limongi D. Long Lasting Cellular Immune Response Induced by mRNA Vaccination: Implication for Prevention Strategies. Front Immunol 2022; 13:836495. [PMID: 35359985 PMCID: PMC8961295 DOI: 10.3389/fimmu.2022.836495] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 02/18/2022] [Indexed: 12/14/2022] Open
Abstract
As the COVID19 pandemic continues to spread and vaccinations are administered throughout the world at different rates and with different strategies, understanding the multiple aspects of the immune response to vaccinations is required to define more efficient vaccination strategies. To date, the duration of protection induced by COVID19 vaccines is still matter of debate. To assess whether 2-doses vaccination with BNT162b2 mRNA COVID-19 vaccine was sufficient to induce a persistent specific cellular immune response, we evaluated the presence of SARS-COV2 Spike-specific B and T lymphocytes in 28 healthcare workers 1 and 7 months after completing the vaccination cycle. The results showed that at 7 months after second dose a population of Spike-specific B lymphocytes was still present in 86% of the immunized subjects, with a higher frequency when compared to not-immunized controls (0.38% ± 0.07 vs 0.13% ± 0.03, p<0.001). Similarly, specific CD4+ and CD8+ T lymphocytes, able to respond in vitro to stimulation with Spike derived peptides, were found at 7 months. These results confirm that vaccination with BNT162b2 is able to induce a specific immune response, potentially long lasting, and could be helpful in defining future vaccination strategies.
Collapse
Affiliation(s)
- Laura Vitiello
- Laboratory of Flow Cytometry, IRCCS San Raffaele Roma, Rome, Italy
| | - Lucia Gatta
- Scientific Direction, IRCCS San Raffaele Roma, Rome, Italy
| | - Sara Ilari
- Department of Health Science, Institute of Research for Food Safety and Health (IRC-FSH), University “Magna Graecia” of Catanzaro, Catanzaro, Italy
| | - Stefano Bonassi
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | | | - Filippo Ciatti
- Laboratory of Clinical Pathology, IRCCS San Raffaele Roma, Rome, Italy
| | - Massimo Fini
- Scientific Direction, IRCCS San Raffaele Roma, Rome, Italy
| | - Stefania Proietti
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Rome, Italy
| | - Patrizia Russo
- Clinical and Molecular Epidemiology, IRCCS San Raffaele Roma, Rome, Italy
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
| | - Carlo Tomino
- Scientific Direction, IRCCS San Raffaele Roma, Rome, Italy
| | - Dolores Limongi
- Department of Human Sciences and Quality of Life Promotion, San Raffaele University, Rome, Italy
- Laboratory of Chronic and Neurodegenerative Diseases, IRCCS San Raffaele Roma, Rome, Italy
| |
Collapse
|
29
|
Kochayoo P, Sanguansuttikul P, Thawornpan P, Wangriatisak K, Adams JH, Ntumngia FB, Chootong P. The presence of circulating antibody secreting cells and long-lived memory B cell responses to reticulocyte binding protein 1a in Plasmodium vivax patients. Malar J 2021; 20:474. [PMID: 34930312 PMCID: PMC8686587 DOI: 10.1186/s12936-021-04015-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Accepted: 12/08/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Development of an effective vaccine against blood-stage malaria requires the induction of long-term immune responses. Plasmodium vivax Reticulocyte Binding Protein 1a (PvRBP1a) is a blood-stage parasite antigen which is associated with invasion of red blood cells and induces antibody responses. Thus, PvRBP1a is considered as a target for design of a blood-stage vaccine against vivax malaria. METHODS Both cross-sectional and cohort studies were used to explore the development and persistence of long-lived antibody and memory B cell responses to PvRBP1a in individuals who lived in an area of low malaria endemicity. Antibody titers and frequency of memory B cells specific to PvRBP1a were measured during infection and following recovery for up to 12 months. RESULTS IgG antibody responses against PvRBP1a were prevalent during acute vivax malaria, predominantly IgG1 subclass responses. High responders to PvRBP1a had persistent antibody responses for at least 12-month post-infection. Further analysis of high responder found a direct relation between antibody titers and frequency of activated and atypical memory B cells. Furthermore, circulating antibody secreting cells and memory B cells specific to PvRBP1a were generated during infection. The PvRBP1a-specific memory B cells were maintained for up to 3-year post-infection, indicating the ability of PvRBP1a to induce long-term humoral immunity. CONCLUSION The study revealed an ability of PvRBP1a protein to induce the generation and maintenance of antibody and memory B cell responses. Therefore, PvRBP1a could be considered as a vaccine candidate against the blood-stage of P. vivax.
Collapse
Affiliation(s)
- Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Pattarawan Sanguansuttikul
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Pongsakorn Thawornpan
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - John H Adams
- Center for Global Health and Infectious Diseases Research, University of South Florida, Tampa, FL, 33612, USA
| | - Francis B Ntumngia
- Center for Global Health and Infectious Diseases Research, University of South Florida, Tampa, FL, 33612, USA.
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| |
Collapse
|
30
|
Cossarizza A, Chang HD, Radbruch A, Abrignani S, Addo R, Akdis M, Andrä I, Andreata F, Annunziato F, Arranz E, Bacher P, Bari S, Barnaba V, Barros-Martins J, Baumjohann D, Beccaria CG, Bernardo D, Boardman DA, Borger J, Böttcher C, Brockmann L, Burns M, Busch DH, Cameron G, Cammarata I, Cassotta A, Chang Y, Chirdo FG, Christakou E, Čičin-Šain L, Cook L, Corbett AJ, Cornelis R, Cosmi L, Davey MS, De Biasi S, De Simone G, del Zotto G, Delacher M, Di Rosa F, Di Santo J, Diefenbach A, Dong J, Dörner T, Dress RJ, Dutertre CA, Eckle SBG, Eede P, Evrard M, Falk CS, Feuerer M, Fillatreau S, Fiz-Lopez A, Follo M, Foulds GA, Fröbel J, Gagliani N, Galletti G, Gangaev A, Garbi N, Garrote JA, Geginat J, Gherardin NA, Gibellini L, Ginhoux F, Godfrey DI, Gruarin P, Haftmann C, Hansmann L, Harpur CM, Hayday AC, Heine G, Hernández DC, Herrmann M, Hoelsken O, Huang Q, Huber S, Huber JE, Huehn J, Hundemer M, Hwang WYK, Iannacone M, Ivison SM, Jäck HM, Jani PK, Keller B, Kessler N, Ketelaars S, Knop L, Knopf J, Koay HF, Kobow K, Kriegsmann K, Kristyanto H, Krueger A, Kuehne JF, Kunze-Schumacher H, Kvistborg P, Kwok I, Latorre D, Lenz D, Levings MK, Lino AC, Liotta F, Long HM, Lugli E, MacDonald KN, Maggi L, Maini MK, Mair F, Manta C, Manz RA, Mashreghi MF, Mazzoni A, McCluskey J, Mei HE, Melchers F, Melzer S, Mielenz D, Monin L, Moretta L, Multhoff G, Muñoz LE, Muñoz-Ruiz M, Muscate F, Natalini A, Neumann K, Ng LG, Niedobitek A, Niemz J, Almeida LN, Notarbartolo S, Ostendorf L, Pallett LJ, Patel AA, Percin GI, Peruzzi G, Pinti M, Pockley AG, Pracht K, Prinz I, Pujol-Autonell I, Pulvirenti N, Quatrini L, Quinn KM, Radbruch H, Rhys H, Rodrigo MB, Romagnani C, Saggau C, Sakaguchi S, Sallusto F, Sanderink L, Sandrock I, Schauer C, Scheffold A, Scherer HU, Schiemann M, Schildberg FA, Schober K, Schoen J, Schuh W, Schüler T, Schulz AR, Schulz S, Schulze J, Simonetti S, Singh J, Sitnik KM, Stark R, Starossom S, Stehle C, Szelinski F, Tan L, Tarnok A, Tornack J, Tree TIM, van Beek JJP, van de Veen W, van Gisbergen K, Vasco C, Verheyden NA, von Borstel A, Ward-Hartstonge KA, Warnatz K, Waskow C, Wiedemann A, Wilharm A, Wing J, Wirz O, Wittner J, Yang JHM, Yang J. Guidelines for the use of flow cytometry and cell sorting in immunological studies (third edition). Eur J Immunol 2021; 51:2708-3145. [PMID: 34910301 PMCID: PMC11115438 DOI: 10.1002/eji.202170126] [Citation(s) in RCA: 239] [Impact Index Per Article: 59.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The third edition of Flow Cytometry Guidelines provides the key aspects to consider when performing flow cytometry experiments and includes comprehensive sections describing phenotypes and functional assays of all major human and murine immune cell subsets. Notably, the Guidelines contain helpful tables highlighting phenotypes and key differences between human and murine cells. Another useful feature of this edition is the flow cytometry analysis of clinical samples with examples of flow cytometry applications in the context of autoimmune diseases, cancers as well as acute and chronic infectious diseases. Furthermore, there are sections detailing tips, tricks and pitfalls to avoid. All sections are written and peer-reviewed by leading flow cytometry experts and immunologists, making this edition an essential and state-of-the-art handbook for basic and clinical researchers.
Collapse
Affiliation(s)
- Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Hyun-Dong Chang
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Institute for Biotechnology, Technische Universität, Berlin, Germany
| | - Andreas Radbruch
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Sergio Abrignani
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Richard Addo
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Mübeccel Akdis
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | - Immanuel Andrä
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Francesco Andreata
- Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Annunziato
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Eduardo Arranz
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - Petra Bacher
- Institute of Immunology, Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
- Institute of Clinical Molecular Biology Christian-Albrechts Universität zu Kiel, Kiel, Germany
| | - Sudipto Bari
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
| | - Vincenzo Barnaba
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
- Center for Life Nano & Neuro Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
- Istituto Pasteur - Fondazione Cenci Bolognetti, Rome, Italy
| | | | - Dirk Baumjohann
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Cristian G. Beccaria
- Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
| | - David Bernardo
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
- Centro de Investigaciones Biomédicas en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Dominic A. Boardman
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Jessica Borger
- Department of Immunology and Pathology, Monash University, Melbourne, Victoria, Australia
| | - Chotima Böttcher
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Leonie Brockmann
- Department of Microbiology & Immunology, Columbia University, New York City, USA
| | - Marie Burns
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Dirk H. Busch
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
- German Center for Infection Research (DZIF), Munich, Germany
| | - Garth Cameron
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Ilenia Cammarata
- Dipartimento di Medicina Interna e Specialità Mediche, Sapienza Università di Roma, Rome, Italy
| | - Antonino Cassotta
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
| | - Yinshui Chang
- Medical Clinic III for Oncology, Hematology, Immuno-Oncology and Rheumatology, University Hospital Bonn, University of Bonn, Bonn, Germany
| | - Fernando Gabriel Chirdo
- Instituto de Estudios Inmunológicos y Fisiopatológicos - IIFP (UNLP-CONICET), Facultad de Ciencias Exactas, Universidad Nacional de La Plata, La Plata, Argentina
| | - Eleni Christakou
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Luka Čičin-Šain
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Laura Cook
- BC Children’s Hospital Research Institute, Vancouver, Canada
- Department of Medicine, The University of British Columbia, Vancouver, Canada
| | - Alexandra J. Corbett
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Rebecca Cornelis
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Lorenzo Cosmi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Martin S. Davey
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Gabriele De Simone
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | - Michael Delacher
- Institute for Immunology, University Medical Center Mainz, Mainz, Germany
- Research Centre for Immunotherapy, University Medical Center Mainz, Mainz, Germany
| | - Francesca Di Rosa
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - James Di Santo
- Innate Immunity Unit, Department of Immunology, Institut Pasteur, Paris, France
- Inserm U1223, Paris, France
| | - Andreas Diefenbach
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Mucosal and Developmental Immunology, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Jun Dong
- Cell Biology, German Rheumatism Research Center Berlin (DRFZ), An Institute of the Leibniz Association, Berlin, Germany
| | - Thomas Dörner
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Regine J. Dress
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Charles-Antoine Dutertre
- Institut National de la Sante Et de la Recherce Medicale (INSERM) U1015, Equipe Labellisee-Ligue Nationale contre le Cancer, Villejuif, France
| | - Sidonia B. G. Eckle
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Pascale Eede
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Maximilien Evrard
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | - Christine S. Falk
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Markus Feuerer
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Simon Fillatreau
- Institut Necker Enfants Malades, INSERM U1151-CNRS, UMR8253, Paris, France
- Université de Paris, Paris Descartes, Faculté de Médecine, Paris, France
- AP-HP, Hôpital Necker Enfants Malades, Paris, France
| | - Aida Fiz-Lopez
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
| | - Marie Follo
- Department of Medicine I, Lighthouse Core Facility, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Gemma A. Foulds
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Julia Fröbel
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Nicola Gagliani
- Department of Medicine, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Germany
| | - Giovanni Galletti
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Anastasia Gangaev
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Natalio Garbi
- Institute of Molecular Medicine and Experimental Immunology, Faculty of Medicine, University of Bonn, Germany
| | - José Antonio Garrote
- Mucosal Immunology Lab, Unidad de Excelencia Instituto de Biomedicina y Genética Molecular de Valladolid (IBGM, Universidad de Valladolid-CSIC), Valladolid, Spain
- Laboratory of Molecular Genetics, Servicio de Análisis Clínicos, Hospital Universitario Río Hortega, Gerencia Regional de Salud de Castilla y León (SACYL), Valladolid, Spain
| | - Jens Geginat
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Nicholas A. Gherardin
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Florent Ginhoux
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Shanghai Institute of Immunology, Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Translational Immunology Institute, SingHealth Duke-NUS Academic Medical Centre, Singapore, Singapore
| | - Dale I. Godfrey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Paola Gruarin
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Claudia Haftmann
- Institute of Experimental Immunology, University of Zurich, Zurich, Switzerland
| | - Leo Hansmann
- Department of Hematology, Oncology, and Tumor Immunology, Charité - Universitätsmedizin Berlin (CVK), Berlin, Germany
- Berlin Institute of Health (BIH), Berlin, Germany
- German Cancer Consortium (DKTK), partner site Berlin, Germany
| | - Christopher M. Harpur
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Sciences, Monash University, Clayton, Victoria, Australia
| | - Adrian C. Hayday
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Guido Heine
- Division of Allergy, Department of Dermatology and Allergy, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Daniela Carolina Hernández
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany
| | - Martin Herrmann
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Oliver Hoelsken
- Laboratory of Innate Immunity, Department of Microbiology, Infectious Diseases and Immunology, Charité – Universitätsmedizin Berlin, Campus Benjamin Franklin, Berlin, Germany
- Mucosal and Developmental Immunology, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Qing Huang
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Samuel Huber
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Johanna E. Huber
- Institute for Immunology, Biomedical Center, Faculty of Medicine, LMU Munich, Planegg-Martinsried, Germany
| | - Jochen Huehn
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Michael Hundemer
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - William Y. K. Hwang
- Cancer & Stem Cell Biology, Duke-NUS Medical School, Singapore, Singapore
- Department of Hematology, Singapore General Hospital, Singapore, Singapore
- Executive Offices, National Cancer Centre Singapore, Singapore
| | - Matteo Iannacone
- Division of Immunology, Transplantation and Infectious Diseases, IRCSS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
- Experimental Imaging Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sabine M. Ivison
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Hans-Martin Jäck
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Peter K. Jani
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Nina Kessler
- Institute of Molecular Medicine and Experimental Immunology, Faculty of Medicine, University of Bonn, Germany
| | - Steven Ketelaars
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Laura Knop
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Jasmin Knopf
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Hui-Fern Koay
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
- Australian Research Council Centre of Excellence in Advanced Molecular Imaging, University of Melbourne, Parkville, Victoria, Australia
| | - Katja Kobow
- Department of Neuropathology, Universitätsklinikum Erlangen, Germany
| | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - H. Kristyanto
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Andreas Krueger
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Jenny F. Kuehne
- Institute of Transplant Immunology, Hannover Medical School, Hannover, Germany
| | - Heike Kunze-Schumacher
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Pia Kvistborg
- Division of Molecular Oncology and Immunology, the Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Immanuel Kwok
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
| | | | - Daniel Lenz
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Megan K. Levings
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, Canada
| | - Andreia C. Lino
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Francesco Liotta
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Heather M. Long
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Katherine N. MacDonald
- BC Children’s Hospital Research Institute, Vancouver, Canada
- School of Biomedical Engineering, The University of British Columbia, Vancouver, Canada
- Michael Smith Laboratories, The University of British Columbia, Vancouver, Canada
| | - Laura Maggi
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Mala K. Maini
- Division of Infection & Immunity, Institute of Immunity & Transplantation, University College London, London, UK
| | - Florian Mair
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Calin Manta
- Department of Hematology, Oncology and Rheumatology, University Heidelberg, Heidelberg, Germany
| | - Rudolf Armin Manz
- Institute for Systemic Inflammation Research, University of Luebeck, Luebeck, Germany
| | | | - Alessio Mazzoni
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - James McCluskey
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, Victoria, Australia
| | - Henrik E. Mei
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Fritz Melchers
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Susanne Melzer
- Clinical Trial Center Leipzig, Leipzig University, Härtelstr.16, −18, Leipzig, 04107, Germany
| | - Dirk Mielenz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Leticia Monin
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Lorenzo Moretta
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Gabriele Multhoff
- Radiation Immuno-Oncology Group, Center for Translational Cancer Research (TranslaTUM), Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
- Department of Radiation Oncology, Technical University of Munich (TUM), Klinikum rechts der Isar, Munich, Germany
| | - Luis Enrique Muñoz
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Miguel Muñoz-Ruiz
- Immunosurveillance Laboratory, The Francis Crick Institute, London, UK
| | - Franziska Muscate
- Department of Medicine, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ambra Natalini
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Katrin Neumann
- Institute of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Lai Guan Ng
- Division of Medical Sciences, National Cancer Centre Singapore, Singapore
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- School of Biological Sciences, Nanyang Technological University, Singapore, Singapore
| | | | - Jana Niemz
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | | | - Samuele Notarbartolo
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Lennard Ostendorf
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Laura J. Pallett
- Division of Infection & Immunity, Institute of Immunity & Transplantation, University College London, London, UK
| | - Amit A. Patel
- Institut National de la Sante Et de la Recherce Medicale (INSERM) U1015, Equipe Labellisee-Ligue Nationale contre le Cancer, Villejuif, France
| | - Gulce Itir Percin
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
| | - Giovanna Peruzzi
- Center for Life Nano & Neuro Science@Sapienza, Istituto Italiano di Tecnologia (IIT), Rome, Italy
| | - Marcello Pinti
- Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - A. Graham Pockley
- John van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Nottingham, UK
- Centre for Health, Ageing and Understanding Disease (CHAUD), School of Science and Technology, Nottingham Trent University, Nottingham, UK
| | - Katharina Pracht
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Immo Prinz
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Institute of Systems Immunology, Hamburg Center for Translational Immunology (HCTI), University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Irma Pujol-Autonell
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
- Peter Gorer Department of Immunobiology, King’s College London, London, UK
| | - Nadia Pulvirenti
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Linda Quatrini
- Department of Immunology, IRCCS Bambino Gesù Children’s Hospital, Rome, Italy
| | - Kylie M. Quinn
- School of Biomedical and Health Sciences, RMIT University, Bundorra, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Helena Radbruch
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Hefin Rhys
- Flow Cytometry Science Technology Platform, The Francis Crick Institute, London, UK
| | - Maria B. Rodrigo
- Institute of Molecular Medicine and Experimental Immunology, Faculty of Medicine, University of Bonn, Germany
| | - Chiara Romagnani
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany
| | - Carina Saggau
- Institute of Immunology, Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | | | - Federica Sallusto
- Institute for Research in Biomedicine, Università della Svizzera italiana, Bellinzona, Switzerland
- Institute of Microbiology, ETH Zurich, Zurich, Switzerland
| | - Lieke Sanderink
- Regensburg Center for Interventional Immunology (RCI), Regensburg, Germany
- Chair for Immunology, University Regensburg, Regensburg, Germany
| | - Inga Sandrock
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - Christine Schauer
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Alexander Scheffold
- Institute of Immunology, Christian-Albrechts Universität zu Kiel & Universitätsklinik Schleswig-Holstein, Kiel, Germany
| | - Hans U. Scherer
- Department of Rheumatology, Leiden University Medical Center, Leiden, The Netherlands
| | - Matthias Schiemann
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
| | - Frank A. Schildberg
- Clinic for Orthopedics and Trauma Surgery, University Hospital Bonn, Bonn, Germany
| | - Kilian Schober
- Institut für Medizinische Mikrobiologie, Immunologie und Hygiene, Technische Universität München, Munich, Germany
- Mikrobiologisches Institut – Klinische Mikrobiologie, Immunologie und Hygiene, Universitätsklinikum Erlangen, Friedrich-Alexander-Universität (FAU) Erlangen-Nürnberg, Germany
| | - Janina Schoen
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Wolfgang Schuh
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Thomas Schüler
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Axel R. Schulz
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Sebastian Schulz
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Julia Schulze
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Sonia Simonetti
- Institute of Molecular Biology and Pathology, National Research Council of Italy (CNR), Rome, Italy
| | - Jeeshan Singh
- Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Department of Medicine 3 – Rheumatology and Immunology and Universitätsklinikum Erlangen, Erlangen, Germany
- Deutsches Zentrum für Immuntherapie, Friedrich-Alexander-University Erlangen-Nürnberg and Universitätsklinikum Erlangen, Erlangen, Germany
| | - Katarzyna M. Sitnik
- Department of Viral Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Regina Stark
- Charité Universitätsmedizin Berlin – BIH Center for Regenerative Therapies, Berlin, Germany
- Sanquin Research – Adaptive Immunity, Amsterdam, The Netherlands
| | - Sarah Starossom
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, Berlin, Germany
| | - Christina Stehle
- Innate Immunity, German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Charité – Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Gastroenterology, Infectious Diseases, Rheumatology, Berlin, Germany
| | - Franziska Szelinski
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Leonard Tan
- Singapore Immunology Network (SIgN), Agency for Science, Technology and Research, Singapore, Singapore
- Department of Microbiology & Immunology, Immunology Programme, Life Science Institute, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Attila Tarnok
- Institute for Medical Informatics, Statistics and Epidemiology (IMISE), University of Leipzig, Leipzig, Germany
- Department of Precision Instrument, Tsinghua University, Beijing, China
- Department of Preclinical Development and Validation, Fraunhofer Institute for Cell Therapy and Immunology IZI, Leipzig, Germany
| | - Julia Tornack
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
| | - Timothy I. M. Tree
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Jasper J. P. van Beek
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Willem van de Veen
- Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland
| | | | - Chiara Vasco
- Istituto Nazionale di Genetica Molecolare Romeo ed Enrica Invernizzi (INGM), Milan, Italy
| | - Nikita A. Verheyden
- Institute for Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany
| | - Anouk von Borstel
- Infection and Immunity Program, Department of Biochemistry and Molecular Biology, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Kirsten A. Ward-Hartstonge
- Department of Surgery, The University of British Columbia, Vancouver, Canada
- BC Children’s Hospital Research Institute, Vancouver, Canada
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency, Medical Center – University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Claudia Waskow
- Immunology of Aging, Leibniz Institute on Aging – Fritz Lipmann Institute, Jena, Germany
- Institute of Biochemistry and Biophysics, Faculty of Biological Sciences, Friedrich-Schiller-University Jena, Jena, Germany
- Department of Medicine III, Technical University Dresden, Dresden, Germany
| | - Annika Wiedemann
- German Rheumatism Research Center Berlin (DRFZ), Berlin, Germany
- Department of Medicine/Rheumatology and Clinical Immunology, Charité Universitätsmedizin Berlin, Berlin, Germany
| | - Anneke Wilharm
- Institute of Immunology, Hannover Medical School, Hannover, Germany
| | - James Wing
- Immunology Frontier Research Center, Osaka University, Japan
| | - Oliver Wirz
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jens Wittner
- Division of Molecular Immunology, Nikolaus-Fiebiger-Center, Department of Internal Medicine III, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Jennie H. M. Yang
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King’s College London, UK
- National Institute for Health Research (NIHR) Biomedical Research Center (BRC), Guy’s and St Thomas’ NHS Foundation Trust and King’s College London, London, UK
| | - Juhao Yang
- Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| |
Collapse
|
31
|
Zhong C, Liu F, Hajnik RJ, Yao L, Chen K, Wang M, Liang Y, Sun J, Soong L, Hou W, Hu H. Type I Interferon Promotes Humoral Immunity in Viral Vector Vaccination. J Virol 2021; 95:e0092521. [PMID: 34495698 PMCID: PMC8549508 DOI: 10.1128/jvi.00925-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2021] [Accepted: 09/01/2021] [Indexed: 11/20/2022] Open
Abstract
Recombinant viral vectors represent an important platform for vaccine delivery. Our recent studies have demonstrated distinct innate immune profiles in responding to viral vectors of different families (e.g., adenovirus versus poxvirus): while human Ad5 vector is minimally innate immune stimulatory, the poxviral vector ALVAC induces strong innate response and stimulates type I interferon (IFN) and inflammasome activation. However, the impact of the innate immune signaling on vaccine-induced adaptive immunity in viral vector vaccination is less clear. Here, we show that Modified Vaccinia Ankara (MVA), another poxviral vector, stimulated a type I IFN response in innate immune cells through cGAS-STING. Using MVA-HIV vaccine as a model, we found that type I IFN signaling promoted the generation of humoral immunity in MVA-HIV vaccination in vivo. Following vaccination, type I IFN receptor-knockout (IFNAR1-/-) mice produced significantly lower levels of total and HIV gp120-specific antibodies compared to wild-type (WT) mice. Consistent with the antibody response, a type I IFN signaling deficiency also led to reduced levels of plasma cells and memory-like B cells compared to WT mice. Furthermore, analysis of vaccine-induced CD4 T cells showed that type I IFN signaling also promoted the generation of a vaccine-specific CD4 T-cell response and a T follicular helper (Tfh) response in mice. Together, our data indicate a role for type I IFN signaling in promoting humoral immunity in poxviral vector vaccination. The study suggests that modulating type I IFN and its associated innate immune pathways will likely affect vaccine efficacy. IMPORTANCE Viral vectors, including MVA, are an important antigen delivery platform and have been commonly used in vaccine development. Understanding the innate host-viral vector interactions and their impact on vaccine-induced immunity is critical but understudied. Using MVA-HIV vaccination of WT and IFNAR1-/- mice as a model, we report that type I IFN signaling promotes humoral immunity in MVA vaccination, including vaccine-induced antibody, B-cell, and Tfh responses. Our findings provide insights that not only add to our basic understanding of host-viral vector interactions but also will aid in improving vaccine design by potentially modulating type I IFN and its associated innate immune pathways in viral vector vaccination.
Collapse
Affiliation(s)
- Chaojie Zhong
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Fengliang Liu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Renee J. Hajnik
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lei Yao
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Kangjing Chen
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Meirong Wang
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Yuejin Liang
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
| | - Jiaren Sun
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Lynn Soong
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Wei Hou
- School of Basic Medical Sciences, Wuhan University, Wuhan, Hubei, China
| | - Haitao Hu
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, Texas, USA
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
32
|
Piano Mortari E, Russo C, Vinci MR, Terreri S, Fernandez Salinas A, Piccioni L, Alteri C, Colagrossi L, Coltella L, Ranno S, Linardos G, Agosta M, Albano C, Agrati C, Castilletti C, Meschi S, Romania P, Roscilli G, Pavoni E, Camisa V, Santoro A, Brugaletta R, Magnavita N, Ruggiero A, Cotugno N, Amodio D, Ciofi Degli Atti ML, Giorgio D, Russo N, Salvatori G, Corsetti T, Locatelli F, Perno CF, Zaffina S, Carsetti R. Highly Specific Memory B Cells Generation after the 2nd Dose of BNT162b2 Vaccine Compensate for the Decline of Serum Antibodies and Absence of Mucosal IgA. Cells 2021; 10:cells10102541. [PMID: 34685521 PMCID: PMC8533837 DOI: 10.3390/cells10102541] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/15/2021] [Accepted: 09/22/2021] [Indexed: 11/16/2022] Open
Abstract
Specific memory B cells and antibodies are a reliable read-out of vaccine efficacy. We analysed these biomarkers after one and two doses of BNT162b2 vaccine. The second dose significantly increases the level of highly specific memory B cells and antibodies. Two months after the second dose, specific antibody levels decline, but highly specific memory B cells continue to increase, thus predicting a sustained protection from COVID-19. We show that although mucosal IgA is not induced by the vaccination, memory B cells migrate in response to inflammation and secrete IgA at mucosal sites. We show that the first vaccine dose may lead to an insufficient number of highly specific memory B cells and low concentration of serum antibodies, thus leaving vaccinees without the immune robustness needed to ensure viral elimination and herd immunity. We also clarify that the reduction of serum antibodies does not diminish the force and duration of the immune protection induced by vaccination. The vaccine does not induce sterilizing immunity. Infection after vaccination may be caused by the lack of local preventive immunity because of the absence of mucosal IgA.
Collapse
Affiliation(s)
- Eva Piano Mortari
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
| | - Cristina Russo
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Maria Rosaria Vinci
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Sara Terreri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
| | - Ane Fernandez Salinas
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Department of Molecular Medicine, Sapienza University of Rome, Viale dell’Università, 37, 00185 Rome, Italy
| | - Livia Piccioni
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Claudia Alteri
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Department of Oncology and Hemato-Oncology, University of Milan, Via festa del Perdono, 7, 20122 Milan, Italy
| | - Luna Colagrossi
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Luana Coltella
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Stefania Ranno
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Giulia Linardos
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Marilena Agosta
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Christian Albano
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
| | - Chiara Agrati
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 2, 00146 Rome, Italy; (C.A.); (C.C.); (S.M.)
| | - Concetta Castilletti
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 2, 00146 Rome, Italy; (C.A.); (C.C.); (S.M.)
| | - Silvia Meschi
- National Institute for Infectious Diseases Lazzaro Spallanzani, IRCCS, Via Portuense, 2, 00146 Rome, Italy; (C.A.); (C.C.); (S.M.)
| | - Paolo Romania
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Department of Molecular Medicine, Sapienza University of Rome, Viale dell’Università, 37, 00185 Rome, Italy
| | - Giuseppe Roscilli
- Takis s.r.l., Via di Castel Romano, 100, 00128 Rome, Italy; (G.R.); (E.P.)
| | - Emiliano Pavoni
- Takis s.r.l., Via di Castel Romano, 100, 00128 Rome, Italy; (G.R.); (E.P.)
| | - Vincenzo Camisa
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Annapaola Santoro
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Rita Brugaletta
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Nicola Magnavita
- Section of Occupational Medicine and Labor Law, Post-Graduate School of Occupational Health, University Cattolica del Sacro Cuore, Largo Francesco Vito, 1, 00168 Rome, Italy;
- Department of Woman, Child & Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Via della Pineta Sacchetti, 217, 00168 Rome, Italy
| | - Alessandra Ruggiero
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (A.R.); (N.C.); (D.A.)
- Department of Neuroscience, Biomedicine and Movement Sciences, University of Verona, Via San Francesco, 22, 37129 Verona, Italy
| | - Nicola Cotugno
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (A.R.); (N.C.); (D.A.)
| | - Donato Amodio
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (A.R.); (N.C.); (D.A.)
| | - Marta Luisa Ciofi Degli Atti
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy;
| | - Daniela Giorgio
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (D.G.); (N.R.); (G.S.)
| | - Nicoletta Russo
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (D.G.); (N.R.); (G.S.)
| | - Guglielmo Salvatori
- Neonatal Intensive Care Unit and Human Milk Bank, Department of Neonatology, Bambino Gesù Children’s Hospital, IRCSS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (D.G.); (N.R.); (G.S.)
| | - Tiziana Corsetti
- Hospital Pharmacy Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy;
| | - Franco Locatelli
- Department of Hematology/Oncology, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy;
- Department of Pediatrics, Sapienza, University of Rome, Viale dell’Università, 37, 00185 Rome, Italy
| | - Carlo Federico Perno
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
| | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo, 15, 00146 Rome, Italy; (M.R.V.); (V.C.); (A.S.); (R.B.); (S.Z.)
- Health Directorate, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy
| | - Rita Carsetti
- Diagnostic Immunology Research Unit, Multimodal Medicine Research Area, Bambino Gesù Children’s Hospital, IRCCS, Viale di San Paolo,15, 00146 Rome, Italy; (E.P.M.); (S.T.); (A.F.S.); (C.A.); (C.A.); (P.R.); (C.F.P.)
- Microbiology and Diagnostic Immunology Unit, Bambino Gesù Children’s Hospital, IRCCS, Piazza Sant’Onofrio, 4, 00165 Rome, Italy; (C.R.); (L.P.); (L.C.); (L.C.); (S.R.); (G.L.); (M.A.)
- Correspondence:
| |
Collapse
|
33
|
The Bone Marrow as Sanctuary for Plasma Cells and Memory T-Cells: Implications for Adaptive Immunity and Vaccinology. Cells 2021; 10:cells10061508. [PMID: 34203839 PMCID: PMC8232593 DOI: 10.3390/cells10061508] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 05/31/2021] [Accepted: 06/08/2021] [Indexed: 12/20/2022] Open
Abstract
The bone marrow (BM) is key to protective immunological memory because it harbors a major fraction of the body’s plasma cells, memory CD4+ and memory CD8+ T-cells. Despite its paramount significance for the human immune system, many aspects of how the BM enables decade-long immunity against pathogens are still poorly understood. In this review, we discuss the relationship between BM survival niches and long-lasting humoral immunity, how intrinsic and extrinsic factors define memory cell longevity and show that the BM is also capable of adopting many responsibilities of a secondary lymphoid organ. Additionally, with more and more data on the differentiation and maintenance of memory T-cells and plasma cells upon vaccination in humans being reported, we discuss what factors determine the establishment of long-lasting immunological memory in the BM and what we can learn for vaccination technologies and antigen design. Finally, using these insights, we touch on how this holistic understanding of the BM is necessary for the development of modern and efficient vaccines against the pandemic SARS-CoV-2.
Collapse
|
34
|
Grasseau A, Boudigou M, Michée-Cospolite M, Delaloy C, Mignen O, Jamin C, Cornec D, Pers JO, Le Pottier L, Hillion S. The diversity of the plasmablast signature across species and experimental conditions: A meta-analysis. Immunology 2021; 164:120-134. [PMID: 34041745 DOI: 10.1111/imm.13344] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 03/15/2021] [Accepted: 04/12/2021] [Indexed: 12/17/2022] Open
Abstract
Antibody-secreting cells (ASC) are divided into two principal subsets, including the long-lived plasma cell (PC) subset residing in the bone marrow and the short-lived subset, also called plasmablast (PB). PB are described as a proliferating subset circulating through the blood and ending its differentiation in tissues. Due to their inherent heterogeneity, the molecular signature of PB is not fully established. The purpose of this study was to decipher a specific PB signature in humans and mice through a comprehensive meta-analysis of different data sets exploring the PB differentiation in both species and across different experimental conditions. The present study used recent analyses using whole RNA sequencing in prdm1-GFP transgenic mice to define a reliable and accurate PB signature. Next, we performed similar analysis using current data sets obtained from human PB and PC. The PB-specific signature is composed of 155 and 113 genes in mouse and human being, respectively. Although only nine genes are shared between the human and mice PB signature, the loss of B-cell identity such as the down-regulation of PAX5, MS4A1, (CD20) CD22 and IL-4R is a conserved feature across species and across the different experimental conditions. Additionally, we observed that the IRF8 and IRF4 transcription factors have a specific dynamic range of expression in human PB. We thus demonstrated that IRF4/IRF8 intranuclear staining was useful to define PB in vivo and in vitro and able to discriminate between atypical PB populations and transient states.
Collapse
Affiliation(s)
| | | | | | - Céline Delaloy
- UMR U1236, INSERM, Etablissement Français du Sang (EFS) de Bretagne, Université de Rennes 1, Rennes, France
| | | | - Christophe Jamin
- UMR1227, LBAI, INSERM, Univ Brest, Brest, France.,UMR1227, LBAI, INSERM, CHU de Brest, Univ Brest, Brest, France
| | - Divi Cornec
- UMR1227, LBAI, INSERM, Univ Brest, Brest, France.,UMR1227, LBAI, INSERM, CHU de Brest, Univ Brest, Brest, France
| | - Jacques-Olivier Pers
- UMR1227, LBAI, INSERM, Univ Brest, Brest, France.,UMR1227, LBAI, INSERM, CHU de Brest, Univ Brest, Brest, France
| | | | - Sophie Hillion
- UMR1227, LBAI, INSERM, Univ Brest, Brest, France.,UMR1227, LBAI, INSERM, CHU de Brest, Univ Brest, Brest, France
| |
Collapse
|
35
|
Bestard O, Couzi L, Crespo M, Kessaris N, Thaunat O. Stratifying the humoral risk of candidates to a solid organ transplantation: a proposal of the ENGAGE working group. Transpl Int 2021; 34:1005-1018. [PMID: 33786891 DOI: 10.1111/tri.13874] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 03/26/2021] [Indexed: 12/30/2022]
Abstract
Detection of circulating antibodies directed against human leukocyte antigen (HLA) molecules, which corresponds to the current definition of 'sensitized patient', has been shown to have a severe impact on both access to transplantation and, if the anti-HLA antibodies are specific to the selected donor, survival of the graft. However, not all donor-specific antibodies (DSA) are equally harmful to the graft and progress in the understanding of humoral memory has led to the conclusion that absence of DSA at transplantation does not rule out the possibility that the patient has a preformed cellular humoral memory against the graft (thereby defining a category of DSA-negative sensitized recipients). Technological progress has led to the generation of new assays that offer unprecedented precision in exploring the different layers (serological and cellular) of alloimmune humoral memory. Based on this recent knowledge, the EuropeaN Guidelines for the mAnagement of Graft rEcipients (ENGAGE) working group to propose an updated definition of sensitization in candidates for solid organ transplantation - one that moves away from the current binary division towards a definition based on homogenous strata with similar humoral risk.
Collapse
Affiliation(s)
- Oriol Bestard
- Kidney Transplant Unit, Nephrology Department, Bellvitge University Hospital, Barcelona, Spain.,Bellvitge Research Institute (IDIBELL), Barcelona, Spain
| | - Lionel Couzi
- Nephrology-Transplantation-Dialysis, CHU Bordeaux, Bordeaux, France.,CNRS-UMR 5164 Immuno ConcEpT, Bordeaux University, Bordeaux, France
| | - Marta Crespo
- Department of Nephrology, Hospital del Mar, Barcelona, Spain.,Nephropathies Research Group, Hospital del Mar Medical Research Institute (IMIM), Barcelona, Spain
| | - Nicos Kessaris
- Department of Nephrology and Transplantation, Guy's Hospital, London, UK.,King's College London, London, UK
| | - Olivier Thaunat
- Service de Transplantation, Néphrologie et Immunologie Clinique, Hôpital Edouard Herriot, Lyon, France.,Université Claude-Bernard Lyon I, Lyon, France.,Institut National de la Santé et de la Recherche Médicale U1111, Lyon, France
| |
Collapse
|
36
|
Park JW, Lagniton PN, Liu Y, Xu RH. mRNA vaccines for COVID-19: what, why and how. Int J Biol Sci 2021; 17:1446-1460. [PMID: 33907508 PMCID: PMC8071766 DOI: 10.7150/ijbs.59233] [Citation(s) in RCA: 188] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Accepted: 03/15/2021] [Indexed: 01/09/2023] Open
Abstract
The Coronavirus disease-19 (COVID-19) pandemic, caused by severe acute respiratory syndrome coronavirus -2 (SARS-CoV-2), has impacted human lives in the most profound ways with millions of infections and deaths. Scientists and pharmaceutical companies have been in race to produce vaccines against SARS-CoV-2. Vaccine generation usually demands years of developing and testing for efficacy and safety. However, it only took less than one year to generate two mRNA vaccines from their development to deployment. The rapid production time, cost-effectiveness, versatility in vaccine design, and clinically proven ability to induce cellular and humoral immune response have crowned mRNA vaccines with spotlights as most promising vaccine candidates in the fight against the pandemic. In this review, we discuss the general principles of mRNA vaccine design and working mechanisms of the vaccines, and provide an up-to-date summary of pre-clinical and clinical trials on seven anti-COVID-19 mRNA candidate vaccines, with the focus on the two mRNA vaccines already licensed for vaccination. In addition, we highlight the key strategies in designing mRNA vaccines to maximize the expression of immunogens and avoid intrinsic innate immune response. We also provide some perspective for future vaccine development against COVID-19 and other pathogens.
Collapse
Affiliation(s)
| | | | | | - Ren-He Xu
- Institute of Translational Medicine, and Centre of Reproduction, Development and Aging, Faculty of Health Sciences, University of Macau, Taipa, Macau, China
| |
Collapse
|
37
|
Grimsholm O, Piano Mortari E, Davydov AN, Shugay M, Obraztsova AS, Bocci C, Marasco E, Marcellini V, Aranburu A, Farroni C, Silvestris DA, Cristofoletti C, Giorda E, Scarsella M, Cascioli S, Barresi S, Lougaris V, Plebani A, Cancrini C, Finocchi A, Moschese V, Valentini D, Vallone C, Signore F, de Vincentiis G, Zaffina S, Russo G, Gallo A, Locatelli F, Tozzi AE, Tartaglia M, Chudakov DM, Carsetti R. The Interplay between CD27 dull and CD27 bright B Cells Ensures the Flexibility, Stability, and Resilience of Human B Cell Memory. Cell Rep 2021; 30:2963-2977.e6. [PMID: 32130900 DOI: 10.1016/j.celrep.2020.02.022] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 12/23/2019] [Accepted: 02/05/2020] [Indexed: 10/24/2022] Open
Abstract
Memory B cells (MBCs) epitomize the adaptation of the immune system to the environment. We identify two MBC subsets in peripheral blood, CD27dull and CD27bright MBCs, whose frequency changes with age. Heavy chain variable region (VH) usage, somatic mutation frequency replacement-to-silent ratio, and CDR3 property changes, reflecting consecutive selection of highly antigen-specific, low cross-reactive antibody variants, all demonstrate that CD27dull and CD27bright MBCs represent sequential MBC developmental stages, and stringent antigen-driven pressure selects CD27dull into the CD27bright MBC pool. Dynamics of human MBCs are exploited in pregnancy, when 50% of maternal MBCs are lost and CD27dull MBCs transit to the more differentiated CD27bright stage. In the postpartum period, the maternal MBC pool is replenished by the expansion of persistent CD27dull clones. Thus, the stability and flexibility of human B cell memory is ensured by CD27dull MBCs that expand and differentiate in response to change.
Collapse
Affiliation(s)
- Ola Grimsholm
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy; Department of Rheumatology and Inflammation Research, University of Gothenburg, Box 480, 405 30 Gothenburg, Sweden
| | - Eva Piano Mortari
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Alexey N Davydov
- Central European Institute of Technology, 625 00 Brno, Czech Republic
| | - Mikhail Shugay
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, 101000 Moscow, Russia
| | - Anna S Obraztsova
- Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, 101000 Moscow, Russia
| | - Chiara Bocci
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Emiliano Marasco
- Division of Rheumatology, Bambino Gesù Children's Hospital IRCCS, 00146 Roma, Italy
| | - Valentina Marcellini
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Alaitz Aranburu
- Department of Rheumatology and Inflammation Research, University of Gothenburg, Box 480, 405 30 Gothenburg, Sweden
| | - Chiara Farroni
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | | | | | - Ezio Giorda
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Marco Scarsella
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Simona Cascioli
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Sabina Barresi
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, 00146 Rome, Italy
| | - Vassilios Lougaris
- Department of Experimental and Clinical Sciences, University of Brescia, 25121 Brescia, Italy
| | - Alessandro Plebani
- DPUO, Division of Immuno-Infectivology, University Department of Pediatrics, 00146 Bambino Gesù Children's Hospital, Rome, Italy
| | - Caterina Cancrini
- DPUO, Division of Immuno-Infectivology, University Department of Pediatrics, 00146 Bambino Gesù Children's Hospital, Rome, Italy; School of Medicine, University of Tor Vergata, 00133 Rome, Italy
| | - Andrea Finocchi
- DPUO, Division of Immuno-Infectivology, University Department of Pediatrics, 00146 Bambino Gesù Children's Hospital, Rome, Italy; School of Medicine, University of Tor Vergata, 00133 Rome, Italy
| | - Viviana Moschese
- Pediatric Immunology Unit, Policlinico Tor Vergata, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Diletta Valentini
- Pediatric and Infectious Disease Unit, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | - Cristina Vallone
- Department of Obstetrics and Gynaecology, Misericordia Hospital Grosseto, Usl Toscana Sud-est, 58100 Grosseto, Italy
| | - Fabrizio Signore
- Department of Obstetrics and Gynaecology, Misericordia Hospital Grosseto, Usl Toscana Sud-est, 58100 Grosseto, Italy
| | | | - Salvatore Zaffina
- Occupational Medicine/Health Technology Assessment and Safety Research Unit, Clinical-Technological Innovations Research Area, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy
| | | | - Angela Gallo
- Oncohaematology Department, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy
| | - Franco Locatelli
- Oncohaematology Department, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy; Department of Pediatrics, Sapienza, University of Rome, 00161 Rome, Italy
| | - Alberto E Tozzi
- Multifactorial Disease and Complex Phenotype Research Area, Bambino Gesù Children's Hospital, IRCSS, 00146 Rome, Italy
| | - Marco Tartaglia
- Genetics and Rare Diseases Research Division, Bambino Gesù Children's Hospital, 00146 Rome, Italy
| | - Dmitriy M Chudakov
- Central European Institute of Technology, 625 00 Brno, Czech Republic; Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, 117997 Moscow, Russia; Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997 Moscow, Russia; Center of Life Sciences, Skolkovo Institute of Science and Technology, 101000 Moscow, Russia
| | - Rita Carsetti
- B Cell Pathophysiology Unit, Immunology Research Area, Bambino Gesù Children's Hospital IRCCS, 00146 Rome, Italy; Diagnostic Immunology Unit, Department of Laboratories, Bambino Gesù Children's Hospital, IRCCS, 00146 Rome, Italy.
| |
Collapse
|
38
|
Sokal A, Chappert P, Barba-Spaeth G, Roeser A, Fourati S, Azzaoui I, Vandenberghe A, Fernandez I, Meola A, Bouvier-Alias M, Crickx E, Beldi-Ferchiou A, Hue S, Languille L, Michel M, Baloul S, Noizat-Pirenne F, Luka M, Mégret J, Ménager M, Pawlotsky JM, Fillatreau S, Rey FA, Weill JC, Reynaud CA, Mahévas M. Maturation and persistence of the anti-SARS-CoV-2 memory B cell response. Cell 2021; 184:1201-1213.e14. [PMID: 33571429 PMCID: PMC7994111 DOI: 10.1016/j.cell.2021.01.050] [Citation(s) in RCA: 236] [Impact Index Per Article: 59.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 01/09/2021] [Accepted: 01/26/2021] [Indexed: 12/15/2022]
Abstract
Memory B cells play a fundamental role in host defenses against viruses, but to date, their role has been relatively unsettled in the context of SARS-CoV-2. We report here a longitudinal single-cell and repertoire profiling of the B cell response up to 6 months in mild and severe COVID-19 patients. Distinct SARS-CoV-2 spike-specific activated B cell clones fueled an early antibody-secreting cell burst as well as a durable synchronous germinal center response. While highly mutated memory B cells, including pre-existing cross-reactive seasonal Betacoronavirus-specific clones, were recruited early in the response, neutralizing SARS-CoV-2 RBD-specific clones accumulated with time and largely contributed to the late, remarkably stable, memory B cell pool. Highlighting germinal center maturation, these cells displayed clear accumulation of somatic mutations in their variable region genes over time. Overall, these findings demonstrate that an antigen-driven activation persisted and matured up to 6 months after SARS-CoV-2 infection and may provide long-term protection.
Collapse
Affiliation(s)
- Aurélien Sokal
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Pascal Chappert
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France; Inovarion, Paris, France
| | | | - Anais Roeser
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Slim Fourati
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France; INSERM U955, équipe 18, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Imane Azzaoui
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Alexis Vandenberghe
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Ignacio Fernandez
- Institut Pasteur, Unité de Virologie Structurale, CNRS UMR 3569, Paris, France
| | - Annalisa Meola
- Institut Pasteur, Unité de Virologie Structurale, CNRS UMR 3569, Paris, France
| | - Magali Bouvier-Alias
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France; INSERM U955, équipe 18, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Etienne Crickx
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Asma Beldi-Ferchiou
- Département Immunologie-Hématologie, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France; INSERM U955, équipe immunorégulation et biothérapie (I-BIOT), Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Sophie Hue
- Département Immunologie-Hématologie, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), 94000 Créteil, France; Institut de Recherche Vaccinale (VRI), Université Paris-Est Créteil (UPEC), Faculté de Médecine, Créteil, France; INSERM U955, équipe 16, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Laetitia Languille
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Marc Michel
- Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Samia Baloul
- Département de Santé Publique, Unité de Recherche Clinique (URC), CEpiA (Clinical Epidemiology and Ageing), EA 7376, Institut Mondor de Recherche Biomédicale (IMRB), Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France
| | - France Noizat-Pirenne
- Etablissement Français du Sang, INSERM U955, Université Paris-Est Créteil (UPEC), Créteil, France
| | - Marine Luka
- Réponses inflammatoires et réseaux transcriptomiques dans les maladies, Institut Imagine, INSERM UMR1163, ATIP-Avenir Team, Université de Paris, Paris, France; Labtech Single-cell@Imagine, Institut Imagine, INSERM UMR 1163, Paris, France
| | - Jérôme Mégret
- Plateforme de Cytométrie en Flux, Structure Fédérative de Recherche Necker, INSERM US24-CNRS UMS3633, Paris, France
| | - Mickaël Ménager
- Réponses inflammatoires et réseaux transcriptomiques dans les maladies, Institut Imagine, INSERM UMR1163, ATIP-Avenir Team, Université de Paris, Paris, France; Labtech Single-cell@Imagine, Institut Imagine, INSERM UMR 1163, Paris, France
| | - Jean-Michel Pawlotsky
- Département de Virologie, Bactériologie, Hygiène et Mycologie-Parasitologie, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Créteil, France; INSERM U955, équipe 18, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France
| | - Simon Fillatreau
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France
| | - Felix A Rey
- Institut Pasteur, Unité de Virologie Structurale, CNRS UMR 3569, Paris, France
| | - Jean-Claude Weill
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France.
| | - Claude-Agnès Reynaud
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France.
| | - Matthieu Mahévas
- Institut Necker Enfants Malades (INEM), INSERM U1151/CNRS UMS 8253, Université de Paris, Paris, France; Service de Médecine Interne, Centre Hospitalier Universitaire Henri-Mondor, Assistance Publique-Hôpitaux de Paris (AP-HP), Université Paris-Est Créteil (UPEC), Créteil, France; INSERM U955, équipe 2, Institut Mondor de Recherche Biomédicale (IMRB), Université Paris-Est Créteil (UPEC), Créteil, France.
| |
Collapse
|
39
|
Adam A, Cuellar S, Wang T. Memory B cell and antibody responses to flavivirus infection and vaccination. Fac Rev 2021; 10:5. [PMID: 33659923 PMCID: PMC7894259 DOI: 10.12703/r/10-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Flaviviruses are a group of mosquito- or tick-borne single-stranded RNA viruses that can cause a wide range of clinical manifestations in humans and animals, including asymptomatic, flu-like febrile illness, hemorrhagic fever, encephalitis, birth defects, and death. Many of them have no licensed vaccines available for human use. Memory B cell development and induction of neutralizing antibody responses, which are important for the control of flavivirus infection and dissemination, have been used as biomarkers for vaccine efficacy. In this review, we will discuss recent findings on memory B cells and antibody responses from studies in clinical specimen and animal models of flavivirus infection and vaccination with a focus on several clinically important flaviviruses, including dengue, West Nile, yellow fever, and Zika viruses.
Collapse
Affiliation(s)
- Awadalkareem Adam
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Servando Cuellar
- School of Medicine, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Tian Wang
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, 77555, USA
- Sealy Institute for Vaccine Sciences, University of Texas Medical Branch, Galveston, TX, 77555, USA
| |
Collapse
|
40
|
Muir YSS, Bryant B, Campbell-Ward M, Higgins DP. Retrospective anti-tetanus antibody responses of zoo-based Asian elephants (Elephas maximus) and rhinoceros (Rhinocerotidae). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 114:103841. [PMID: 32861731 DOI: 10.1016/j.dci.2020.103841] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Revised: 08/19/2020] [Accepted: 08/19/2020] [Indexed: 06/11/2023]
Abstract
Tetanus toxoids (TT) commercially available for use in horses and livestock are commonly used to vaccinate elephants and rhinoceros that are in human care. Although recommendations for booster intervals have changed in human and horse protocols to reduce the risks associated with hyper-immunity (i.e. B-cell anergy and hypersensitivity reactions) these have generally not been adopted in zoo protocols. Additionally, there is no evidence to demonstrate commercial TT immunogenicity in rhinoceros. In this study, a preliminary analysis of rhinoceros antibody responses to TT was conducted, in addition to an exploration of the impact of various booster frequencies on antibody responses in elephant. Retrospective analysis of archived serum samples was conducted for 9 Asian elephants (Elephas maximus), 7 southern black (Diceros bicornis minor), one southern white (Ceratotherium simum simum), and two greater one-horned (Rhinoceros unicornis) rhinoceros. Pre-vaccination (baseline) samples and those following priming vaccination (rhinoceros only), annual and non-annual boosters were targeted. A commercially available competitive ELISA kit was used to quantify serum anti-TT antibodies. Average baseline and post-vaccination anti-tetanus antibody concentrations were greater in elephant (92 mg/L ± 42, n = 3, N = 3; 125 ± 76, n = 82, N = 9) than in rhinoceros (47 mg/L ± 39, n = 8, N = 8; 44 mg/L ± 37, n = 16, N = 7). Rhinoceros antibody concentrations did not differ markedly following vaccinations from their naturally acquired high pre-vaccination concentrations. Eight elephants demonstrated antibody maintenance for 3-5 years without a tetanus booster. Additionally, although five out of nine elephants developed local reactions consistent with delayed type IV hypersensitivity following some boosters, there was no association between high antibody concentrations and increased incidence of adverse reactions. In addition, no decrease in antibody concentrations was detected as a result of annual vaccination in elephants, though this does not entirely rule out potential for B-cell anergy.
Collapse
Affiliation(s)
| | - Benn Bryant
- Taronga Western Plains Zoo, Obley Rd, Dubbo, 2830, NSW, Australia
| | | | - Damien P Higgins
- Sydney School of Veterinary Science, The University of Sydney, Camperdown, 2006, NSW, Australia.
| |
Collapse
|
41
|
Skin-Associated B Cells in the Pathogenesis of Cutaneous Autoimmune Diseases-Implications for Therapeutic Approaches. Cells 2020; 9:cells9122627. [PMID: 33297481 PMCID: PMC7762338 DOI: 10.3390/cells9122627] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/01/2020] [Accepted: 12/04/2020] [Indexed: 12/11/2022] Open
Abstract
B lymphocytes are crucial mediators of systemic immune responses and are known to be substantial in the pathogenesis of autoimmune diseases with cutaneous manifestations. Amongst them are lupus erythematosus, dermatomyositis, systemic sclerosis and psoriasis, and particularly those driven by autoantibodies such as pemphigus and pemphigoid. However, the concept of autoreactive skin-associated B cells, which may reside in the skin and locally contribute to chronic inflammation, is gradually evolving. These cells are believed to differ from B cells of primary and secondary lymphoid organs and may provide additional features besides autoantibody production, including cytokine expression and crosstalk to autoreactive T cells in an antigen-presenting manner. In chronically inflamed skin, B cells may appear in tertiary lymphoid structures. Those abnormal lymph node-like structures comprise a network of immune and stromal cells possibly enriched by vascular structures and thus constitute an ideal niche for local autoimmune responses. In this review, we describe current considerations of different B cell subsets and their assumed role in skin autoimmunity. Moreover, we discuss traditional and B cell-associated approaches for the treatment of autoimmune skin diseases, including drugs targeting B cells (e.g., CD19- and CD20-antibodies), plasma cells (e.g., proteasome inhibitors, CXCR4 antagonists), activated pathways (such as BTK- and PI3K-inhibitors) and associated activator molecules (BLyS, APRIL).
Collapse
|
42
|
Ni Z, Xing D, Zhang T, Ding N, Xiang D, Zhao Z, Qu J, Hu C, Shen X, Xue X, Zhou J. Tumor-infiltrating B cell is associated with the control of progression of gastric cancer. Immunol Res 2020; 69:43-52. [PMID: 33236222 DOI: 10.1007/s12026-020-09167-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Accepted: 11/16/2020] [Indexed: 01/13/2023]
Abstract
This study aimed to further explore the clinicopathological correlation of B cell infiltration in gastric cancer (GC) and its impact on prognostic. By immunohistochemical method, CD20+ B cells, CD3+ T cells, CD66b+ tumor-associated neutrophils, CD163+ tumor-associated macrophages, and CD57+ natural killer cells were analyzed in consecutive sections of 584 GC tissues and 69 normal adjacent tissues. Kaplan-Meier and Cox regression analyses determined the relationship between clinical relevance or prognosis and B cell infiltration. The correlation between total B cell infiltration and various T cell subtype infiltration in GC tissues from 407 patients in the TCGA data was also analyzed. Kaplan-Meier and Cox regression analyses determined the effects of total B cell infiltration and various B cell subtype infiltration on the prognosis of patients with GC. The infiltration level of CD20+ B cells was positively correlated with that of T cells (risk ratio [RR] = 0.0930), especially CD4+ T cells and CD8+ T cells (P < 0.05). A high level of CD20+ B cell infiltration was significantly associated with low lymph node involvement and low TNM stage (P < 0.05). High levels of CD20+ B cell infiltration were significantly associated with improvements in overall survival and disease-free survival. Univariate Cox regression and multivariate Cox regression analysis showed that CD20+ B cell infiltration was an independent protective factor of prognosis. Higher levels of class-switched memory B cell and plasma cell also reflected better overall survival, and class-switched memory B cell and plasma cell were independent protective factors for prognosis. The findings indicate that B cell infiltration in GC, especially switched memory B cells and plasma cells, has a significant effect on tumor progression and prognosis.
Collapse
Affiliation(s)
- Zhonglin Ni
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China
| | - Dong Xing
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, University Town, Chashan, Wenzhou, 325035, Zhejiang Province, China
| | - Teming Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Ning Ding
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, University Town, Chashan, Wenzhou, 325035, Zhejiang Province, China
| | - Dan Xiang
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, University Town, Chashan, Wenzhou, 325035, Zhejiang Province, China
| | - Zhiguang Zhao
- Department of Pathology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, No. 109 West Xueyuan Road, Lucheng District, Wenzhou, 325027, Zhejiang Province, China.
| | - Jinmiao Qu
- Department of Oncology, The First Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325035, Zhejiang Province, China
| | - Changyuan Hu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, Zhejiang Province, China
| | - Xian Shen
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang Province, China
| | - Xiangyang Xue
- Department of Microbiology and Immunology, Institute of Molecular Virology and Immunology, School of Basic Medical Sciences, Wenzhou Medical University, University Town, Chashan, Wenzhou, 325035, Zhejiang Province, China.
| | - Jie Zhou
- Division of Hepatobiliopancreatic Surgery, Department of General Surgery, Nanfang Hospital, Southern Medical University, No. 1838 North Guangzhou Avenue, Guangzhou, 510515, Guangdong Province, China.
| |
Collapse
|
43
|
Al‐Katib AM, Gaith H, Sano D, Al‐Katib S, Bonnett M, Kafri Z. Emergence of overt myeloma in a patient with chronic lymphocytic leukemia on ibrutinib therapy. Clin Case Rep 2020; 8:1797-1801. [PMID: 32983498 PMCID: PMC7495781 DOI: 10.1002/ccr3.3019] [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: 02/19/2020] [Revised: 04/20/2020] [Accepted: 05/12/2020] [Indexed: 11/09/2022] Open
Abstract
Ibrutinib is approved for chronic lymphocytic leukemia (CLL). However, its role in the treatment of multiple myeloma (MM) is not clear and is under investigation. We report a case of CLL that developed MM while on therapy with ibrutinib indicating that this drug may not be active against MM.
Collapse
Affiliation(s)
- Ayad M. Al‐Katib
- Lymphoma Research LaboratoryDepartment of Internal MedicineWayne State University School of Medicine (WSU SOM)DetroitMIUSA
| | - Hussein Gaith
- Lymphoma Research LaboratoryDepartment of Internal MedicineWayne State University School of Medicine (WSU SOM)DetroitMIUSA
| | - Dahlia Sano
- Department of OncologyKarmanos Cancer InstituteWSU SOMDetroitMIUSA
| | - Sayf Al‐Katib
- Department of Diagnostic Radiology and Molecular ImagingBeaumont HealthOakland University William Beaumont School of MedicineRoyal OakMIUSA
| | - Michelle Bonnett
- Department of PathologyAscension St. John Hospital and Medical CenterDetroitMIUSA
| | - Zyad Kafri
- Van Elslander Cancer CenterGrosse Pointe WoodsMIUSA
| |
Collapse
|
44
|
Higashioka K, Kikushige Y, Ayano M, Kimoto Y, Mitoma H, Kikukawa M, Akahoshi M, Arinobu Y, Horiuchi T, Akashi K, Niiro H. Generation of a novel CD30 + B cell subset producing GM-CSF and its possible link to the pathogenesis of systemic sclerosis. Clin Exp Immunol 2020; 201:233-243. [PMID: 32538493 PMCID: PMC7419935 DOI: 10.1111/cei.13477] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 05/26/2020] [Accepted: 06/06/2020] [Indexed: 12/12/2022] Open
Abstract
Systemic sclerosis (SSc) is a T helper type 2 (Th2)-associated autoimmune disease characterized by vasculopathy and fibrosis. Efficacy of B cell depletion therapy underscores antibody-independent functions of B cells in SSc. A recent study showed that the Th2 cytokine interleukin (IL)-4 induces granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing effector B cells (GM-Beffs ) in humans. In this study, we sought to elucidate the generation mechanism of GM-Beffs and also determine a role of this subset in SSc. Among Th-associated cytokines, IL-4 most significantly facilitated the generation of GM-Beffs within memory B cells in healthy controls (HCs). In addition, the profibrotic cytokine transforming growth factor (TGF)-β further potentiated IL-4- and IL-13-induced GM-Beffs . Of note, tofacitinib, a Janus kinase (JAK) inhibitor, inhibited the expression of GM-CSF mRNA and protein in memory B cells induced by IL-4, but not by TGF-β. GM-Beffs were enriched within CD20+ CD30+ CD38-/low cells, a distinct population from plasmablasts, suggesting that GM-Beffs exert antibody-independent functions. GM-Beffs were also enriched in a CD30+ fraction of freshly isolated B cells. GM-Beffs generated under Th2 conditions facilitated the differentiation from CD14+ monocytes to DC-SIGN+ CD1a+ CD14- CD86+ cells, which significantly promoted the proliferation of naive T cells. CD30+ GM-Beffs were more pronounced in patients with SSc than in HCs. A subpopulation of SSc patients with the diffuse type and concomitant interstitial lung disease exhibited high numbers of GM-Beffs . Together, these findings suggest that human GM-Beffs are enriched in a CD30+ B cell subset and play a role in the pathogenesis of SSc.
Collapse
Affiliation(s)
- K. Higashioka
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Y. Kikushige
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - M. Ayano
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Y. Kimoto
- Department of Internal MedicineKyushu University Beppu HospitalTsurumiharaBeppuOitaJapan
| | - H. Mitoma
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - M. Kikukawa
- Department of Medical EducationFaculty of Medical SciencesKyushu UniversityFukuokaJapan
| | - M. Akahoshi
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - Y. Arinobu
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - T. Horiuchi
- Department of Internal MedicineKyushu University Beppu HospitalTsurumiharaBeppuOitaJapan
| | - K. Akashi
- Department of Medicine and Biosystemic ScienceGraduate School of Medical SciencesKyushu UniversityFukuokaJapan
| | - H. Niiro
- Department of Medical EducationFaculty of Medical SciencesKyushu UniversityFukuokaJapan
| |
Collapse
|
45
|
Chandrasekhar JL, Cox KM, Erickson LD. B Cell Responses in the Development of Mammalian Meat Allergy. Front Immunol 2020; 11:1532. [PMID: 32765532 PMCID: PMC7379154 DOI: 10.3389/fimmu.2020.01532] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2020] [Accepted: 06/10/2020] [Indexed: 12/11/2022] Open
Abstract
Studies of meat allergic patients have shown that eating meat poses a serious acute health risk that can induce severe cutaneous, gastrointestinal, and respiratory reactions. Allergic reactions in affected individuals following meat consumption are mediated predominantly by IgE antibodies specific for galactose-α-1,3-galactose (α-gal), a blood group antigen of non-primate mammals and therefore present in dietary meat. α-gal is also found within certain tick species and tick bites are strongly linked to meat allergy. Thus, it is thought that exposure to tick bites promotes cutaneous sensitization to tick antigens such as α-gal, leading to the development of IgE-mediated meat allergy. The underlying immune mechanisms by which skin exposure to ticks leads to the production of α-gal-specific IgE are poorly understood and are key to identifying novel treatments for this disease. In this review, we summarize the evidence of cutaneous exposure to tick bites and the development of mammalian meat allergy. We then provide recent insights into the role of B cells in IgE production in human patients with mammalian meat allergy and in a novel mouse model of meat allergy. Finally, we discuss existing data more generally focused on tick-mediated immunomodulation, and highlight possible mechanisms for how cutaneous exposure to tick bites might affect B cell responses in the skin and gut that contribute to loss of oral tolerance.
Collapse
Affiliation(s)
- Jessica L Chandrasekhar
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Kelly M Cox
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| | - Loren D Erickson
- Beirne B. Carter Center for Immunology Research, University of Virginia School of Medicine, Charlottesville, VA, United States.,Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, United States
| |
Collapse
|
46
|
Waltari E, Carabajal E, Sanyal M, Friedland N, McCutcheon KM. Adaption of a conventional ELISA to a 96-well ELISA-Array for measuring the antibody responses to influenza virus proteins and vaccines. J Immunol Methods 2020; 481-482:112789. [PMID: 32380014 DOI: 10.1016/j.jim.2020.112789] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Accepted: 04/12/2020] [Indexed: 12/18/2022]
Abstract
We describe an adaptation of conventional ELISA methods to an ELISA-Array format using non-contact Piezo printing of up to 30 spots of purified recombinant viral fusion proteins and vaccine on 96 well high-protein binding plates. Antigens were printed in 1 nanoliter volumes of protein stabilizing buffer using as little as 0.25 nanograms of protein, 2000-fold less than conventional ELISA. The performance of the ELISA-Array was demonstrated by serially diluting n = 9 human post-flu vaccination plasma samples starting at a 1/1000 dilution and measuring binding to the array of Influenza antigens. Plasma polyclonal antibody levels were detected using a cocktail of biotinylated anti-human kappa and lambda light chain antibodies, followed by a Streptavidin-horseradish peroxidase conjugate and the dose-dependent signal was developed with a precipitable TMB substrate. Intra- and inter-assay precision of absorbance units among the eight donor samples showed mean CVs of 4.8% and 10.8%, respectively. The plasma could be differentiated by donor and antigen with titer sensitivities ranging from 1 × 103 to 4 × 106, IC50 values from 1 × 104 to 9 × 106, and monoclonal antibody sensitivities in the ng/mL range. Equivalent sensitivities of ELISA versus ELISA-Array, compared using plasma and an H1N1 HA trimer, were achieved on the ELISA-Array printed at 0.25 ng per 200um spot and 1000 ng per ELISA 96-well. Vacuum-sealed array plates were shown to be stable when stored for at least 2 days at ambient temperature and up to 1 month at 4-8 °C. By the use of any set of printed antigens and analyte matrices the methods of this multiplexed ELISA-Array format can be broadly applied in translational research.
Collapse
Affiliation(s)
| | | | - Mrinmoy Sanyal
- Stanford ChEM-H and Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Natalia Friedland
- Stanford ChEM-H and Department of Biochemistry, Stanford University School of Medicine, Stanford, CA, USA
| | | |
Collapse
|
47
|
Waickman AT, Gromowski GD, Rutvisuttinunt W, Li T, Siegfried H, Victor K, Kuklis C, Gomootsukavadee M, McCracken MK, Gabriel B, Mathew A, Grinyo I Escuer A, Fouch ME, Liang J, Fernandez S, Davidson E, Doranz BJ, Srikiatkhachorn A, Endy T, Thomas SJ, Ellison D, Rothman AL, Jarman RG, Currier JR, Friberg H. Transcriptional and clonal characterization of B cell plasmablast diversity following primary and secondary natural DENV infection. EBioMedicine 2020; 54:102733. [PMID: 32315970 PMCID: PMC7170960 DOI: 10.1016/j.ebiom.2020.102733] [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: 01/02/2020] [Revised: 02/19/2020] [Accepted: 03/10/2020] [Indexed: 01/06/2023] Open
Abstract
Antibody-mediated humoral immunity is thought to play a central role in mediating the immunopathogenesis of acute DENV infection, but limited data are available on the diversity, specificity, and functionality of the antibody response at the molecular level elicited by primary or secondary DENV infection. In order to close this functional gap in our understanding of DENV-specific humoral immunity, we utilized high-throughput single cell RNA sequencing to investigate B cells circulating in both primary and secondary natural DENV infections. We captured full-length paired immunoglobulin receptor sequence data from 9,027 B cells from a total of 6 subjects, including 2,717 plasmablasts. In addition to IgG and IgM class-switched cells, we unexpectedly found a high proportion of the DENV-elicited plasmablasts expressing IgA, principally in individuals with primary DENV infections. These IgA class-switched cells were extensively hypermutated even in individuals with a serologically confirmed primary DENV infection. Utilizing a combination of conventional biochemical assays and high-throughput shotgun mutagenesis, we determined that DENV-reactive IgA class-switched antibodies represent a significant fraction of DENV-reactive Igs generated in response to DENV infection, and that they exhibit a comparable epitope specificity to DENV-reactive IgG antibodies. These results provide insight into the molecular-level diversity of DENV-elicited humoral immunity and identify a heretofore unappreciated IgA plasmablast response to DENV infection.
Collapse
Affiliation(s)
- Adam T Waickman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States.
| | - Gregory D Gromowski
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Wiriya Rutvisuttinunt
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Tao Li
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Hayden Siegfried
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Kaitlin Victor
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Caitlin Kuklis
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Methee Gomootsukavadee
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | - Michael K McCracken
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Benjamin Gabriel
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Anuja Mathew
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | | | | | - Jenny Liang
- Integral Molecular, Philadelphia, PA, United States
| | - Stefan Fernandez
- Department of Virology, Armed Forces Research Institute of Medical Sciences, Bangkok, Thailand
| | | | | | - Anon Srikiatkhachorn
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States; Faculty of Medicine, King Mongkut's Institute of Technology Ladkrabang, Bangkok, Thailand
| | - Timothy Endy
- Department of Microbiology and Immunology, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Stephen J Thomas
- Institute for Global Health and Translational Sciences, State University of New York Upstate Medical University, Syracuse, New York, USA
| | - Damon Ellison
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Alan L Rothman
- Department of Cell and Molecular Biology, Institute for Immunology and Informatics, University of Rhode Island, Providence, RI, United States
| | - Richard G Jarman
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Jeffrey R Currier
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| | - Heather Friberg
- Viral Diseases Branch, Walter Reed Army Institute of Research, Silver Spring, MD, United States
| |
Collapse
|
48
|
López-Puertollano D, Agulló C, Mercader JV, Abad-Somovilla A, Abad-Fuentes A. Click Chemistry-Assisted Bioconjugates for Hapten Immunodiagnostics. Bioconjug Chem 2020; 31:956-964. [DOI: 10.1021/acs.bioconjchem.0c00099] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
- Daniel López-Puertollano
- Department of Organic Chemistry, Universitat de València, Doctor Moliner 50, 46100 Burjassot, València, Spain
| | - Consuelo Agulló
- Department of Organic Chemistry, Universitat de València, Doctor Moliner 50, 46100 Burjassot, València, Spain
| | - Josep V. Mercader
- Institute of Agrochemistry and Food Technology (IATA), Spanish National Research Council (CSIC), Agustí Escardino 7, 46980 Paterna, València, Spain
| | - Antonio Abad-Somovilla
- Department of Organic Chemistry, Universitat de València, Doctor Moliner 50, 46100 Burjassot, València, Spain
| | - Antonio Abad-Fuentes
- Institute of Agrochemistry and Food Technology (IATA), Spanish National Research Council (CSIC), Agustí Escardino 7, 46980 Paterna, València, Spain
| |
Collapse
|
49
|
Lerner LK, Nguyen TV, Castro LP, Vilar JB, Munford V, Le Guillou M, Mohammad MM, Vergé V, Rosselli F, Menck CFM, Sarasin A, Aoufouchi S. Large deletions in immunoglobulin genes are associated with a sustained absence of DNA Polymerase η. Sci Rep 2020; 10:1311. [PMID: 31992747 PMCID: PMC6987143 DOI: 10.1038/s41598-020-58180-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Accepted: 01/08/2020] [Indexed: 01/27/2023] Open
Abstract
Somatic hypermutation of immunoglobulin genes is a highly mutagenic process that is B cell-specific and occurs during antigen-driven responses leading to antigen specificity and antibody affinity maturation. Mutations at the Ig locus are initiated by Activation-Induced cytidine Deaminase and are equally distributed at G/C and A/T bases. This requires the establishment of error-prone repair pathways involving the activity of several low fidelity DNA polymerases. In the physiological context, the G/C base pair mutations involve multiple error-prone DNA polymerases, while the generation of mutations at A/T base pairs depends exclusively on the activity of DNA polymerase η. Using two large cohorts of individuals with xeroderma pigmentosum variant (XP-V), we report that the pattern of mutations at Ig genes becomes highly enriched with large deletions. This observation is more striking for patients older than 50 years. We propose that the absence of Pol η allows the recruitment of other DNA polymerases that profoundly affect the Ig genomic landscape.
Collapse
Affiliation(s)
- Leticia K Lerner
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Thuy V Nguyen
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France
- Department of Genetics, Faculty of Biology, University of Science, Ho Chi Minh City, Vietnam
| | - Ligia P Castro
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Juliana B Vilar
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Veridiana Munford
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Morwenna Le Guillou
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France
- Université Paris-Saclay, 91400, Orsay, France
| | - Mahwish Mian Mohammad
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France
- Sorbonne Université, Paris, 75006, France
| | - Véronique Vergé
- Haematology Unit, Gustave Roussy, 94805, Villejuif, France
- Université Paris-Saclay, 91400, Orsay, France
| | - Filippo Rosselli
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France
- Université Paris-Saclay, 91400, Orsay, France
| | - Carlos F M Menck
- Department of Microbiology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil
| | - Alain Sarasin
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France
- Université Paris-Saclay, 91400, Orsay, France
| | - Said Aoufouchi
- Centre National de la Recherche Scientifique UMR8200, Gustave Roussy, 94805, Villejuif, France.
- Université Paris-Saclay, 91400, Orsay, France.
- Sorbonne Université, Paris, 75006, France.
| |
Collapse
|
50
|
Abstract
The importance of B cell and antibody-mediated immune response in the acute and long-term persistence of transplanted solid organs has become increasingly evident in recent years. A variety of therapeutic innovations target antibodies directed toward HLA or blood groups (ABO) to allow better allocation and posttransplant longevity of organs. Antibodies originate from plasma cells (PCs), which are terminally differentiated B cells. Long-term production and persistence of these antibodies is partly due to fast reactivation of previously generated memory B cells; however, there is increasing evidence that some differentiated PCs can persist independently in the bone marrow for years or even decades, producing specific antibodies or even experiencing regeneration without proliferation without need to be replaced by newly differentiating B cells. This review outlines the currently presumed pathways of differentiation, antibody, and memory generation on both B-cell and PC levels. On this background, current therapeutic concepts for antibody reduction before and after solid organ transplantation are considered, to better understand their mechanisms, possible synergisms, and specific risks. Specific differences in regards to ABO versus HLA antibodies as well as practical relevance for generation of desensitization and posttransplant antibody-directed therapy protocols are discussed.
Collapse
|