1
|
Neirinck J, Buysse M, De Vriendt C, Hofmans M, Bonroy C. The role of immunophenotyping in common variable immunodeficiency: a narrative review. Crit Rev Clin Lab Sci 2024:1-20. [PMID: 39364936 DOI: 10.1080/10408363.2024.2404842] [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: 06/24/2024] [Revised: 08/06/2024] [Accepted: 09/12/2024] [Indexed: 10/05/2024]
Abstract
Common variable immunodeficiency (CVID) is a heterogeneous primary immunodeficiency (PID) characterized by an impaired immunoglobulin production, in association with an increased susceptibility to infections and a diversity of clinical manifestations. This narrative review summarizes immunophenotypic abnormalities in CVID patients and their relevance for diagnosis and disease classification. A comprehensive search across four databases - PubMED, Web of Science, EMBASE and Google Scholar - yielded 170 relevant studies published between 1988 and April 31, 2023. Over the past decades, the role of immunophenotyping in CVID diagnosis has become evident by identifying "hallmark" immunophenotypic aberrancies in patient subsets, with some now integrated in the consensus diagnostic criteria. Furthermore, the role of immunophenotyping in subclassifying CVID in relation to clinical presentation and prognosis has been extensively studied. Certain immunophenotypic patterns consistently correlate with clinical manifestations and/or subsets of CVID, particularly those associated with noninfectious complications (i.e. low switched memory B cells, shifts in follicular helper T cell subsets, low naïve CD4+ T cells, low regulatory T cells, and expansion of CD21low B cells, often associated with autoimmunity and/or splenomegaly). Also, efforts to associate subset levels of innate immune cells, such as Natural Killer (NK) cells, invariant (i)NKT cells, innate lymphoid cells (ILCs), and dendritic cells (DCs) to CVID complications are evident albeit in a lesser degree. However, inconsistencies regarding the role of flow cytometry in classification and prognosis persist, reflecting the disease complexity, but probably also cohort variations and methodological differences between published studies. This underscores the need for collaborative efforts to integrate emerging concepts, such as standardized flow cytometry and computational tools, for a more precise CVID classification approach. Additionally, recent studies suggest a potential value of (epi)genetic-based molecular assays to this effort.
Collapse
Affiliation(s)
- Jana Neirinck
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Malicorne Buysse
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Ciel De Vriendt
- Department of Haematology, University Hospital Ghent, Ghent, Belgium
| | - Mattias Hofmans
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
2
|
Fryer HA, Geers D, Gommers L, Zaeck LM, Tan NH, Jones-Freeman B, Goorhuis A, Postma DF, Visser LG, Hogarth PM, Koopmans MPG, GeurtsvanKessel CH, O'Hehir RE, van der Kuy PHM, de Vries RD, van Zelm MC. Fourth dose bivalent COVID-19 vaccines outperform monovalent boosters in eliciting cross-reactive memory B cells to Omicron subvariants. J Infect 2024; 89:106246. [PMID: 39127451 DOI: 10.1016/j.jinf.2024.106246] [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: 05/15/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Bivalent COVID-19 vaccines comprising ancestral Wuhan-Hu-1 (WH1) and the Omicron BA.1 or BA.5 subvariant elicit enhanced serum antibody responses to emerging Omicron subvariants. Here, we characterized the RBD-specific memory B cell (Bmem) response following a fourth dose with a BA.1 or BA.5 bivalent vaccine, in direct comparison with a WH1 monovalent fourth dose. Healthcare workers previously immunized with mRNA or adenoviral vector monovalent vaccines were sampled before and one month after a fourth dose with a monovalent or a BA.1 or BA.5 bivalent vaccine. Serum neutralizing antibodies (NAb) were quantified, as well as RBD-specific Bmem with an in-depth spectral flow cytometry panel including recombinant RBD proteins of the WH1, BA.1, BA.5, BQ.1.1, and XBB.1.5 variants. Both bivalent vaccines elicited higher NAb titers against Omicron subvariants compared to the monovalent vaccine. Following either vaccine type, recipients had slightly increased WH1 RBD-specific Bmem numbers. Both bivalent vaccines significantly increased WH1 RBD-specific Bmem binding of all Omicron subvariants tested by flow cytometry, while recognition of Omicron subvariants was not enhanced following monovalent vaccination. IgG1+ Bmem dominated the response, with substantial IgG4+ Bmem only detected in recipients of an mRNA vaccine for their primary dose. Thus, Omicron-based bivalent vaccines can significantly boost NAb and Bmem specific for ancestral WH1 and Omicron variants and improve recognition of descendent subvariants by pre-existing, WH1-specific Bmem beyond that of a monovalent vaccine. This provides new insights into the capacity of variant-based mRNA booster vaccines to improve immune memory against emerging SARS-CoV-2 variants and potentially protect against severe disease. ONE-SENTENCE SUMMARY: Omicron BA.1 and BA.5 bivalent COVID-19 boosters, used as a fourth dose, increase RBD-specific Bmem cross-recognition of Omicron subvariants, both those encoded by the vaccines and antigenically distinct subvariants, further than a monovalent booster.
Collapse
Affiliation(s)
- Holly A Fryer
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Daryl Geers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Lennert Gommers
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Luca M Zaeck
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Ngoc H Tan
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Bernadette Jones-Freeman
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia
| | - Abraham Goorhuis
- Center of Tropical Medicine and Travel Medicine, Department of Infectious Diseases, Amsterdam University Medical Centers, Amsterdam, the Netherlands; Infection and Immunity, Amsterdam Public Health, University of Amsterdam, Amsterdam, the Netherlands
| | - Douwe F Postma
- Department of Internal Medicine and Infectious Diseases, University Medical Center Groningen, Groningen, the Netherlands
| | - Leo G Visser
- Department of Infectious Diseases, Leiden University Medical Center, Leiden, Netherlands
| | - P Mark Hogarth
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Immune Therapies Group, Burnet Institute, Melbourne, Victoria, Australia
| | - Marion P G Koopmans
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | | | - Robyn E O'Hehir
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia
| | - P Hugo M van der Kuy
- Dept. Hospital Pharmacy, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Rory D de Vries
- Dept. Viroscience, Erasmus MC, University Medical Center, Rotterdam, the Netherlands
| | - Menno C van Zelm
- Dept. Immunology, School of Translational Medicine, Monash University, Melbourne, Victoria, Australia; Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, Victoria, Australia; Dept. Immunology, Erasmus MC, University Medical Center, Rotterdam, the Netherlands.
| |
Collapse
|
3
|
Hartley GE, Fryer HA, Gill PA, Boo I, Bornheimer SJ, Hogarth PM, Drummer HE, O'Hehir RE, Edwards ESJ, van Zelm MC. Homologous but not heterologous COVID-19 vaccine booster elicits IgG4+ B-cells and enhanced Omicron subvariant binding. NPJ Vaccines 2024; 9:129. [PMID: 39013889 PMCID: PMC11252355 DOI: 10.1038/s41541-024-00919-8] [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: 10/25/2023] [Accepted: 06/27/2024] [Indexed: 07/18/2024] Open
Abstract
Booster vaccinations are recommended to improve protection against severe disease from SARS-CoV-2 infection. With primary vaccinations involving various adenoviral vector and mRNA-based formulations, it remains unclear if these differentially affect the immune response to booster doses. We examined the effects of homologous (mRNA/mRNA) and heterologous (adenoviral vector/mRNA) vaccination on antibody and memory B cell (Bmem) responses against ancestral and Omicron subvariants. Healthy adults who received primary BNT162b2 (mRNA) or ChAdOx1 (vector) vaccination were sampled 1-month and 6-months after their 2nd and 3rd dose (homologous or heterologous) vaccination. Recombinant spike receptor-binding domain (RBD) proteins from ancestral, Omicron BA.2 and BA.5 variants were produced for ELISA-based serology, and tetramerized for immunophenotyping of RBD-specific Bmem. Dose 3 boosters significantly increased ancestral RBD-specific plasma IgG and Bmem in both cohorts. Up to 80% of ancestral RBD-specific Bmem expressed IgG1+. IgG4+ Bmem were detectable after primary mRNA vaccination, and expanded significantly to 5-20% after dose 3, whereas heterologous boosting did not elicit IgG4+ Bmem. Recognition of Omicron BA.2 and BA.5 by ancestral RBD-specific plasma IgG increased from 20% to 60% after the 3rd dose in both cohorts. Reactivity of ancestral RBD-specific Bmem to Omicron BA.2 and BA.5 increased following a homologous booster from 40% to 60%, but not after a heterologous booster. A 3rd mRNA dose generates similarly robust serological and Bmem responses in homologous and heterologous vaccination groups. The expansion of IgG4+ Bmem after mRNA priming might result from the unique vaccine formulation or dosing schedule affecting the Bmem response duration and antibody maturation.
Collapse
Affiliation(s)
- Gemma E Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Holly A Fryer
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Paul A Gill
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Irene Boo
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
| | | | - P Mark Hogarth
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Heidi E Drummer
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Robyn E O'Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| | - Emily S J Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, School of Translational Medicine, Monash University, Melbourne, VIC, Australia.
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia.
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
4
|
Materne E, Zhou B, DiGiacomo D, Farmer JR, Fuleihan R, Sullivan KE, Cunningham-Rundles C, Ballas ZK, Suez D, Barmettler S. Renal complications in patients with predominantly antibody deficiency in the United States Immune Deficiency Network (USIDNET). J Allergy Clin Immunol 2024; 154:237-242.e1. [PMID: 38555979 DOI: 10.1016/j.jaci.2024.03.014] [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: 12/16/2023] [Revised: 03/03/2024] [Accepted: 03/18/2024] [Indexed: 04/02/2024]
Abstract
BACKGROUND Prior studies have reported that renal insufficiency occurs in a small percentage of patients with predominantly antibody deficiency (PAD) and in about 2% of patients with common variable immunodeficiency. OBJECTIVE The goal of our study was to understand and evaluate the prevalence and type of renal complications in patients with PAD in the United States Immunodeficiency Network (USIDNET) cohort. We hypothesized that there is an association between certain renal complications and severity of immunophenotype in patients with PAD. METHODS We performed a query of patients with PAD from the USIDNET cohort with renal complications. Patients with documented renal disease such as chronic kidney disease (CKD), nephrolithiasis, nephritis, and renal failure syndrome were included. We compared immunophenotype, flow cytometry findings, and immunoglobulin levels of patients with PAD accompanied by renal complications with those of the total USIDNET cohort of patients with PAD. RESULTS We determined that 140 of 2071 patients with PAD (6.8%) had renal complications. Of these 140 patients, 50 (35.7%) had CKD, 46 (32.9%) had nephrolithiasis, 18 (12.9 %) had nephritis, and 50 (35.7%) had other renal complications. Compared with the total USIDNET cohort of patients with PAD, patients with CKD had lower absolute lymphocyte counts, CD3+ T-cell counts, CD4+ T-cell counts, CD19+ B-cell counts, CD20+ B-cell counts, and CD27+IgD- B-cell counts (P < .05 for all). Patients with nephritis had lower absolute lymphocyte counts, CD19+ B-cell counts, CD27+ B-cell counts, and IgE levels (P < .05 for all) than patients with PAD without renal disease. CONCLUSIONS We determined that 6.8% of the USIDNET cohort of patients with PAD had a documented renal complication. Compared with the overall cohort of patients with PAD, those patients with nephritis and CKD had a more severe immunophenotype.
Collapse
Affiliation(s)
- Emma Materne
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass.
| | - Baijun Zhou
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass
| | - Daniel DiGiacomo
- Hackensack Meridian Health, Jersey Shore University Medical Center, Neptune, NJ
| | - Jocelyn R Farmer
- Clinical Immunodeficiency Program, Division of Allergy and Inflammation, Beth Israel Lahey Health, Burlington, Mass
| | - Ramsay Fuleihan
- Division of Pediatric Allergy, Immunology and Rheumatology, Columbia University, New York, NY
| | - Kathleen E Sullivan
- Division of Allergy and Immunology, Children's Hospital of Pennsylvania, Philadelphia, Pa
| | | | - Zuhair K Ballas
- Division of Internal Medicine, Immunology, University of Iowa, Iowa City, Iowa
| | | | - Sara Barmettler
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Boston, Mass; Harvard Medical School, Boston, Mass
| |
Collapse
|
5
|
Chang-Rabley E, van Zelm MC, Ricotta EE, Edwards ESJ. An Overview of the Strategies to Boost SARS-CoV-2-Specific Immunity in People with Inborn Errors of Immunity. Vaccines (Basel) 2024; 12:675. [PMID: 38932404 PMCID: PMC11209597 DOI: 10.3390/vaccines12060675] [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: 05/03/2024] [Revised: 06/09/2024] [Accepted: 06/12/2024] [Indexed: 06/28/2024] Open
Abstract
The SARS-CoV-2 pandemic has heightened concerns about immunological protection, especially for individuals with inborn errors of immunity (IEI). While COVID-19 vaccines elicit strong immune responses in healthy individuals, their effectiveness in IEI patients remains unclear, particularly against new viral variants and vaccine formulations. This uncertainty has led to anxiety, prolonged self-isolation, and repeated vaccinations with uncertain benefits among IEI patients. Despite some level of immune response from vaccination, the definition of protective immunity in IEI individuals is still unknown. Given their susceptibility to severe COVID-19, strategies such as immunoglobulin replacement therapy (IgRT) and monoclonal antibodies have been employed to provide passive immunity, and protection against both current and emerging variants. This review examines the efficacy of COVID-19 vaccines and antibody-based therapies in IEI patients, their capacity to recognize viral variants, and the necessary advances required for the ongoing protection of people with IEIs.
Collapse
Affiliation(s)
- Emma Chang-Rabley
- The Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| | - Menno C. van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3800, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC 3000, Australia
- Department of Immunology, Erasmus MC, University Medical Center, 3015 GD Rotterdam, The Netherlands
| | - Emily E. Ricotta
- The Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
- Department of Preventive Medicine and Biostatistics, Uniform Services University of the Health Sciences, Bethesda, MD 20814, USA
| | - Emily S. J. Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC 3800, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies in Melbourne, Melbourne, VIC 3000, Australia
| |
Collapse
|
6
|
Ramirez NJ, Schulze JJ, Walter S, Werner J, Mrovecova P, Olek S, Sachsenmaier C, Grimbacher B, Salzer U. Epigenetic immune cell quantification for diagnostic evaluation and monitoring of patients with inborn errors of immunity and secondary immune deficiencies. Clin Immunol 2024; 260:109920. [PMID: 38307474 DOI: 10.1016/j.clim.2024.109920] [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: 06/08/2023] [Revised: 01/30/2024] [Accepted: 01/31/2024] [Indexed: 02/04/2024]
Abstract
BACKGROUND Early detection and monitoring of primary immunodeficiencies (PID) in humans require quantitative determination of immune cells from fresh blood analyzed by flow cytometry. However, epigenetic immune cell quantification allows analysis from fresh, frozen, or dried blood samples. We demonstrate the utility of epigenetic immune cell quantification for patients with PID. METHODS Epigenetic quantification of basic lymphocyte subpopulations of 259 samples from PID patients were compared to flow cytometric data. Epigenetic analysis was extended to T-cell subsets (Treg, Th17, Tfh, PD-1+, CCR6+) and memory B-cells and compared between venous EDTA and dried blood. RESULTS A high correlation of >0.9 was observed for basic T- and B-cell subsets. Extended epigenetic analysis showed quantitative trends within PID subgroups, but individually these varied substantially within these groups. Epigenetic analysis of dried blood samples was equivalent to EDTA blood. CONCLUSION Epigenetic immune cell quantification is suitable for immune cell profiling in PID patients.
Collapse
Affiliation(s)
- Neftali J Ramirez
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | | | | | | | - Pavla Mrovecova
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sven Olek
- Ivana Turbachova Laboratory for Epigenetics, Precision for Medicine GmbH, Berlin, Germany
| | | | - Bodo Grimbacher
- Institute for Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Clinic of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany; DZIF - German Center for Infection Research, Satellite Center Freiburg, Germany; CIBSS - Centre for Integrative Biological Signalling Studies, Albert-Ludwigs University, Freiburg, Germany; RESIST - Cluster of Excellence 2155 to Hanover Medical School, Satellite Center Freiburg, Germany.
| | - Ulrich Salzer
- Center for Chronic Immunodeficiency (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Clinic of Rheumatology and Clinical Immunology, Center for Chronic Immunodeficiency (CCI), Medical Center, Faculty of Medicine, Albert-Ludwigs-University of Freiburg, Germany.
| |
Collapse
|
7
|
Torres-Valle A, Aragon L, Silva SL, Serrano C, Marcos M, Melero J, Bonroy C, Arenas-Caro PP, Casado DM, Olaizola PMR, Neirinck J, Hofmans M, de Arriba S, Jara M, Prieto C, Sousa AE, Prada Á, van Dongen JJM, Pérez-Andrés M, Orfao A. In-depth blood immune profiling of Good syndrome patients. Front Immunol 2023; 14:1285088. [PMID: 38035080 PMCID: PMC10684950 DOI: 10.3389/fimmu.2023.1285088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 10/23/2023] [Indexed: 12/02/2023] Open
Abstract
Introduction Good syndrome (GS) is a rare adult-onset immunodeficiency first described in 1954. It is characterized by the coexistence of a thymoma and hypogammaglobulinemia, associated with an increased susceptibility to infections and autoimmunity. The classification and management of GS has been long hampered by the lack of data about the underlying immune alterations, a controversy existing on whether it is a unique diagnostic entity vs. a subtype of Common Variable Immune Deficiency (CVID). Methods Here, we used high-sensitive flow cytometry to investigate the distribution of up to 70 different immune cell populations in blood of GS patients (n=9) compared to age-matched CVID patients (n=55) and healthy donors (n=61). Results All 9 GS patients displayed reduced B-cell counts -down to undetectable levels (<0.1 cells/μL) in 8/9 cases-, together with decreased numbers of total CD4+ T-cells, NK-cells, neutrophils, and basophils vs. age-matched healthy donors. In contrast, they showed expanded TCRγδ+ T-cells (p ≤ 0.05). Except for a deeper B-cell defect, the pattern of immune cell alteration in blood was similar in GS and (age-matched) CVID patients. In depth analysis of CD4+ T-cells revealed significantly decreased blood counts of naïve, central memory (CM) and transitional memory (TM) TCD4+ cells and their functional compartments of T follicular helper (TFH), regulatory T cells (Tregs), T helper (Th)2, Th17, Th22, Th1/Th17 and Th1/Th2 cells. In addition, GS patients also showed decreased NK-cell, neutrophil, basophil, classical monocyte and of both CD1c+ and CD141+ myeloid dendritic cell counts in blood, in parallel to an expansion of total and terminal effector TCRγδ+ T-cells. Interestingly, those GS patients who developed hypogammaglobulinemia several years after the thymoma presented with an immunological and clinical phenotype which more closely resembled a combined immune humoral and cellular defect, with poorer response to immunoglobulin replacement therapy, as compared to those in whom the thymoma and hypogammaglobulinemia were simultaneously detected. Discussion Our findings provide a more accurate definition of the immune cell defects of GS patients and contribute to a better discrimination among GS patients between those with a pure B-cell defect vs. those suffering from a combined immunodeficiency with important consequences on the diagnosis and management of the disease.
Collapse
Affiliation(s)
- Alba Torres-Valle
- Translational and Clinical Research Program, Centro de investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
| | - Larraitz Aragon
- Immunology Department, Donostia University Hospital, Osakidetza Basque Health Service, San Sebastián, Spain
| | - Susana L. Silva
- Serviço de Imunoalergologia, Centro Hospitalar Universitário Lisboa Norte, Lisbon, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | | | - Miguel Marcos
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Internal Medicine, University Hospital of Salamanca, Salamanca, Spain
- Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Josefa Melero
- Servicio de inmunología y genética, Hospital Universitario de Badajoz, Badajoz, Spain
| | - Carolien Bonroy
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Pedro Pablo Arenas-Caro
- Immunology Department, Donostia University Hospital, Osakidetza Basque Health Service, San Sebastián, Spain
| | - David Monzon Casado
- Immunology Department, Donostia University Hospital, Osakidetza Basque Health Service, San Sebastián, Spain
| | | | - Jana Neirinck
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Mattias Hofmans
- Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium
| | - Sonia de Arriba
- Pediatrics Department, University Hospital of Salamanca, Salamanca, Spain
| | - María Jara
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
- DNA Sequencing Service (NUCLEUS), University of Salamanca, Salamanca, Spain
| | - Carlos Prieto
- Bioinformatics service (NUCLEUS), University of Salamanca, Salamanca, Spain
| | - Ana E. Sousa
- Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Álvaro Prada
- Immunology Department, Donostia University Hospital, Osakidetza Basque Health Service, San Sebastián, Spain
| | - Jacques J. M. van Dongen
- Translational and Clinical Research Program, Centro de investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, Salamanca, Spain
- Department of Immunology, Leiden University Medical Center, Leiden, Netherlands
| | - Martín Pérez-Andrés
- Translational and Clinical Research Program, Centro de investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| | - Alberto Orfao
- Translational and Clinical Research Program, Centro de investigación del Cáncer (CIC), Instituto de Biología Molecular y Celular del Cáncer (IBMCC), Consejo Superior de Investigaciones Científicas (CSIC) and University of Salamanca (USAL), Salamanca, Spain
- Cytometry Service, NUCLEUS, Department of Medicine, University of Salamanca, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Biomedical Research Networking Centre Consortium of Oncology (CIBERONC), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
8
|
LaBere B, Nguyen AA, Habiballah SB, Elkins M, Imperial J, Li B, Devana S, Timilsina S, Stubbs SB, Joerger J, Chou J, Platt CD. Clinical utility of measuring CD4 + T follicular cells in patients with immune dysregulation. J Autoimmun 2023; 140:103088. [PMID: 37549449 PMCID: PMC10839119 DOI: 10.1016/j.jaut.2023.103088] [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: 05/25/2023] [Revised: 06/23/2023] [Accepted: 07/11/2023] [Indexed: 08/09/2023]
Abstract
Mechanistic studies of autoimmune disorders have identified circulating T follicular helper (cTfh) cells as drivers of autoimmunity. However, the quantification of cTfh cells is not yet used in clinical practice due to the lack of age-stratified normal ranges and the unknown sensitivity and specificity of this test for autoimmunity. We enrolled 238 healthy participants and 130 patients with common and rare disorders of autoimmunity or autoinflammation. Patients with infections, active malignancy, or any history of transplantation were excluded. In 238 healthy controls, median cTfh percentages (range 4.8%-6.2%) were comparable among age groups, sexes, races, and ethnicities, apart from a significantly lower percentages in children less than 1 year of age (median 2.1%, CI: 0.4%-6.8, p < 0.0001). Among 130 patients with over 40 immune regulatory disorders, a cTfh percentage exceeding 12% had 88% sensitivity and 94% specificity for differentiating disorders with adaptive immune cell dysregulation from those with predominantly innate cell defects. This threshold had a sensitivity of 86% and specificity of 100% for active autoimmunity and normalized with effective treatment. cTfh percentages exceeding 12% distinguish autoimmunity from autoinflammation, thereby differentiating two endotypes of immune dysregulation with overlapping symptoms and different therapies.
Collapse
Affiliation(s)
- Brenna LaBere
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Alan A Nguyen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Saddiq B Habiballah
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Pediatrics, Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Megan Elkins
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Juliet Imperial
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Betty Li
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | | | - Suraj Timilsina
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Spencer B Stubbs
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jill Joerger
- Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Janet Chou
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA
| | - Craig D Platt
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Department of Laboratory Medicine, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
9
|
Fryer HA, Hartley GE, Edwards ESJ, Varese N, Boo I, Bornheimer SJ, Hogarth PM, Drummer HE, O'Hehir RE, van Zelm MC. COVID-19 Adenoviral Vector Vaccination Elicits a Robust Memory B Cell Response with the Capacity to Recognize Omicron BA.2 and BA.5 Variants. J Clin Immunol 2023; 43:1506-1518. [PMID: 37322095 PMCID: PMC10499924 DOI: 10.1007/s10875-023-01527-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Accepted: 05/27/2023] [Indexed: 06/17/2023]
Abstract
Following the COVID-19 pandemic, novel vaccines have successfully reduced severe disease and death. Despite eliciting lower antibody responses, adenoviral vector vaccines are nearly as effective as mRNA vaccines. Therefore, protection against severe disease may be mediated by immune memory cells. We here evaluated plasma antibody and memory B cells (Bmem) targeting the SARS-CoV-2 Spike receptor-binding domain (RBD) elicited by the adenoviral vector vaccine ChAdOx1 (AstraZeneca), their capacity to bind Omicron subvariants, and compared this to the response to mRNA BNT162b2 (Pfizer-BioNTech) vaccination. Whole blood was sampled from 31 healthy adults pre-vaccination and 4 weeks after dose one and dose two of ChAdOx1. Neutralizing antibodies (NAb) against SARS-CoV-2 were quantified at each time point. Recombinant RBDs of the Wuhan-Hu-1 (WH1), Delta, BA.2, and BA.5 variants were produced for ELISA-based quantification of plasma IgG and incorporated separately into fluorescent tetramers for flow cytometric identification of RBD-specific Bmem. NAb and RBD-specific IgG levels were over eight times lower following ChAdOx1 vaccination than BNT162b2. In ChAdOx1-vaccinated individuals, median plasma IgG recognition of BA.2 and BA.5 as a proportion of WH1-specific IgG was 26% and 17%, respectively. All donors generated resting RBD-specific Bmem, which were boosted after the second dose of ChAdOx1 and were similar in number to those produced by BNT162b2. The second dose of ChAdOx1 boosted Bmem that recognized VoC, and 37% and 39% of WH1-specific Bmem recognized BA.2 and BA.5, respectively. These data uncover mechanisms by which ChAdOx1 elicits immune memory to confer effective protection against severe COVID-19.
Collapse
Affiliation(s)
- Holly A Fryer
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Gemma E Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Emily S J Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Nirupama Varese
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
| | - Irene Boo
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
| | | | - P Mark Hogarth
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Heidi E Drummer
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
- Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia
- Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Robyn E O'Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
10
|
Edwards JM, Andrews MC, Burridge H, Smith R, Owens C, Edinger M, Pilkington K, Desfrancois J, Shackleton M, Senthi S, van Zelm MC. Design, optimisation and standardisation of a high-dimensional spectral flow cytometry workflow assessing T-cell immunophenotype in patients with melanoma. Clin Transl Immunology 2023; 12:e1466. [PMID: 37692904 PMCID: PMC10484688 DOI: 10.1002/cti2.1466] [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: 03/30/2023] [Revised: 06/26/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023] Open
Abstract
Objectives Despite the success of immune checkpoint blockade, most metastatic melanoma patients fail to respond to therapy or experience severe toxicity. Assessment of biomarkers and immunophenotypes before or early into treatment will help to understand favourable responses and improve therapeutic outcomes. Methods We present a high-dimensional approach for blood T-cell profiling using three multi-parameter cytometry panels: (1) a TruCount panel for absolute cell counts, (2) a 27-colour spectral panel assessing T-cell markers and (3) a 20-colour spectral panel evaluating intracellular cytokine expression. Pre-treatment blood mononuclear cells from patients and healthy controls were cryopreserved before staining across 11 batches. Batch effects were tracked using a single-donor control and the suitability of normalisation was assessed. The data were analysed using manual gating and high-dimensional strategies. Results Batch-to-batch variation was minimal, as demonstrated by the dimensionality reduction of batch-control samples, and normalisation did not improve manual or high-dimensional analysis. Application of the workflow demonstrated the capacity of the panels and showed that patients had fewer lymphocytes than controls (P = 0.0027), due to lower naive CD4+ (P = 0.015) and CD8+ (P = 0.011) T cells and follicular helper T cells (P = 0.00076). Patients showed trends for higher proportions of Ki67 and IL-2-expressing cells within CD4+ and CD8+ memory subsets, and increased CD57 and EOMES expression within TCRγδ+ T cells. Conclusion Our optimised high-parameter spectral cytometry approach provided in-depth profiling of blood T cells and found differences in patient immunophenotype at baseline. The robustness of our workflow, as demonstrated by minimal batch effects, makes this approach highly suitable for the longitudinal evaluation of immunotherapy effects.
Collapse
Affiliation(s)
- Jack M Edwards
- Alfred Health Radiation OncologyThe Alfred HospitalMelbourneVICAustralia
- Department of Immunology, Central Clinical SchoolMonash University and Alfred HospitalMelbourneVICAustralia
| | - Miles C Andrews
- Department of Medicine, Central Clinical SchoolMonash UniversityMelbourneVICAustralia
- Department of Medical OncologyThe Alfred HospitalMelbourneVICAustralia
| | - Hayley Burridge
- Department of Medical OncologyThe Alfred HospitalMelbourneVICAustralia
| | - Robin Smith
- Alfred Health Radiation OncologyThe Alfred HospitalMelbourneVICAustralia
| | - Carole Owens
- Alfred Health Radiation OncologyThe Alfred HospitalMelbourneVICAustralia
| | | | | | | | - Mark Shackleton
- Department of Medicine, Central Clinical SchoolMonash UniversityMelbourneVICAustralia
- Department of Medical OncologyThe Alfred HospitalMelbourneVICAustralia
| | - Sashendra Senthi
- Alfred Health Radiation OncologyThe Alfred HospitalMelbourneVICAustralia
| | - Menno C van Zelm
- Department of Immunology, Central Clinical SchoolMonash University and Alfred HospitalMelbourneVICAustralia
| |
Collapse
|
11
|
Edwards ESJ, Ojaimi S, Ngui J, Seo GH, Kim J, Chunilal S, Yablonski D, O'Hehir RE, van Zelm MC. Combined immunodeficiency and impaired PI3K signaling in a patient with biallelic LCP2 variants. J Allergy Clin Immunol 2023; 152:807-813.e7. [PMID: 37211057 DOI: 10.1016/j.jaci.2023.04.020] [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: 04/21/2022] [Revised: 04/25/2023] [Accepted: 04/28/2023] [Indexed: 05/23/2023]
Abstract
BACKGROUND Inborn errors affecting components of the T-cell receptor signaling cascade cause combined immunodeficiency with various degrees of severity. Recently, homozygous variants in LCP2 were reported to cause pediatric onset of severe combined immunodeficiency with neutrophil, platelet, and T- and B-cell defects. OBJECTIVE We sought to unravel the genetic cause of combined immunodeficiency and early-onset immune dysregulation in a 26-year-old man who presented with specific antibody deficiency, autoimmunity, and inflammatory bowel disease since early childhood. METHODS The patient was subjected to whole-exome sequencing of genomic DNA and examination of blood neutrophils, platelets, and T and B cells. Expression levels of the Src homology domain 2-containing leukocyte protein of 76 kDa (SLP76) and tonic and ligand-induced PI3K signaling were evaluated by flow-cytometric detection of phosphorylated ribosomal protein S6 in B and T cells. RESULTS Compound heterozygous missense variants were identified in LCP2, affecting the proline-rich repeat domain of SLP76 (p.P190R and p.R204W). The patient's total B- and T-cell numbers were within the normal range, as was platelet function. However, neutrophil function, numbers of unswitched and class-switched memory B cells, and serum IgA were decreased. Moreover, intracellular SLP76 protein levels were reduced in the patient's B cells, CD4+ and CD8+ T cells, and natural killer cells. Tonic and ligand-induced levels of phosphorylated ribosomal protein S6 and ligand-induced phosphorylated PLCγ1 were decreased in the patient's B cells and CD4+ and CD8+ T cells. CONCLUSIONS Biallelic variants in LCP2 impair neutrophil function and T-cell and B-cell antigen-receptor signaling and can cause combined immunodeficiency with early-onset immune dysregulation, even in the absence of platelet defects.
Collapse
Affiliation(s)
- Emily S J Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, Australia; Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia
| | - Samar Ojaimi
- Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Monash Pathology, Monash Health, Melbourne, Australia; Monash Infectious Diseases, Monash Health, Melbourne, Australia; Monash Lung Sleep Allergy Immunology, Monash Health, Melbourne, Australia; Department of Medicine, Southern Clinical School, Monash Health and Monash University, Melbourne, Australia.
| | - James Ngui
- Monash Pathology, Monash Health, Melbourne, Australia
| | - Go Hun Seo
- Division of Medical Genetics, 3billion Inc, Seoul, Korea
| | - JiHye Kim
- Division of Medical Genetics, 3billion Inc, Seoul, Korea
| | - Sanjeev Chunilal
- Department of Pathology and Radiology, Monash Health, Melbourne, Australia
| | - Deborah Yablonski
- Department of Immunology, Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
| | - Robyn E O'Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, Australia; Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, Australia
| | - Menno C van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology, Central Clinical School, Monash University, Melbourne, Australia; Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, Australia; Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Alfred Hospital, Melbourne, Australia.
| |
Collapse
|
12
|
LaBere B, Chu A, Platt CD, Chou J. The Integration of Patient-Reported Quality of Life and Systemic Biomarkers in Patients with Immune Dysregulation. RESEARCH SQUARE 2023:rs.3.rs-3270389. [PMID: 37674702 PMCID: PMC10479437 DOI: 10.21203/rs.3.rs-3270389/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Background Patient-reported quality of life measurements are an important method for improving the treatment of patients with a variety of diseases. These tools have been minimally investigated in patients with inborn errors of immunity (IEI). Patients with IEI may have immune dysregulation and autoimmune-mediated multi-system organ involvement, making treatment optimization vitally important. Routine laboratory and radiologic testing are typically used for treatment monitoring; however, these modalities have the potential to miss early organ damage. T follicular helper cells are T cells that contribute to antibody production and are known to be expanded in patients with active autoimmunity. We hypothesized that a combination of patient-reported quality of life measurements, in addition to T follicular helper cell percentages, would help us to better understand the level of disease activity in patients with IEI and autoimmunity. Methods Patients with immune dysregulation were consented to provide a blood sample and to complete a questionnaire. The Centers for Disease Control HRQOL-14 tool was utilized for the questionnaire portion, and T follicular helper cell levels were measured from whole blood using surface staining and flow cytometry analysis. Patient disease activity was abstracted from the patient medical record, and this was compared to the questionnaire and whole blood assay results. Results A total of 20 patients participated in the study; 8 patients had active disease and the remaining were found to be quiescent. There was no significant difference between the patient-reported general health ratings based on sex, age, disease activity, or category of immune dysregulation (p > 0.05). The cTfh percentages were expanded in patients with active disease as compared to those with quiescent (p < 0.05). However, there was no significant correlation between cTfh percentage and patient-reported unhealthy days from the questionnaire (R2 = 0.113, p > 0.05). Conclusions Patients with active immune dysregulation were found to have expanded cTfh percentages as compared to those with quiescent disease, however this was not reflected in patient-reported quality of life questionnaires. Better understanding of disease activity and the patient experience is vital to optimize appropriate treatments and outcomes for patients with IEI and immune dysregulation, and more investigation is needed.
Collapse
|
13
|
LaBere B, Nguyen AA, Habiballah SB, Elkins M, Imperial J, Li B, Devana S, Timilsina S, Stubbs SB, Joerger J, Chou J, Platt CD. Clinical utility of measuring CD4 + T follicular cells in patients with immune dysregulation. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2023:2023.06.06.23291032. [PMID: 37333344 PMCID: PMC10274986 DOI: 10.1101/2023.06.06.23291032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Mechanistic studies of autoimmune disorders have identified circulating T follicular helper (cTfh) cells as drivers of autoimmunity. However, the quantification of cTfh cells is not yet used in clinical practice due to the lack of age-stratified normal ranges and the unknown sensitivity and specificity of this test for autoimmunity. We enrolled 238 healthy participants and 130 patients with common and rare disorders of autoimmunity or autoinflammation. Patients with infections, active malignancy, or any history of transplantation were excluded. In 238 healthy controls, median cTfh percentages (range 4.8% - 6.2%) were comparable among age groups, sexes, races, and ethnicities, apart from a significantly lower percentages in children less than 1 year of age (median 2.1%, CI: 0.4% - 6.8, p< 0.0001). Among 130 patients with over 40 immune regulatory disorders, a cTfh percentage exceeding 12% had 88% sensitivity and 94% specificity for differentiating disorders with adaptive immune cell dysregulation from those with predominantly innate cell defects. This threshold had a sensitivity of 86% and specificity of 100% for active autoimmunity and normalized with effective treatment. cTfh percentages exceeding 12% distinguish autoimmunity from autoinflammation, thereby differentiating two endotypes of immune dysregulation with overlapping symptoms and different therapies.
Collapse
|
14
|
Hlongwa L, Peter J, Mayne E. Value of diagnostic vaccination in diagnosis of humoral inborn errors of immunity. Hum Immunol 2023:S0198-8859(23)00066-6. [PMID: 37080873 DOI: 10.1016/j.humimm.2023.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2022] [Revised: 04/11/2023] [Accepted: 04/11/2023] [Indexed: 04/22/2023]
Abstract
Inborn errors of immunity (IEIs) or primary immunodeficiency diseases, are disorders caused by genetic defects affecting immune function. Clinically, IEI presents mainly as recurrent or severe infections, immune dysregulation (autoimmunity or autoinflammatory disorders), and lymphoproliferation with or without dysmorphic features. Humoral IEIs are the largest subgroup of IEI, with a wide spectrum of quantitative and qualitative antibody defects. These disorders are normally diagnosed based on immunological evaluation; diagnostic vaccination is part of this evaluation. This review examines the importance and relevance of diagnostic vaccination in the diagnosis of humoral IEIs and different technologies which can be utilised in diagnosis.
Collapse
Affiliation(s)
- Luyanda Hlongwa
- Division of Immunology, Department of Pathology, Faculty of Health Science, University of Cape Town, South Africa
| | - Jonathan Peter
- Division of Allergology and Clinical Immunology, Groote Schuur Hospital, University of Cape Town, South Africa
| | - Elizabeth Mayne
- Division of Immunology, Department of Pathology, Faculty of Health Science, University of Cape Town, South Africa; Division of Immunology, National Health Laboratory Service, South Africa.
| |
Collapse
|
15
|
McKenzie CI, Varese N, Aui PM, Reinwald S, Wines BD, Hogarth PM, Thien F, Hew M, Rolland JM, O'Hehir RE, van Zelm MC. RNA sequencing of single allergen-specific memory B cells after grass pollen immunotherapy: Two unique cell fates and CD29 as a biomarker for treatment effect. Allergy 2023; 78:822-835. [PMID: 36153670 PMCID: PMC10952829 DOI: 10.1111/all.15529] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 08/26/2022] [Accepted: 08/29/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND Sublingual immunotherapy (SLIT) for grass pollen allergy can modify the natural history of allergic rhinitis and is associated with increased allergen-specific IgG4 . IgG4 competitively inhibits functional IgE on the surface of effector cells, such as mast cells and basophils, from binding to allergens. To further understand the important role memory B-cell (Bmem) responses play in mediating the beneficial effects of SLIT, we assessed changes in allergen-specific Bmem subsets induced by SLIT for grass pollen allergy. METHODS Blood samples were collected twice outside the pollen season from twenty-seven patients with sensitization to ryegrass pollen (RGP; Lolium perenne) and seasonal rhinoconjunctivitis. Thirteen received 4-month pre-seasonal SLIT for grass pollen allergy, and 14 received standard pharmacotherapy only. Single-cell RNA sequencing was performed on FACS-purified Lol p 1-specific Bmem before and after SLIT from four patients, and significant genes were validated by flow cytometry on the total cohort. RESULTS Four months of SLIT increased RGP-specific IgE and IgG4 in serum and induced two Lol p 1-specific Bmem subsets with unique transcriptional profiles. Both subsets had upregulated expression of beta 1 integrin ITGB1 (CD29), whereas IGHE (IgE), IGHG4 (IgG4 ), FCER2 (CD23), and IL13RA1 were upregulated in one subset. There was an increase in the proportion of Lol p 1+ Bmem expressing surface IgG4 , CD23, and CD29 after SLIT. CONCLUSIONS A clinically successful 4 months course of SLIT for grass pollen allergy induces two transcriptionally unique Bmem fates. Associated changes in surface-expressed proteins on these Bmem subsets can be used as early biomarkers for treatment effects.
Collapse
Affiliation(s)
- Craig I. McKenzie
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Nirupama Varese
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Pei Mun Aui
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Simone Reinwald
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Bruce D. Wines
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
| | - P. Mark Hogarth
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Immune Therapies GroupBurnet InstituteMelbourneVictoriaAustralia
- Department of PathologyThe University of MelbourneParkvilleVictoriaAustralia
| | - Francis Thien
- Respiratory Medicine, Eastern HealthBox Hill and Monash UniversityMelbourneVictoriaAustralia
| | - Mark Hew
- School of Public Health and Preventive MedicineMonash UniversityMelbourneVictoriaAustralia
- Allergy, Asthma and Clinical ImmunologyAlfred HealthMelbourneVictoriaAustralia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Robyn E. O'Hehir
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Allergy, Asthma and Clinical ImmunologyAlfred HealthMelbourneVictoriaAustralia
| | - Menno C. van Zelm
- Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
- Allergy, Asthma and Clinical ImmunologyAlfred HealthMelbourneVictoriaAustralia
| |
Collapse
|
16
|
The Autoimmune Manifestations in Patients with Genetic Defects in the B Cell Development and Differentiation Stages. J Clin Immunol 2023; 43:819-834. [PMID: 36790564 PMCID: PMC10110688 DOI: 10.1007/s10875-023-01442-6] [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: 11/02/2022] [Accepted: 01/22/2023] [Indexed: 02/16/2023]
Abstract
PURPOSE Primary B cell defects manifesting as predominantly antibody deficiencies result from variable inborn errors of the B cell lineage and their development, including impairments in early bone marrow development, class switch recombination (CSR), or terminal B cell differentiation. In this study, we aimed to investigate autoimmunity in monogenic patients with B cell development and differentiation defects. METHODS Patients with known genetic defects in the B cell development and differentiation were recruited from the Iranian inborn errors of immunity registry. RESULTS A total of 393 patients with a known genetic defect in the B cell development and differentiation (257 males; 65.4%) with a median age of 12 (6-20) years were enrolled in this study. After categorizing patients, 109 patients had intrinsic B cell defects. More than half of the patients had defects in one of the ATM (85 patients), BTK (76 patients), LRBA (34 patients), and DOCK8 (33 patients) genes. Fifteen patients (3.8%) showed autoimmune complications as their first manifestation. During the course of the disease, autoimmunity was reported in 81 (20.6%) patients at a median age of 4 (2-7) years, among which 65 patients had mixed intrinsic and extrinsic and 16 had intrinsic B cell defects. The comparison between patients with the mentioned four main gene defects showed that the patient group with LRBA defect had a significantly higher frequency of autoimmunity compared to those with other gene defects. Based on the B cell defect stage, 13% of patients with early B cell defect, 17% of patients with CSR defect, and 40% of patients who had terminal B cell defect presented at least one type of autoimmunity. CONCLUSION Our results demonstrated that gene mutations involved in human B cell terminal stage development mainly LRBA gene defect have the highest association with autoimmunity.
Collapse
|
17
|
Long-Term Immunological Memory of SARS-CoV-2 Is Present in Patients with Primary Antibody Deficiencies for up to a Year after Vaccination. Vaccines (Basel) 2023; 11:vaccines11020354. [PMID: 36851231 PMCID: PMC9959530 DOI: 10.3390/vaccines11020354] [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: 01/02/2023] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Some studies have found increased coronavirus disease-19 (COVID-19)-related morbidity and mortality in patients with primary antibody deficiencies. Immunization against COVID-19 may, therefore, be particularly important in these patients. However, the durability of the immune response remains unclear in such patients. In this study, we evaluated the cellular and humoral response to severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) antigens in a cross-sectional study of 32 patients with primary antibody deficiency (n = 17 with common variable immunodeficiency (CVID) and n = 15 with selective IgA deficiency) and 15 healthy controls. Serological and cellular responses were determined using enzyme-linked immunosorbent assay and interferon-gamma release assays. The subsets of B and T lymphocytes were measured using flow cytometry. Of the 32 patients, 28 had completed the vaccination regimen with a median time after vaccination of 173 days (IQR = 142): 27 patients showed a positive spike-peptide-specific antibody response, and 26 patients showed a positive spike-peptide-specific T-cell response. The median level of antibody response in CVID patients (5.47 ratio (IQR = 4.08)) was lower compared to healthy controls (9.43 ratio (IQR = 2.13)). No difference in anti-spike T-cell response was found between the groups. The results of this study indicate that markers of the sustained SARS-CoV-2 spike-specific immune response are detectable several months after vaccination in patients with primary antibody deficiencies comparable to controls.
Collapse
|
18
|
Albert MH, Sirait T, Eikema DJ, Bakunina K, Wehr C, Suarez F, Fox ML, Mahlaoui N, Gennery AR, Lankester AC, Beier R, Bernardo ME, Bigley V, Lindemans CA, Burns SO, Carpenter B, Dybko J, Güngör T, Hauck F, Lum SH, Balashov D, Meisel R, Moshous D, Schulz A, Speckmann C, Slatter MA, Strahm B, Uckan-Cetinkaya D, Meyts I, Vallée TC, Wynn R, Neven B, Morris EC, Aiuti A, Maschan A, Aljurf M, Gedde-Dahl T, Gurman G, Bordon V, Kriván G, Locatelli F, Porta F, Valcárcel D, Beguin Y, Faraci M, Kröger N, Kulagin A, Shaw PJ, Veelken JH, Diaz de Heredia C, Fagioli F, Felber M, Gruhn B, Holter W, Rössig C, Sedlacek P, Apperley J, Ayas M, Bodova I, Choi G, Cornelissen JJ, Sirvent A, Khan A, Kupesiz A, Lenhoff S, Ozdogu H, von der Weid N, Rovira M, Schots R, Vinh DC. Hematopoietic stem cell transplantation for adolescents and adults with inborn errors of immunity: an EBMT IEWP study. Blood 2022; 140:1635-1649. [PMID: 35344580 DOI: 10.1182/blood.2022015506] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/17/2022] [Indexed: 11/20/2022] Open
Abstract
Allogeneic hematopoietic stem cell transplantation (HSCT) is the gold standard curative therapy for infants and children with many inborn errors of immunity (IEI), but adolescents and adults with IEI are rarely referred for transplant. Lack of published HSCT outcome data outside small, single-center studies and perceived high risk of transplant-related mortality have delayed the adoption of HSCT for IEI patients presenting or developing significant organ damage later in life. This large retrospective, multicenter HSCT outcome study reports on 329 IEI patients (age range, 15-62.5 years at HSCT). Patients underwent first HSCT between 2000 and 2019. Primary endpoints were overall survival (OS) and event-free survival (EFS). We also evaluated the influence of IEI-subgroup and IEI-specific risk factors at HSCT, including infections, bronchiectasis, colitis, malignancy, inflammatory lung disease, splenectomy, hepatic dysfunction, and systemic immunosuppression. At a median follow-up of 44.3 months, the estimated OS at 1 and 5 years post-HSCT for all patients was 78% and 71%, and EFS was 65% and 62%, respectively, with low rates of severe acute (8%) or extensive chronic (7%) graft-versus-host disease. On univariate analysis, OS and EFS were inferior in patients with primary antibody deficiency, bronchiectasis, prior splenectomy, hepatic comorbidity, and higher hematopoietic cell transplant comorbidity index scores. On multivariable analysis, EFS was inferior in those with a higher number of IEI-associated complications. Neither age nor donor had a significant effect on OS or EFS. We have identified age-independent risk factors for adverse outcome, providing much needed evidence to identify which patients are most likely to benefit from HSCT.
Collapse
Affiliation(s)
- Michael H Albert
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Tiarlan Sirait
- Statistical Unit and Data Office, European Society for Blood and Marrow Transplantation (EBMT), Leiden, The Netherlands
| | - Dirk-Jan Eikema
- Statistical Unit and Data Office, European Society for Blood and Marrow Transplantation (EBMT), Leiden, The Netherlands
| | - Katerina Bakunina
- Statistical Unit and Data Office, European Society for Blood and Marrow Transplantation (EBMT), Leiden, The Netherlands
| | - Claudia Wehr
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Medicine I, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Felipe Suarez
- Department of Adult Hematology, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Maria Laura Fox
- Department of Hematology, Hospital Universitari Vall d'Hebron, Experimental Hematology, Vall d'Hebron Institute of Oncology (VHIO), Vall d'Hebron Hospital Campus, Barcelona, Spain
| | - Nizar Mahlaoui
- Pediatric Immuno-Hematology and Rheumatology Unit, Necker-Enfants University Hospital and French National Reference Center for Primary Immunodeficiencies (CEREDIH), AP-HP, Paris, France
| | - Andrew R Gennery
- Department of Pediatric Immunology & Haematopoietic Stem Cell Transplantation (HSCT), Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Arjan C Lankester
- Department of Pediatrics, Pediatric Stem Cell Transplantation Program, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Rita Beier
- Department of Pediatric Hematology and Oncology, Medizinische Hochschule Hannover (MHH), Hannover, Germany
| | - Maria Ester Bernardo
- Department of Pediatric Immunohematology, Istituto Di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, Milan, Italy
| | - Venetia Bigley
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
- Northern Center for Cancer Care, Newcastle upon Tyne Hospitals National Health Service (NHS) Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Caroline A Lindemans
- Department of Pediatric Immunology, University Medical Center Utrecht, Utrecht, The Netherlands
- Department of Pediatric Blood and Bone Marrow Transplantation, Princess Maxima Center, Utrecht, The Netherlands
| | - Siobhan O Burns
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, United Kingdom
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
| | - Ben Carpenter
- Department of Clinical Hematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Jaroslaw Dybko
- Department of Hematology and Cellular Transplantation, Lower Silesian Center of Oncology, Wroclaw, Poland
| | - Tayfun Güngör
- Department of Hematology/Oncology/Immunology, Gene-Therapy, and Stem Cell Transplantation, University Children's Hospital Zurich - Eleonore Foundation & Children's Research Center (CRC), Zürich, Switzerland
| | - Fabian Hauck
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Su Han Lum
- Department of Pediatric Immunology & Haematopoietic Stem Cell Transplantation (HSCT), Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
| | - Dmitry Balashov
- Department of Hematopoietic Stem Cell Transplantation, Dmitriy Rogachev National Center for Pediatric Hematology, Oncology, and Immunology, Moscow, Russian Federation
| | - Roland Meisel
- Division of Pediatric Stem Cell Therapy, Department of Paediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Despina Moshous
- Department of Pediatric Immunology, Hematology, and Rheumatology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Carsten Speckmann
- Institute for Immunodeficiency, Center for Chronic Immunodeficiency, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Department of Pediatric Hematology and Oncology, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Mary A Slatter
- Department of Pediatric Immunology & Haematopoietic Stem Cell Transplantation (HSCT), Great North Children's Hospital, Newcastle upon Tyne, United Kingdom
- Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Brigitte Strahm
- Department of Pediatric Hematology and Oncology, Center for Pediatrics and Adolescent Medicine, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Duygu Uckan-Cetinkaya
- Department of Pediatrics, Bone Marrow Transplantation (BMT) Unit, Hacettepe University, Faculty of Medicine, Ankara, Turkey
| | - Isabelle Meyts
- Department of Pediatrics, University Hospitals Leuven, Leuven, Belgium
- Department of Microbiology, Immunology, and Transplantation, Katholieke Universiteit (KU) Leuven, Leuven, Belgium
| | - Tanja C Vallée
- Department of Pediatrics, Dr. von Hauner Children's Hospital, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany
| | - Robert Wynn
- Blood and Marrow Transplant Program, Royal Manchester Children's Hospital, Manchester, United Kingdom
| | - Bénédicte Neven
- Department of Pediatric Immunology, Hematology, and Rheumatology, Necker-Enfants Malades University Hospital, AP-HP, Paris, France
| | - Emma C Morris
- Department of Immunology, Royal Free London Hospitals NHS Foundation Trust, London, United Kingdom
- Institute of Immunity and Transplantation, University College London (UCL), London, United Kingdom
- Department of Clinical Hematology, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hartley GE, Edwards ESJ, O’Hehir RE, van Zelm MC. New insights into human immune memory from SARS-CoV-2 infection and vaccination. Allergy 2022; 77:3553-3566. [PMID: 36048132 PMCID: PMC9538469 DOI: 10.1111/all.15502] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 08/13/2022] [Accepted: 08/29/2022] [Indexed: 01/28/2023]
Abstract
Since early 2020, the world has been embroiled in an ongoing viral pandemic with SARS-CoV-2 and emerging variants resulting in mass morbidity and an estimated 6 million deaths globally. The scientific community pivoted rapidly, providing unique and innovative means to identify infected individuals, technologies to evaluate immune responses to infection and vaccination, and new therapeutic strategies to treat infected individuals. Never before has immunology been so critically at the forefront of combatting a global pandemic. It has now become evident that not just antibody responses, but formation and durability of immune memory cells following vaccination are associated with protection against severe disease from SARS-CoV-2 infection. Furthermore, the emergence of variants of concern (VoC) highlight the need for immunological markers to quantify the protective capacity of Wuhan-based vaccines. Thus, harnessing and modulating the immune response is key to successful vaccination and treatment of disease. We here review the latest knowledge about immune memory generation and durability following natural infection and vaccination, and provide insights into the attributes of immune memory that may protect from emerging variants.
Collapse
Affiliation(s)
- Gemma E. Hartley
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Emily S. J. Edwards
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Robyn E. O’Hehir
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia,Allergy, Asthma and Clinical Immunology ServiceAlfred HospitalMelbourneVictoriaAustralia
| | - Menno C. van Zelm
- Allergy and Clinical Immunology Laboratory, Department of Immunology and Pathology, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia,Allergy, Asthma and Clinical Immunology ServiceAlfred HospitalMelbourneVictoriaAustralia
| |
Collapse
|
20
|
Gill PA, Muir JG, Gibson PR, van Zelm MC. A Randomized Dietary Intervention to Increase Colonic and Peripheral Blood Short-Chain Fatty Acids Modulates the Blood B- and T-cell Compartments in Healthy Humans. Am J Clin Nutr 2022; 116:1354-1367. [PMID: 36084000 PMCID: PMC9630882 DOI: 10.1093/ajcn/nqac246] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 08/15/2022] [Accepted: 09/01/2022] [Indexed: 11/25/2022] Open
Abstract
BACKGROUND Short-chain fatty acids (SCFA) have immune-modulating effects in animal models of disease. However, there is limited evidence that this may occur in humans. OBJECTIVES This study aimed to determine the effects of increased exposure to SCFA via dietary manipulation on colonic fermentation and adaptive immune cells. METHODS Twenty healthy, young adults (18-45 years of age) underwent a blinded, randomized, cross-over dietary intervention, consuming a high-SCFA producing diet and matched low-SCFA diet for 21 days with 21-day wash-out in between. SCFA were provided through resistant starch, inulin and apple cider vinegar. Blood and 3-day total fecal output were collected at baseline and at the end of each diet. Gas chromatography was used to measure fecal and plasma SCFA. Flow cytometry was used for peripheral blood immuno-phenotyping. RESULTS High-SCFA diet was associated with significantly (paired samples Wilcoxon test) higher median [IQR] fecal SCFA concentrations (86.6 [59.0] vs 75.4 [56.2] µmol/g, P = 0.02) and significantly lower median fecal ammonia concentrations (26.2 [14.7] vs 33.4 [18.5] µmol/g, P = 0.04) than the low-SCFA diet. Plasma propionate (9.87 [12.3] vs 4.72 [7.6] µmol/L, P = 0.049) and butyrate (2.85 [1.35] vs 2.02 [1.29] µmol/L, P = 0.03) were significantly higher after high-SCFA diet than after low-SCFA diet. Blood total B cells (184 [112] vs 199 [143] cells/µL, P = 0.04), naive B cells (83 [66] vs 95 [89] cells/µL, P = 0.02), Th1 cells (22 [19] vs 29 [16] cells/µL, P = 0.03) and mucosal-associated invariant T (MAIT) cells (62 [83] vs 69 [114] cells/µL, P = 0.02) were significantly lower after high-SCFA diet than low-SCFA diet. CONCLUSION Increasing colonic and peripheral blood SCFA has discrete effects on circulating immune cells in healthy humans following 3-week intervention. Further studies, e.g., in patients with inflammatory disease, are necessary to determine if these changes have immunomodulatory effects, whether these are therapeutically beneficial, and whether prolonged intake might be required. Clinical trial registry: Australian New Zealand Clinical trials registry: ACTRN12618001054202. <https://www.anzctr.org.au/Trial/Registration/TrialReview.aspx?id=375342&isReview=true>.
Collapse
Affiliation(s)
| | - Jane G Muir
- Department of Gastroenterology and Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Australia
| | - Peter R Gibson
- Department of Gastroenterology and Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Australia
| |
Collapse
|
21
|
Ratnayake G, Reinwald S, Edwards J, Wong N, Yu D, Ward R, Smith R, Haydon A, Au PM, van Zelm MC, Senthi S. Blood T-cell profiling in metastatic melanoma patients as a marker for response to immune checkpoint inhibitors combined with radiotherapy. Radiother Oncol 2022; 173:299-305. [PMID: 35772575 DOI: 10.1016/j.radonc.2022.06.016] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/15/2022] [Accepted: 06/19/2022] [Indexed: 10/17/2022]
Abstract
BACKGROUND The addition of stereotactic ablative radiotherapy (SABR) to immune checkpoint inhibitors (ICIs) has the potential to significantly improve outcomes in the treatment of metastatic melanoma. We analysed peripheral blood immune cells of patients receiving combination SABR and ICI to detect the effect of treatment and identify potential biomarkers that predict outcome. METHODS 24 polymetastatic melanoma patients participated in the SABR IMPACT trial, receiving standard dose immunotherapy with anti-PD-1 and/or anti-CTLA-4 and stereotactic ablative radiotherapy to one site. Comprehensive immunophenotyping of T-cells was performed with flow cytometry on blood samples from 13 patients at baseline and following the first 4 cycles of treatment. RESULTS Following four cycles of immunotherapy and SABR, the proportion of naïve subsets were reduced within both the CD4 and CD8 T-cell lineages. Independently, SABR resulted in increased expression of PD-1 (p = 0.019) and ICOS (p = 0.046) on the CD8+ T-cells, accompanied by a reduction in regulatory T-cell frequencies (p = 0.048). A multivariate discriminant analysis revealed a baseline signature of lower levels of CD8+ naive T-cells and higher expression of TIM-3 on regulatory T-cells and memory T-cells better predicted response. CONCLUSION The combination of immunotherapy and SABR changed the immunophenotype of blood T cells, with some shifts attributable to SABR. Importantly, we identified a T-cell signature at baseline that best predicted response. Validation of these findings in an independent cohort could confirm these as biomarkers at baseline or early during treatment, and whether these can be utilised to stratify patients for high or low intensity treatment to reduce toxicity.
Collapse
Affiliation(s)
- Gishan Ratnayake
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia; Radiation Oncology Princess Alexandra Hospital Raymond Terrace, Brisbane, Australia; Central Clinical School, Monash University, Melbourne, Australia.
| | - Simone Reinwald
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Jack Edwards
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia; Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Nicholas Wong
- Central Clinical School, Monash University, Melbourne, Australia; Monash Bioinformatic Platform, Monash University, Melbourne, Australia
| | - Di Yu
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia
| | - Rachel Ward
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia
| | - Robin Smith
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia
| | - Andrew Haydon
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia
| | - Pei M Au
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia; Monash Bioinformatic Platform, Monash University, Melbourne, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, Australia; Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Australia
| | - Sashendra Senthi
- Alfred Health Radiation Oncology, The Alfred Hospital, Melbourne, Australia; Radiation Oncology Princess Alexandra Hospital Raymond Terrace, Brisbane, Australia
| |
Collapse
|
22
|
Chawla S, Jindal AK, Arora K, Tyagi R, Dhaliwal M, Rawat A. T Cell Abnormalities in X-Linked Agammaglobulinaemia: an Updated Review. Clin Rev Allergy Immunol 2022:10.1007/s12016-022-08949-7. [PMID: 35708830 PMCID: PMC9201264 DOI: 10.1007/s12016-022-08949-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/31/2022] [Indexed: 12/03/2022]
Abstract
X-linked agammaglobulinaemia (XLA) is a primary immunodeficiency (PID) resulting from a defect in the B cell development. It has conventionally been thought that T cells play a major role in the development and function of the B cell compartment. However, it has also been shown that B cells and T cells undergo bidirectional interactions and B cells also influence the structure and function of the T cell compartment. Patients with XLA offer a unique opportunity to understand the effect of absent B cells on the T cell compartment. In this review, we provide an update on abnormalities in the T cell compartment in patients with XLA. Studies have shown impaired memory T cells, follicular helper T cells, T regulatory cells and T helper 17 in patients with XLA. In addition, these patients have also been reported to have abnormal delayed cell-mediated immune responses and vaccine-specific T cell-mediated immune responses; defective T helper cell polarization and impaired T cell receptor diversity. At present, the clinical significance of these T cell abnormalities has not been studied in detail. However, these abnormalities may result in an increased risk of viral infections, autoimmunity, autoinflammation and possibly chronic lung disease. Abnormal response to SARS-Cov2 vaccine in patients with XLA and prolonged persistence of SARS-Cov2 virus in the respiratory tract of these patients may be related to abnormalities in the T cell compartment.
Collapse
Affiliation(s)
- Sanchi Chawla
- Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Ankur Kumar Jindal
- Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India.
| | - Kanika Arora
- Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Rahul Tyagi
- Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Manpreet Dhaliwal
- Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Paediatrics, Advanced Paediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, 160012, India
| |
Collapse
|
23
|
Distinct CD8 T Cell Populations with Differential Exhaustion Profiles Associate with Secondary Complications in Common Variable Immunodeficiency. J Clin Immunol 2022; 42:1254-1269. [PMID: 35589883 PMCID: PMC9537220 DOI: 10.1007/s10875-022-01291-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/09/2022] [Indexed: 11/08/2022]
Abstract
Purpose Common variable immunodeficiency (CVID) is the most frequent symptomatic primary immunodeficiency, with heterogeneous clinical presentation. Our goal was to analyze CD8 T cell homeostasis in patients with infection only CVID, compared to those additionally affected by dysregulatory and autoimmune phenomena. Methods We used flow and mass cytometry evaluation of peripheral blood of 40 patients with CVID and 17 healthy donors. Results CD8 T cells are skewed in patients with CVID, with loss of naïve and increase of effector memory stages, expansion of cell clusters with high functional exhaustion scores, and a highly activated population of cells with immunoregulatory features, producing IL-10. These findings correlate to clinically widely used B cell-based EURO classification. Features of exhaustion, including loss of CD127 and CD28, and expression of TIGIT and PD-1 in CD8 T cells are strongly associated with interstitial lung disease and autoimmune cytopenias, whereas CD8 T cell activation with elevated HLA-DR and CD38 expression predict non-infectious diarrhea. Conclusion We demonstrate features of advanced differentiation, exhaustion, activation, and immunoregulatory capabilities within CD8 T cells of CVID patients. Assessment of CD8 T cell phenotype may allow risk assessment of CVID patients and provide new insights into CVID pathogenesis, including a better understanding of mechanisms underlying T cell exhaustion and regulation. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-022-01291-9.
Collapse
|
24
|
Bergman P, Wullimann D, Gao Y, Wahren Borgström E, Norlin AC, Lind Enoksson S, Aleman S, Ljunggren HG, Buggert M, Smith CIE. Elevated CD21 low B Cell Frequency Is a Marker of Poor Immunity to Pfizer-BioNTech BNT162b2 mRNA Vaccine Against SARS-CoV-2 in Patients with Common Variable Immunodeficiency. J Clin Immunol 2022; 42:716-727. [PMID: 35290571 PMCID: PMC8922070 DOI: 10.1007/s10875-022-01244-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/24/2022] [Indexed: 12/18/2022]
Abstract
PURPOSE Limited data is available on the effect of COVID-19 vaccination in immunocompromised individuals. Here, we provide the results from vaccinating a single-center cohort of patients with common variable immunodeficiency (CVID). METHODS In a prospective, open-label clinical trial, 50 patients with CVID and 90 age-matched healthy controls (HC) were analyzed for SARS-CoV-2 spike antibody (Ab) production after one or two doses of the Pfizer-BioNTech BNT162b2 mRNA vaccine. Additionally, in selected patients, SARS-CoV-2 spike-specific T-cells were assessed. RESULTS A potent vaccine-induced anti-spike-specific IgG Ab response was observed in all the HC. In contrast, only 68.3% of the CVID patients seroconverted, with median titers of specific Ab being 83-fold lower than in HC. In fact, only 4/46 patients (8.6%) of patients who were seronegative at baseline reached the threshold for an optimal response (250 U/mL). Using the EUROclass definition, patients with either a reduced proportion, but not absolute counts, of switched memory B-cells or having an increased frequency of CD21low B-cells generally generated poor vaccine responses. Overall, CVID-patients had reduced spike-specific IFN-γ positive CD4+ T cell responses 2 weeks after the second dose, compared to HC. The total CD4 and CD4 central memory cell counts correlated with humoral immunity to the vaccine. CONCLUSIONS CVID patients with low frequency of switched memory B-cells or an increased frequency of CD21low B-cells according to the EUROclass definition demonstrated poor responses to Pfizer-BioNTech BNT162b2 mRNA vaccination. Cellular immune responses were significantly affected, affirming that the defect in CVID is not limited to humoral immunity.
Collapse
Affiliation(s)
- Peter Bergman
- Department of Infectious Diseases, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden.
- Department of Laboratory Medicine, Clinical Microbiology, Karolinska Institutet, Stockholm, Sweden.
| | - David Wullimann
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Yu Gao
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Emilie Wahren Borgström
- Department of Infectious Diseases, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Anna-Carin Norlin
- Department of Infectious Diseases, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden
| | - Sara Lind Enoksson
- Department of Clinical immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
- Department of Clinical Science, Investigation and Technology (CLINTEC), Karolinska Institutet, Stockholm, Sweden
| | - Soo Aleman
- Department of Infectious Diseases, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Infectious Diseases, Karolinska Institutet, Stockholm, Sweden
| | - Hans-Gustaf Ljunggren
- Department of Infectious Diseases, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institutet, Stockholm, Sweden
| | - C I Edvard Smith
- Department of Infectious Diseases, Immunodeficiency Unit, Karolinska University Hospital, Stockholm, Sweden
- Department of Laboratory Medicine, Biomolecular and Cellular Medicine, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
25
|
Wenyang Huazhuo Tuihuang Formula Inhibits the Th17/Treg Cell Imbalance and Protects against Acute-on-Chronic Liver Failure. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:5652172. [PMID: 35399641 PMCID: PMC8986372 DOI: 10.1155/2022/5652172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/22/2022] [Accepted: 03/07/2022] [Indexed: 11/18/2022]
Abstract
Objective Acute-on-chronic liver failure (ACLF) is a group of chronic liver diseases and caused by acute internal and external liver injury. Wenyang Huazhuo Tuihuang (WYHZTH) formula had a good clinical effect on promoting the resolution of jaundice. The aim of this study is to further investigate the mechanism of the WYHZTH formula in the ACLF rat model. Methods The ACLF rat model was constructed by combining human serum albumin with LPS and D-gal. WYHZTH was used to intervene and treat. The cytokines IL-17, IL-23, IL-10, and TGF-β were detected by ELISA and fluorescence-quantitative PCR. Flow cytometry was used to detect the percentage of Th17 and Treg cells in the peripheral blood and liver tissues of each group of rats. The pathological changes in the liver tissue were detected by hematoxylin-eosin staining, immunohistochemistry, and electron microscopy. Results Compared with the ACLF group, the WYHZTH formula and Thy significantly decreased the levels of ALT, AST, and CHE in the ACLF group. After drug intervention, apoptosis was significantly reduced. The PCNA expression decreased in the ACLF model group but increased in the WYHZTH or Thy group. Under transmission electron microscope, hepatocytes in the ACLF group showed obvious necrosis. After drug intervention, hepatocyte necrosis was reduced with most of the structure returning to normal. Conclusion This present study demonstrated that WYHZTH formula may protect against acute-on-chronic liver failure, which may be related to the inhibition of Th17/Treg cell imbalance.
Collapse
|
26
|
Hermans MAW, Heeringa JJ, Swagemakers SGA, Schrijver B, van Daele PLA, van der Spek PJ, van Hagen PM, van Zelm MC, Dik WA. Altered leukocyte subsets and immune proteome indicate pro-inflammatory mechanisms in mastocytosis. J Allergy Clin Immunol 2022; 150:146-156.e10. [PMID: 35026208 DOI: 10.1016/j.jaci.2021.12.786] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/26/2022]
Abstract
BACKGROUND Indolent systemic mastocytosis (ISM) is characterized by pathological accumulation of mast cells. The mechanism behind its phenotypic heterogeneity is not well understood. Interaction of mast cells with other immune cells might cause systemic inflammation and thereby associated symptoms. OBJECTIVE To investigate peripheral leukocyte compartments and serum immune proteome in ISM. METHODS Peripheral blood leukocyte phenotyping using flowcytometry in a cohort of 18 adults with ISM and 12 healthy controls. Targeted proteomics was performed to measure 169 proteins associated with inflammation on serum of another 20 ISM patients and 20 healthy controls. RESULTS Proportions of plasmacytoid dendritic cells (pDC) and monocytes were significantly decreased while T-helper 2 cells were increased in peripheral blood of ISM patients. Furthermore, a shift from naïve to memory T-cells was observed. Hierarchical clustering of the serum proteome revealed two distinct subgroups within ISM patients. In subgroup A (n=8), 62 proteins were significantly overexpressed, whereas subgroup B (n=12) was comparable to healthy controls. Patients in subgroup A displayed upregulated signaling pathways downstream of Toll-like receptor 4, TNF-α and interferon-γFatigue was more often present in subgroup A compared to B (75% vs. 33% respectively, p 0.06). CONCLUSIONS Altered distribution of leukocyte subsets and a pro-inflammatory proteome were observed in subsequent two cohorts of ISM patients. We hypothesize that neoplastic mast cells recruit and activate pDC, monocytes and T-cells leading to a vicious cycle of inflammation. CLINICAL IMPLICATION Characterization of inflammatory mechanisms in ISM might uncover additional targets for treatment, in particular for constitutional symptoms such as fatigue.
Collapse
Affiliation(s)
- M A W Hermans
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands.
| | - J J Heeringa
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - S G A Swagemakers
- Department of Pathology and Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - B Schrijver
- Laboratory of Medical Immunology, department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P L A van Daele
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands; Laboratory of Medical Immunology, department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P J van der Spek
- Department of Pathology and Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - P M van Hagen
- Department of Internal Medicine, section Allergy & Clinical Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands; Laboratory of Medical Immunology, department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - M C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University and Alfred Hospital, Melbourne, Victoria, Australia
| | - W A Dik
- Laboratory of Medical Immunology, department of Immunology, Erasmus University Medical Center, Rotterdam, The Netherlands
| |
Collapse
|
27
|
McKenzie CI, Varese N, Aui PM, Wines BD, Hogarth PM, Thien F, Hew M, Rolland JM, O’Hehir RE, Zelm MC. CytoBas: Precision component-resolved diagnostics for allergy using flow cytometric staining of basophils with recombinant allergen tetramers. Allergy 2021; 76:3028-3040. [PMID: 33772805 PMCID: PMC8518718 DOI: 10.1111/all.14832] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 02/18/2021] [Accepted: 02/19/2021] [Indexed: 12/26/2022]
Abstract
BACKGROUND Diagnostic tests for allergy rely on detecting allergen-specific IgE. Component-resolved diagnostics incorporate multiple defined allergen components to improve the quality of diagnosis and patient care. OBJECTIVE To develop a new approach for determining sensitization to specific allergen components that utilizes fluorescent protein tetramers for direct staining of IgE on blood basophils by flow cytometry. METHODS Recombinant forms of Lol p 1 and Lol p 5 proteins from ryegrass pollen (RGP) and Api m 1 from honeybee venom (BV) were produced, biotinylated, and tetramerized with streptavidin-fluorochrome conjugates. Blood samples from 50 RGP-allergic, 41 BV-allergic, and 26 controls were incubated with fluorescent protein tetramers for flow cytometric evaluation of basophil allergen binding and activation. RESULTS Allergen tetramers bound to and activated basophils from relevant allergic patients but not controls. Direct fluorescence staining of Api m 1 and Lol p 1 tetramers had greater positive predictive values than basophil activation for BV and RGP allergy, respectively, as defined with receiver operator characteristics (ROC) curves. Staining intensities of allergen tetramers correlated with allergen-specific IgE levels in serum. Inclusion of multiple allergens coupled with distinct fluorochromes in a single-tube assay enabled rapid detection of sensitization to both Lol p 1 and Lol p 5 in RGP-allergic patients and discriminated between controls, BV-allergic, and RGP-allergic patients. CONCLUSION Our novel flow cytometric assay, termed CytoBas, enables rapid and reliable detection of clinically relevant allergic sensitization. The intensity of fluorescent allergen tetramer staining of basophils has a high positive predictive value for disease, and the assay can be multiplexed for a component-resolved and differential diagnostic test for allergy.
Collapse
Affiliation(s)
- Craig I. McKenzie
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Nirupama Varese
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University Melbourne VIC Australia
| | - Pei M. Aui
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
| | - Bruce D. Wines
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Immune Therapies Group Burnet Institute Melbourne Australia
- Department of Pathology The University of Melbourne Parkville VIC Australia
| | - Philip Mark Hogarth
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Immune Therapies Group Burnet Institute Melbourne Australia
- Department of Pathology The University of Melbourne Parkville VIC Australia
| | - Francis Thien
- Respiratory Medicine Eastern HealthBox Hill and Monash University Melbourne VIC Australia
| | - Mark Hew
- School of Public Health and Preventive Medicine Monash University Melbourne VIC Australia
- Allergy, Asthma and Clinical Immunology Alfred Health Melbourne VIC Australia
| | - Jennifer M. Rolland
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University Melbourne VIC Australia
| | - Robyn E. O’Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Monash University Melbourne VIC Australia
- Allergy, Asthma and Clinical Immunology Alfred Health Melbourne VIC Australia
| | - Menno C. Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia
- Allergy, Asthma and Clinical Immunology Alfred Health Melbourne VIC Australia
| |
Collapse
|
28
|
Looman KIM, van Mierlo MMF, van Zelm MC, Hu C, Duijts L, de Jongste JC, Nijsten T, Pardo LM, Kiefte-de Jong JC, Moll HA, Pasmans SGMA. Increased Th22 cell numbers in a general pediatric population with filaggrin haploinsufficiency: The Generation R Study. Pediatr Allergy Immunol 2021; 32:1360-1368. [PMID: 33715246 PMCID: PMC8451856 DOI: 10.1111/pai.13502] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/09/2021] [Indexed: 12/15/2022]
Abstract
BACKGROUND Mutations in the filaggrin gene (FLG) affect epidermal barrier function and increase the risk of atopic dermatitis (AD). We hypothesized that FLG mutations affect immune cell composition in a general pediatric population. Therefore, we investigated whether school-aged children with and without FLG mutations have differences in T- and B-cell subsets. METHODS This study was embedded in a population-based prospective cohort study, the Generation R Study, and included 523 children of European genetic ancestry aged 10 years. The most common FLG mutations in the European population (R501X, S1085CfsX36, R2447X, and S3247X) were genotyped. Additionally, 11-color flow cytometry was performed on peripheral blood samples to determine helper T (Th), regulatory T (Treg), and CD27+ and CD27- memory B cells. Subset analysis was performed in 358 non-AD and 102 AD cases, assessed by parental questionnaires. RESULTS FLG mutations were observed in 8.4% of the total population and in 15.7% of the AD cases. Children with any FLG mutation had higher Th22 cell numbers compared to FLG wild-type children in the general and non-AD population. Children with and without FLG mutations had no difference in Th1, Th2, Th17, Treg, or memory B-cell numbers. Furthermore, in children with AD, FLG mutation carriership was not associated with differences in T- and B-cell subsets. CONCLUSIONS School-aged children of a general population with FLG mutations have higher Th22 cell numbers, which reflects the immunological response to the skin barrier dysfunction. FLG mutations did not otherwise affect the composition of the adaptive immunity in this general pediatric population.
Collapse
Affiliation(s)
- Kirsten I M Looman
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of General Pediatrics, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Minke M F van Mierlo
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Menno C van Zelm
- Department Immunology and Pathology, Central Clinical School, Monash University and The Alfred Hospital, Melbourne, VIC, Australia
| | - Chen Hu
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Dermatology, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Liesbeth Duijts
- Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Neonatology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Johan C de Jongste
- The Generation R Study Group, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands.,Department of Pediatrics, Division of Respiratory Medicine and Allergology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Tamar Nijsten
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Luba M Pardo
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| | - Jessica C Kiefte-de Jong
- Department of Public Health and Primary Care, Leiden University Medical Center/LUMC Campus, Leiden, The Netherlands
| | - Henriëtte A Moll
- Department of General Pediatrics, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital, Rotterdam, The Netherlands
| | - Suzanne G M A Pasmans
- Department of Dermatology, Erasmus MC, University Medical Center Rotterdam-Sophia Children's Hospital-Center of Pediatric Dermatology, Rotterdam, The Netherlands
| |
Collapse
|
29
|
Więsik-Szewczyk E, Rutkowska E, Kwiecień I, Korzeniowska M, Sołdacki D, Jahnz-Różyk K. Patients with Common Variable Immunodeficiency Complicated by Autoimmune Phenomena Have Lymphopenia and Reduced Treg, Th17, and NK Cells. J Clin Med 2021; 10:3356. [PMID: 34362140 PMCID: PMC8348468 DOI: 10.3390/jcm10153356] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 12/11/2022] Open
Abstract
Most patients with primary immune deficiency suffer from recurrent infections; however, paradoxical autoimmune phenomena can also manifest. The aim of this study was to identify immunological markers of autoimmune phenomena associated with common variable immunodeficiency (CVID). The study included 33 adults with CVID divided into two groups: (1) those with noninfectious autoimmune complications (CVID-C (n = 24)) and (2) those with only infectious symptoms (CVID-OI (n = 9)). Flow cytometry of peripheral blood was performed and compared with systemic lupus erythematosus (SLE) patients (n = 17) and healthy controls (n = 20). We found that all lymphocytes were lower in CVID-C and SLE. NK cells were lowest in CVID-C. Th17 cells were significantly reduced in CVID-C and SLE. Tregs were significantly lower in CVID-C and SLE. Bregs did not significantly differ between any groups. Class-switched memory B cells were significantly lower in CVID-C and CVID-OI. Lastly, plasmablasts were significantly higher in SLE. Among the T cell subsets, CVID-C patients had lower naive and recent thymic emigrant CD4+ T cells. In conclusion, reduced Treg, Th17, and NK cells are features of CVID with autoimmune complications, and class-switched memory B cells can help distinguish patients with different causes of autoimmunity. Future studies are needed to confirm whether reductions of Treg, Th17, and NK cells might be a biomarker of more complicated CVID cases.
Collapse
Affiliation(s)
- Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
| | - Elżbieta Rutkowska
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (E.R.); (I.K.)
| | - Iwona Kwiecień
- Laboratory of Hematology and Flow Cytometry, Department of Internal Medicine and Hematology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (E.R.); (I.K.)
| | - Marcelina Korzeniowska
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
| | - Dariusz Sołdacki
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
- Department of Clinical Immunology, Medical University of Warsaw, 02-691 Warsaw, Poland
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Military Institute of Medicine, Szaserów 128, 04-141 Warsaw, Poland; (M.K.); (D.S.); (K.J.-R.)
| |
Collapse
|
30
|
Tofighi Zavareh F, Mirshafiey A, Yazdani R, Keshtkar AA, Abolhassani H, Bagheri Y, Rezaei A, Delavari S, Rezaei N, Aghamohammadi A. Lymphocytes subsets in correlation with clinical profile in CVID patients without monogenic defects. Expert Rev Clin Immunol 2021; 17:1041-1051. [PMID: 34252322 DOI: 10.1080/1744666x.2021.1954908] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Objectives: Common variable immunodeficiency (CVID) patients experience clinical manifestations rather than recurrent respiratory infections including autoimmunity, enteropathy, and lymphoproliferation. We evaluated the correlation of lymphocyte subpopulations with such manifestations.Methods: Twenty-six genetically unsolved CVID patients were subdivided into four phenotypes: infection only (IO), autoimmunity (AI), chronic enteropathy (CE), and lymphoproliferative disorders (LP) and examined for lymphocyte subsets by flow cytometry and TCD4+ proliferation by Carboxyfluorescein succinimidyl ester (CFSE) test.Results: We detected reduced memory B and increased total, effector memory (EM), cytotoxic, and activated TCD8+ in IO, AI and CE, decreased plasmablasts, total and naive TCD4+, Regulatory TCD4+ (Treg) and naive TCD8+ in IO and CE, elevated CD21low B and terminally differentiated effector memory (TEMRA) TCD8+ in IO and AI, increased helper T (Th2) and Th17 in IO, decreased Th1 in AI and defective total and naive B and central memory (CM) TCD4+ in CE. IO showed reduced TCD4+ proliferation response.Conclusions: In genetically unsolved CVID patients, increased Th2 and Th17 and reduced Treg is associated with IO, increased CD21low B and TEMRA TCD8+ and reduced Th1 is contributed to AI and reduced total and naive B, CM TCD4+ and naive TCD8+ and expanded total TCD8+ is correlated with CE.
Collapse
Affiliation(s)
- Farzaneh Tofighi Zavareh
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran.,Primary Immunodeficiency Diseases Network (Pidnet), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Abbas Mirshafiey
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Department of Immunology, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Reza Yazdani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Ali Keshtkar
- Department of Health Sciences Education Development, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Yasser Bagheri
- Clinical Research Development Unit (CRDU), 5 Azar Hospital, Golestan University of Medical Sciences, Gorgan, Iran
| | - Arezou Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Samaneh Delavari
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Nima Rezaei
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.,Primary Immunodeficiency Diseases Network (Pidnet), Universal Scientific Education and Research Network (USERN), Tehran, Iran.,Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
31
|
Berbers RM, van der Wal MM, van Montfrans JM, Ellerbroek PM, Dalm VASH, van Hagen PM, Leavis HL, van Wijk F. Chronically Activated T-cells Retain Their Inflammatory Properties in Common Variable Immunodeficiency. J Clin Immunol 2021; 41:1621-1632. [PMID: 34247288 PMCID: PMC8452589 DOI: 10.1007/s10875-021-01084-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Accepted: 06/07/2021] [Indexed: 12/04/2022]
Abstract
Purpose Immune dysregulation complications cause significant morbidity and mortality in common variable immunodeficiency (CVID), but the underlying pathophysiology is poorly understood. While CVID is primarily considered a B-cell defect, resulting in the characteristic hypogammaglobulinemia, T-cells may also contribute to immune dysregulation complications. Here, we aim to further characterize T-cell activation and regulation in CVID with immune dysregulation (CVIDid). Methods Flow cytometry was performed to investigate T-cell differentiation, activation and intracellular cytokine production, negative regulators of immune activation, regulatory T-cells (Treg), and homing markers in 12 healthy controls, 12 CVID patients with infections only (CVIDio), and 20 CVIDid patients. Results Both CD4 + and CD8 + T-cells in CVIDid showed an increased activation profile (HLA-DR + , Ki67 + , IFNγ +) when compared to CVIDio, with concomitant upregulation of negative regulators of immune activation PD1, LAG3, CTLA4, and TIGIT. PD1 + and LAG3 + subpopulations contained equal or increased frequencies of cells with the capacity to produce IFNγ, Ki67, and/or GzmB. The expression of PD1 correlated with serum levels of CXCL9, 10, and 11. Treg frequencies were normal to high in CVIDid, but CVIDid Tregs had reduced CTLA-4 expression, especially on CD27 + effector Tregs. Increased migratory capacity to inflamed and mucosal tissue was also observed in CVIDid T-cells. Conclusion CVIDid was characterized by chronic activation of peripheral T-cells with preserved inflammatory potential rather than functional exhaustion, and increased tissue migratory capacity. While Treg numbers were normal in CVIDid Tregs, low levels of CTLA-4 indicate possible Treg dysfunction. Combined studies of T-cell dysfunction and circulating inflammatory proteins may direct future treatment strategies. Supplementary Information The online version contains supplementary material available at 10.1007/s10875-021-01084-6.
Collapse
Affiliation(s)
- Roos-Marijn Berbers
- Department of Rheumatology and Clinical Immunology, University Medical Center and Utrecht University, Utrecht, The Netherlands
| | - M Marlot van der Wal
- Center for Translational Immunology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Pauline M Ellerbroek
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Virgil A S H Dalm
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Immunology, Academic Center for Rare Immunological Diseases (RIDC), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine, Division of Clinical Immunology, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,Department of Immunology, Academic Center for Rare Immunological Diseases (RIDC), Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - Helen L Leavis
- Department of Rheumatology and Clinical Immunology, University Medical Center and Utrecht University, Utrecht, The Netherlands.
| | - Femke van Wijk
- Center for Translational Immunology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| |
Collapse
|
32
|
Edwards ESJ, Bosco JJ, Ojaimi S, O'Hehir RE, van Zelm MC. Beyond monogenetic rare variants: tackling the low rate of genetic diagnoses in predominantly antibody deficiency. Cell Mol Immunol 2021; 18:588-603. [PMID: 32801365 PMCID: PMC8027216 DOI: 10.1038/s41423-020-00520-8] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Accepted: 07/26/2020] [Indexed: 02/07/2023] Open
Abstract
Predominantly antibody deficiency (PAD) is the most prevalent form of primary immunodeficiency, and is characterized by broad clinical, immunological and genetic heterogeneity. Utilizing the current gold standard of whole exome sequencing for diagnosis, pathogenic gene variants are only identified in less than 20% of patients. While elucidation of the causal genes underlying PAD has provided many insights into the cellular and molecular mechanisms underpinning disease pathogenesis, many other genes may remain as yet undefined to enable definitive diagnosis, prognostic monitoring and targeted therapy of patients. Considering that many patients display a relatively late onset of disease presentation in their 2nd or 3rd decade of life, it is questionable whether a single genetic lesion underlies disease in all patients. Potentially, combined effects of other gene variants and/or non-genetic factors, including specific infections can drive disease presentation. In this review, we define (1) the clinical and immunological variability of PAD, (2) consider how genetic defects identified in PAD have given insight into B-cell immunobiology, (3) address recent technological advances in genomics and the challenges associated with identifying causal variants, and (4) discuss how functional validation of variants of unknown significance could potentially be translated into increased diagnostic rates, improved prognostic monitoring and personalized medicine for PAD patients. A multidisciplinary approach will be the key to curtailing the early mortality and high morbidity rates in this immune disorder.
Collapse
Affiliation(s)
- Emily S J Edwards
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia
| | - Julian J Bosco
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University and Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| | - Samar Ojaimi
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia
- Department of Infectious Diseases, Monash Health, Clayton, VIC, Australia
- Centre for Inflammatory Diseases, Monash Health, Clayton, VIC, Australia
- Department of Allergy and Immunology, Monash Health, Clayton, VIC, Australia
| | - Robyn E O'Hehir
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University and Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia.
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies, Melbourne, VIC, Australia.
- Department of Allergy, Immunology and Respiratory Medicine, Central Clinical School, Monash University and Allergy, Asthma and Clinical Immunology Service, Alfred Hospital, Melbourne, VIC, Australia.
| |
Collapse
|
33
|
Hartley GE, Edwards ESJ, Aui PM, Varese N, Stojanovic S, McMahon J, Peleg AY, Boo I, Drummer HE, Hogarth PM, O'Hehir RE, van Zelm MC. Rapid generation of durable B cell memory to SARS-CoV-2 spike and nucleocapsid proteins in COVID-19 and convalescence. Sci Immunol 2021; 5:5/54/eabf8891. [PMID: 33443036 PMCID: PMC7877496 DOI: 10.1126/sciimmunol.abf8891] [Citation(s) in RCA: 202] [Impact Index Per Article: 67.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 12/18/2020] [Indexed: 12/13/2022]
Abstract
Lasting immunity following SARS-CoV-2 infection is questioned because serum antibodies decline in convalescence. However, functional immunity is mediated by long-lived memory T and B (Bmem) cells. Therefore, we generated fluorescently-labeled tetramers of the spike receptor binding domain (RBD) and nucleocapsid protein (NCP) to determine the longevity and immunophenotype of SARS-CoV-2-specific Bmem cells in COVID-19 patients. A total of 36 blood samples were obtained from 25 COVID-19 patients between 4 and 242 days post-symptom onset including 11 paired samples. While serum IgG to RBD and NCP was identified in all patients, antibody levels began declining at 20 days post-symptom onset. RBD- and NCP-specific Bmem cells predominantly expressed IgM+ or IgG1+ and continued to rise until 150 days. RBD-specific IgG+ Bmem were predominantly CD27+, and numbers significantly correlated with circulating follicular helper T cell numbers. Thus, the SARS-CoV-2 antibody response contracts in convalescence with persistence of RBD- and NCP-specific Bmem cells. Flow cytometric detection of SARS-CoV-2-specific Bmem cells enables detection of long-term immune memory following infection or vaccination for COVID-19.
Collapse
Affiliation(s)
- Gemma E Hartley
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Emily S J Edwards
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Pei M Aui
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia
| | - Nirupama Varese
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Department of Allergy, Immunology & Respiratory Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Stephanie Stojanovic
- Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, VIC, Australia
| | - James McMahon
- Department of Infectious Diseases, The Alfred and Central Clinical school, Monash University, Melbourne, VIC, Australia.,Department of Infectious Diseases, Monash Health, Melbourne, VIC, Australia
| | - Anton Y Peleg
- Department of Infectious Diseases, The Alfred and Central Clinical school, Monash University, Melbourne, VIC, Australia.,Infection and Immunity Program, Monash Biomedicine Discovery Institute, Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - Irene Boo
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia
| | - Heidi E Drummer
- Viral Entry and Vaccines Group, Burnet Institute, Melbourne, VIC, Australia.,Department of Microbiology and Immunology, Peter Doherty Institute for Infection and Immunity, University of Melbourne, Melbourne, VIC, Australia.,Department of Microbiology, Monash University, Clayton, VIC, Australia
| | - P Mark Hogarth
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Immune Therapies Group, Burnet Institute, Melbourne, VIC, Australia.,Department of Pathology, The University of Melbourne, Parkville, VIC, Australia
| | - Robyn E O'Hehir
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia.,Department of Allergy, Immunology & Respiratory Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia.,Allergy, Asthma and Clinical Immunology, Alfred Health, Melbourne, VIC, Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University, Melbourne, VIC, Australia. .,Department of Allergy, Immunology & Respiratory Medicine, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
34
|
Kim JH, Jang JH, Lee SH, Yang EM, Jang SH, Jung KS, Park HS. Specific Antibody Deficiency in Adult Patients With IgG or IgG Subclass Deficiency. ALLERGY, ASTHMA & IMMUNOLOGY RESEARCH 2021; 13:271-283. [PMID: 33474861 PMCID: PMC7840874 DOI: 10.4168/aair.2021.13.2.271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Revised: 07/24/2020] [Accepted: 07/31/2020] [Indexed: 11/23/2022]
Abstract
Purpose Specific antibody deficiency (SAD) involves a deficient response to a polysaccharide vaccine despite having normal immunoglobulin levels. The failure of the polysaccharide response can be observed as a component of various primary antibody deficiencies. However, only a few studies have described the clinical and immunological profiles in SAD and/or other primary immunodeficiencies (PIDs) in adults. Methods A total of 47 patients who had a clinical history suggestive of antibody deficiency or had already been diagnosed with various antibody deficiencies were enrolled. Polysaccharide responses to 7 pneumococcal serotypes (4, 6B, 9V, 14, 18C, 19F and 23F) were measured using the World Health Organization enzyme-linked immunosorbent assay (WHO-ELISA), and postvaccination immunoglobulin G (IgG) titers were compared to clinical and laboratory parameters. Results Based on the American Academy of Allergy, Asthma, and Immunology (AAAAI) criteria for the WHO-ELISA, 11 (23.4%) patients were diagnosed as having SAD. Sixteen-three percent of them had combined with other types of PID, such as IgG subclass deficiency and hypogammaglobulinemia. Postvaccination IgG titers for the serotypes 4/9V/18C correlated with IgG2 (P = 0.012, P = 0.001, and P = 0.004) and for 6B/9V/14 with IgG3 (P = 0.003, P = 0.041, and P = 0.036, respectively). The IgG3 subclass levels negatively correlated with forced expiratory volume in 1 second (FEV1, %) and FEV1/forced vital capacity (P < 0.001 and P = 0.001, respectively). Conclusion SAD can be diagnosed in patients with normal IgG levels as well as in those deficient in IgG or the IgG3 subclass, implicating that restricted responses to Streptococcus pneumoniae polysaccharide antigens commonly exist in patients with predominantly antibody deficiency.
Collapse
Affiliation(s)
- Joo Hee Kim
- Department of Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Jae Hyuk Jang
- Department of Allergy & Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - So Hee Lee
- Department of Allergy & Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Eun Mi Yang
- Department of Allergy & Clinical Immunology, Ajou University School of Medicine, Suwon, Korea
| | - Seung Hun Jang
- Department of Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Ki Suck Jung
- Department of Medicine, Hallym University Sacred Heart Hospital, Hallym University College of Medicine, Anyang, Korea
| | - Hae Sim Park
- Department of Allergy & Clinical Immunology, Ajou University School of Medicine, Suwon, Korea.
| |
Collapse
|
35
|
Berbers RM, Drylewicz J, Ellerbroek PM, van Montfrans JM, Dalm VASH, van Hagen PM, Keller B, Warnatz K, van de Ven A, van Laar JM, Nierkens S, Leavis HL. Targeted Proteomics Reveals Inflammatory Pathways that Classify Immune Dysregulation in Common Variable Immunodeficiency. J Clin Immunol 2020; 41:362-373. [PMID: 33190167 PMCID: PMC7858548 DOI: 10.1007/s10875-020-00908-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022]
Abstract
Patients with common variable immunodeficiency (CVID) can develop immune dysregulation complications such as autoimmunity, lymphoproliferation, enteritis, and malignancy, which cause significant morbidity and mortality. We aimed to (i) assess the potential of serum proteomics in stratifying patients with immune dysregulation using two independent cohorts and (ii) identify cytokine and chemokine signaling pathways that underlie immune dysregulation in CVID. A panel of 180 markers was measured in two multicenter CVID cohorts using Olink Protein Extension Assay technology. A classification algorithm was trained to distinguish CVID with immune dysregulation (CVIDid, n = 14) from CVID with infections only (CVIDio, n = 16) in the training cohort, and validated on a second testing cohort (CVIDid n = 23, CVIDio n = 24). Differential expression in both cohorts was used to determine relevant signaling pathways. An elastic net classifier using MILR1, LILRB4, IL10, IL12RB1, and CD83 could discriminate between CVIDid and CVIDio patients with a sensitivity of 0.83, specificity of 0.75, and area under the curve of 0.73 in an independent testing cohort. Activated pathways (fold change > 1.5, FDR-adjusted p < 0.05) in CVIDid included Th1 and Th17-associated signaling, as well as IL10 and other immune regulatory markers (LAG3, TNFRSF9, CD83). Targeted serum proteomics provided an accurate and reproducible tool to discriminate between patients with CVIDid and CVIDio. Cytokine profiles provided insight into activation of Th1 and Th17 pathways and indicate a possible role for chronic inflammation and exhaustion in immune dysregulation. These findings serve as a first step towards the development of biomarkers for immune dysregulation in CVID.
Collapse
Affiliation(s)
- Roos-Marijn Berbers
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Julia Drylewicz
- Center for Translational Immunology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Pauline M Ellerbroek
- Department of Internal Medicine and Infectious Diseases, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Joris M van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Virgil A S H Dalm
- Department of Internal Medicine, Division of Clinical Immunology; Department of Immunology; Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - P Martin van Hagen
- Department of Internal Medicine, Division of Clinical Immunology; Department of Immunology; Academic Center for Rare Immunological Diseases (RIDC), Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - 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 (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - 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 (CCI), Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Annick van de Ven
- Departments of Internal Medicine and Allergology, Rheumatology and Clinical Immunology, University Medical Center Groningen, Groningen, The Netherlands
| | - Jaap M van Laar
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - Stefan Nierkens
- Center for Translational Immunology, University Medical Center Utrecht and Utrecht University, Utrecht, The Netherlands
| | - Helen L Leavis
- Department of Rheumatology & Clinical Immunology, University Medical Center Utrecht and Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Hartley GE, Edwards ESJ, Bosco JJ, Ojaimi S, Stirling RG, Cameron PU, Flanagan K, Plebanski M, Hogarth PM, O'Hehir RE, van Zelm MC. Influenza-specific IgG1 + memory B-cell numbers increase upon booster vaccination in healthy adults but not in patients with predominantly antibody deficiency. Clin Transl Immunology 2020; 9:e1199. [PMID: 33088507 PMCID: PMC7563650 DOI: 10.1002/cti2.1199] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 09/15/2020] [Accepted: 09/28/2020] [Indexed: 12/18/2022] Open
Abstract
Background Annual influenza vaccination is recommended to all individuals over 6 months of age, including predominantly antibody deficiency (PAD) patients. Vaccination responses are typically evaluated by serology, and because PAD patients are by definition impaired in generating IgG and receive immunoglobulin replacement therapy (IgRT), it remains unclear whether they can mount an antigen-specific response. Objective To quantify and characterise the antigen-specific memory B (Bmem) cell compartment in healthy controls and PAD patients following an influenza booster vaccination. Methods Recombinant hemagglutinin (HA) from the A/Michigan/2015 H1N1 (AM15) strain with an AviTag was generated in a mammalian cell line, and following targeted biotinylation, was tetramerised with BUV395 or BUV737 streptavidin conjugates. Multicolour flow cytometry was applied on blood samples before and 28 days after booster influenza vaccination in 16 healthy controls and five PAD patients with circulating Bmem cells. Results Recombinant HA tetramers were specifically recognised by 0.5-1% of B cells in previously vaccinated healthy adults. HA-specific Bmem cell numbers were significantly increased following booster vaccination and predominantly expressed IgG1. Similarly, PAD patients carried HA-specific Bmem cells, predominantly expressing IgG1. However, these numbers were lower than in controls and did not increase following booster vaccination. Conclusion We have successfully identified AM15-specific Bmem cells in healthy controls and PAD patients. The presence of antigen-specific Bmem cells could offer an additional diagnostic tool to aid in the clinical diagnosis of PAD. Furthermore, alterations in the number or immunophenotype of HA-specific Bmem cells post-booster vaccination could assist in the evaluation of immune responses in individuals receiving IgRT.
Collapse
Affiliation(s)
- Gemma E Hartley
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia
| | - Emily S J Edwards
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia
| | - Julian J Bosco
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia.,Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Alfred Hospital Monash University and Allergy, Asthma and Clinical Immunology Service Melbourne VIC Australia
| | - Samar Ojaimi
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia.,Infectious Diseases Monash Health Clayton VIC Australia.,Immunology Laboratory Monash Pathology Clayton VIC Australia.,Allergy and Immunology Monash Health Clayton VIC Australia
| | - Robert G Stirling
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia.,Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Alfred Hospital Monash University and Allergy, Asthma and Clinical Immunology Service Melbourne VIC Australia
| | - Paul U Cameron
- The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia.,Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Alfred Hospital Monash University and Allergy, Asthma and Clinical Immunology Service Melbourne VIC Australia
| | - Katie Flanagan
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia.,School of Medicine University of Tasmania Launceston TAS Australia.,School of Health and Biomedical Sciences RMIT Bundoora VIC Australia
| | | | - Philip Mark Hogarth
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia.,Immune Therapies Group Burnet Institute Melbourne VIC Australia
| | - Robyn E O'Hehir
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia.,Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Alfred Hospital Monash University and Allergy, Asthma and Clinical Immunology Service Melbourne VIC Australia
| | - Menno C van Zelm
- Department of Immunology and Pathology Central Clinical School Monash University Melbourne VIC Australia.,The Jeffrey Modell Diagnostic and Research Centre for Primary Immunodeficiencies Melbourne VIC Australia.,Department of Allergy, Immunology and Respiratory Medicine Central Clinical School Alfred Hospital Monash University and Allergy, Asthma and Clinical Immunology Service Melbourne VIC Australia
| |
Collapse
|
37
|
Więsik-Szewczyk E, Jahnz-Różyk K. From infections to autoimmunity: Diagnostic challenges in common variable immunodeficiency. World J Clin Cases 2020; 8:3942-3955. [PMID: 33024751 PMCID: PMC7520788 DOI: 10.12998/wjcc.v8.i18.3942] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Revised: 05/29/2020] [Accepted: 08/26/2020] [Indexed: 02/05/2023] Open
Abstract
Common variable immunodeficiency (CVID) is the most common clinically significant primary antibody deficiency diagnosed in adults. The early symptoms are not specific. They include common infections, mainly of the respiratory tract, caused by typical microorganisms, so cases can be missed in primary care. In the majority of patients increased susceptibility to infections coexists with signs or symptoms of autoimmunity, inflammation or polyclonal lymphoproliferation, which can divert diagnosis from immune deficiency. The overall incidence of malignancy is increased in CVID and certain cancers are significantly more common. Lymphomas and gastric carcinoma are the most frequently reported malignancies in CVID, so a high index of suspicion is recommended. Diagnostic delay in CVID is seen worldwide. The main goal of this paper is to increase the awareness about CVID among health care professionals. We aim to present features which can be helpful in CVID diagnosis in order to shorten the “latency” of proper management of CVID patients. We review clinical symptoms, complications and laboratory abnormalities of CVID. Immunoglobulin replacement therapy is regarded as the cornerstone of pharmacological intervention. New modes of Ig application, mainly subcutaneously and via the hyaluronidase-facilitated subcutaneous route, help to adjust therapy to patients’ needs and preferences. Still there remain unmet needs. It remains to be seen whether CVID complications can be avoided by earlier diagnosis, treatment and thorough monitoring in the context of increased risk of malignancy. Development of patient tailored protocols depending on the clinical phenotype and risk factors might be more appropriate. The most important consideration is to diagnose suspected cases and stratify patients in a precise and timely way. Work is needed to define features predictive of unfavorable prognosis.
Collapse
Affiliation(s)
- Ewa Więsik-Szewczyk
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Central Clinical Hospital of the Ministry of National Defense, Military Institute of Medicine, Warsaw 04-141, Poland
| | - Karina Jahnz-Różyk
- Department of Internal Medicine, Pulmonology, Allergy and Clinical Immunology, Central Clinical Hospital of the Ministry of National Defense, Military Institute of Medicine, Warsaw 04-141, Poland
| |
Collapse
|
38
|
Banday AZ, Jindal AK, Arora K, Rawat A. Extensive Molluscum Contagiosum in X-Linked Agammaglobulinemia. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2020; 9:985. [PMID: 32811796 DOI: 10.1016/j.jaip.2020.07.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Revised: 07/21/2020] [Accepted: 07/22/2020] [Indexed: 11/25/2022]
Affiliation(s)
- Aaqib Zaffar Banday
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Ankur Kumar Jindal
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| | - Kanika Arora
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
39
|
Tang Y, He H, Hu P, Xu X. T lymphocytes in IgA nephropathy. Exp Ther Med 2020; 20:186-194. [PMID: 32509008 PMCID: PMC7271719 DOI: 10.3892/etm.2020.8673] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2019] [Accepted: 03/19/2020] [Indexed: 02/07/2023] Open
Abstract
Immunoglobulin A nephropathy (IgAN), the most common primary glomerulonephritis worldwide, is the main cause of end-stage renal disease. IgAN is characterized by the accumulation of immune complexes in the circulation, which contain abnormal levels of IgA. IgAN primarily results from galactose-deficient IgA1 (Gd-IgA1) and Gd-IgA1 deposition in the renal mesangium, causing local proliferation and matrix expansion. Gd-IgA1 has been confirmed as one of the key effectors in the pathogenesis of IgAN, but the origin of Gd-IgA1 is not clear. Recent studies have shown that Gd-IgA1 deposition could be the result of mucosally primed plasma cells and is associated with T cell dysregulation. T cells contribute to the IgA response and play an important role in the development of IgAN. In the present review, the latest discoveries regarding the role of T lymphocytes in the pathogenesis of IgAN have been summarized. Understanding these advances will allow novel therapeutic strategies for the treatment of IgAN.
Collapse
Affiliation(s)
- Yuyan Tang
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Haidong He
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Pin Hu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| | - Xudong Xu
- Department of Nephrology, Minhang Hospital, Fudan University, Shanghai 201199, P.R. China
| |
Collapse
|
40
|
Grosserichter-Wagener C, Franco-Gallego A, Ahmadi F, Moncada-Vélez M, Dalm VA, Rojas JL, Orrego JC, Correa Vargas N, Hammarström L, Schreurs MW, Dik WA, van Hagen PM, Boon L, van Dongen JJ, van der Burg M, Pan-Hammarström Q, Franco JL, van Zelm MC. Defective formation of IgA memory B cells, Th1 and Th17 cells in symptomatic patients with selective IgA deficiency. Clin Transl Immunology 2020; 9:e1130. [PMID: 32355559 PMCID: PMC7190975 DOI: 10.1002/cti2.1130] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 03/12/2020] [Accepted: 03/30/2020] [Indexed: 12/16/2022] Open
Abstract
Objective Selective IgA deficiency (sIgAD) is the most common primary immunodeficiency in Western countries. Patients can suffer from recurrent infections and autoimmune diseases because of a largely unknown aetiology. To increase insights into the pathophysiology of the disease, we studied memory B and T cells and cytokine concentrations in peripheral blood. Methods We analysed 30 sIgAD patients (12 children, 18 adults) through detailed phenotyping of peripheral B‐cell, CD8+ T‐cell and CD4+ T‐cell subsets, sequence analysis of IGA and IGG transcripts, in vitro B‐cell activation and blood cytokine measurements. Results All patients had significantly decreased numbers of T‐cell‐dependent (TD; CD27+) and T‐cell‐independent (TI; CD27−) IgA memory B cells and increased CD21low B‐cell numbers. IgM+IgD− memory B cells were decreased in children and normal in adult patients. IGA and IGG transcripts contained normal SHM levels. In sIgAD children, IGA transcripts more frequently used IGA2 than controls (58.5% vs. 25.1%), but not in adult patients. B‐cell activation after in vitro stimulation was normal. However, adult sIgAD patients exhibited increased blood levels of TGF‐β1, BAFF and APRIL, whereas they had decreased Th1 and Th17 cell numbers. Conclusion Impaired IgA memory formation in sIgAD patients is not due to a B‐cell activation defect. Instead, decreased Th1 and Th17 cell numbers and high blood levels of BAFF, APRIL and TGF‐β1 might reflect disturbed regulation of IgA responses in vivo. These insights into B‐cell extrinsic immune defects suggest the need for a broader immunological focus on genomics and functional analyses to unravel the pathogenesis of sIgAD.
Collapse
Affiliation(s)
| | | | - Fatemeh Ahmadi
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
| | - Marcela Moncada-Vélez
- Grupo de Inmunodeficiencias Primarias Universidad de Antioquia UdeA Medellín Colombia
| | - Virgil Ash Dalm
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands.,Department of Internal Medicine Erasmus MC University Medical Center Rotterdam The Netherlands
| | - Jessica Lineth Rojas
- Grupo de Inmunodeficiencias Primarias Universidad de Antioquia UdeA Medellín Colombia
| | - Julio César Orrego
- Grupo de Inmunodeficiencias Primarias Universidad de Antioquia UdeA Medellín Colombia
| | - Natalia Correa Vargas
- Grupo de Inmunodeficiencias Primarias Universidad de Antioquia UdeA Medellín Colombia
| | - Lennart Hammarström
- Clinical Immunology Department of Laboratory Medicine Karolinska Institutet at Karolinska University Hospital Huddinge Sweden
| | - Marco Wj Schreurs
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
| | - Willem A Dik
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands
| | - P Martin van Hagen
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands.,Department of Internal Medicine Erasmus MC University Medical Center Rotterdam The Netherlands
| | | | - Jacques Jm van Dongen
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands.,Department of Immunohematology and Blood Transfusion Leiden University Medical Center Leiden The Netherlands
| | - Mirjam van der Burg
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands.,Laboratory for Immunology Department of Pediatrics Leiden University Medical Center Leiden The Netherlands
| | - Qiang Pan-Hammarström
- Clinical Immunology Department of Laboratory Medicine Karolinska Institutet at Karolinska University Hospital Huddinge Sweden
| | - José L Franco
- Grupo de Inmunodeficiencias Primarias Universidad de Antioquia UdeA Medellín Colombia
| | - Menno C van Zelm
- Department of Immunology Erasmus MC University Medical Center Rotterdam The Netherlands.,Department of Immunology and Pathology Central Clinical School Monash University and The Alfred Hospital Melbourne VIC Australia.,The Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies in Melbourne Melbourne VIC Australia
| |
Collapse
|