101
|
Königs C, Schultze-Strasser S, Quaiser A, Bochennek K, Schwabe D, Klingebiel TE, Koehl U, Cappel C, Rolle U, Bader P, Bremm M, Huenecke S, Bakhtiar S. An Exponential Regression Model Reveals the Continuous Development of B Cell Subpopulations Used as Reference Values in Children. Front Pediatr 2018; 6:121. [PMID: 29780793 PMCID: PMC5945839 DOI: 10.3389/fped.2018.00121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Accepted: 04/13/2018] [Indexed: 11/13/2022] Open
Abstract
B lymphocytes are key players in humoral immunity, expressing diverse surface immunoglobulin receptors directed against specific antigenic epitopes. The development and profile of distinct subpopulations have gained awareness in the setting of primary immunodeficiency disorders, primary or secondary autoimmunity and as therapeutic targets of specific antibodies in various diseases. The major B cell subpopulations in peripheral blood include naïve (CD19+ or CD20+IgD+CD27-), non-switched memory (CD19+ or CD20+IgD+CD27+) and switched memory B cells (CD19+ or CD20+IgD-CD27+). Furthermore, less common B cell subpopulations have also been described as having a role in the suppressive capacity of B cells to maintain self-tolerance. Data on reference values for B cell subpopulations are limited and only available for older age groups, neglecting the continuous process of human B cell development in children and adolescents. This study was designed to establish an exponential regression model to produce continuous reference values for main B cell subpopulations to reflect the dynamic maturation of the human immune system in healthy children.
Collapse
Affiliation(s)
- Christoph Königs
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | | | - Andrea Quaiser
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Konrad Bochennek
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Dirk Schwabe
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Thomas E. Klingebiel
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Ulrike Koehl
- GMP Development, Integriertes Forschungs- und Behandlungszentrum Transplantation (IFB-TX), Hannover Medical School, Institute of Cellular Therapeutics, Hannover, Germany
| | - Claudia Cappel
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Udo Rolle
- Department of Pediatric Surgery, University Hospital Frankfurt, Frankfurt, Germany
| | - Peter Bader
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Melanie Bremm
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Sabine Huenecke
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| | - Shahrzad Bakhtiar
- Department of Pediatric and Adolescent Medicine, University Hospital Frankfurt, Frankfurt, Germany
| |
Collapse
|
102
|
June CH, O'Connor RS, Kawalekar OU, Ghassemi S, Milone MC. CAR T cell immunotherapy for human cancer. Science 2018; 359:1361-1365. [PMID: 29567707 DOI: 10.1126/science.aar6711] [Citation(s) in RCA: 2014] [Impact Index Per Article: 287.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Adoptive T cell transfer (ACT) is a new area of transfusion medicine involving the infusion of lymphocytes to mediate antitumor, antiviral, or anti-inflammatory effects. The field has rapidly advanced from a promising form of immuno-oncology in preclinical models to the recent commercial approvals of chimeric antigen receptor (CAR) T cells to treat leukemia and lymphoma. This Review describes opportunities and challenges for entering mainstream oncology that presently face the CAR T field, with a focus on the challenges that have emerged over the past several years.
Collapse
Affiliation(s)
- Carl H June
- Center for Cellular Immunotherapies, Perlman School of Medicine, Philadelphia, PA, USA. .,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perlman School of Medicine, Philadelphia, PA, USA
| | - Roddy S O'Connor
- Center for Cellular Immunotherapies, Perlman School of Medicine, Philadelphia, PA, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Omkar U Kawalekar
- Center for Cellular Immunotherapies, Perlman School of Medicine, Philadelphia, PA, USA
| | - Saba Ghassemi
- Center for Cellular Immunotherapies, Perlman School of Medicine, Philadelphia, PA, USA.,Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael C Milone
- Center for Cellular Immunotherapies, Perlman School of Medicine, Philadelphia, PA, USA.,Department of Pathology and Laboratory Medicine, Perlman School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
103
|
Abstract
Primary immunodeficiency diseases are genetic disorders that mostly cause susceptibility to infections and are sometimes associated with autoimmune and malignant diseases. For early detection and management of these diseases, flow cytometric procedures allow an encompassing assessment of cellular phenotypes and cellular functions. State-of-the art cytometry is based today on 8- to 10-color staining and includes an assessment of lineage maturation and functional markers.
Collapse
Affiliation(s)
- Andreas Boldt
- Medical Faculty, Department of Diagnostics, Institute of Clinical Immunology, University of Leipzig, Johannisallee 30, Leipzig D-04103, Germany.
| | - Michael Bitar
- Medical Faculty, Department of Diagnostics, Institute of Clinical Immunology, University of Leipzig, Johannisallee 30, Leipzig D-04103, Germany
| | - Ulrich Sack
- Medical Faculty, Department of Diagnostics, Institute of Clinical Immunology, University of Leipzig, Johannisallee 30, Leipzig D-04103, Germany
| |
Collapse
|
104
|
Forsthuber TG, Cimbora DM, Ratchford JN, Katz E, Stüve O. B cell-based therapies in CNS autoimmunity: differentiating CD19 and CD20 as therapeutic targets. Ther Adv Neurol Disord 2018; 11:1756286418761697. [PMID: 29593838 PMCID: PMC5865455 DOI: 10.1177/1756286418761697] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2017] [Accepted: 02/01/2018] [Indexed: 01/05/2023] Open
Abstract
Increasing recognition of the role of B cells in the adaptive immune response makes B cells an important therapeutic target in autoimmunity. Numerous current and developmental immunotherapies target B cells for elimination through recognition of cell-surface proteins expressed specifically on B cells, in particular CD19 and CD20. Similarities and differences in the function and expression of these two molecules predict some shared, and some distinct, pharmacological effects of agents targeting CD19 versus CD20, potentially leading to differences in the clinical safety and efficacy of such agents. Here, we review current knowledge of CD19 and CD20 function and biology, survey current and developmental therapies that target these molecules, and discuss potential differences in elimination of B cells by drugs that target CD19 versus CD20, with particular focus on the central nervous system autoimmune diseases multiple sclerosis and neuromyelitis optica. The principles and mechanisms herein discussed might also be relevant to a variety of other nervous system autoimmune disorders, including NMDA (N-methyl-D-aspartate) receptor encephalitis, transverse myelitis and myasthenia gravis.
Collapse
Affiliation(s)
| | | | | | | | - Olaf Stüve
- Neurology Section, VA North Texas Health Care System, Medical Service, Dallas, TX, USA
| |
Collapse
|
105
|
Teplyakov A, Obmolova G, Luo J, Gilliland GL. Crystal structure of B-cell co-receptor CD19 in complex with antibody B43 reveals an unexpected fold. Proteins 2018; 86:495-500. [PMID: 29490423 DOI: 10.1002/prot.25485] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2017] [Revised: 02/11/2018] [Accepted: 02/24/2018] [Indexed: 12/27/2022]
Abstract
CD19 is a transmembrane protein expressed on malignant B cells, but not in other lineages or other tissues, which makes it an attractive target for monoclonal antibody-mediated immunotherapy. Anti-CD19 antibody B43 was utilized in a bispecific T-cell engager (BiTE) blinatumomab that demonstrated potency for the treatment of relapsed acute lymphoblastic leukemia. To gain insight into the mechanism of action of the antibody, the crystal structure of B43 Fab was determined in complex with CD19 and in the unbound form. The structure revealed the binding epitope, explained the lack of cross-reactivity toward non-human species, and suggested the key-and-lock mechanism of antigen recognition. Most unexpectedly, the structure revealed a unique molecular topology of CD19. Rather than a tandem of c-type immunoglobulin folds predicted from the amino acid sequence, the extracellular domain of CD19 exhibits an elongated β-sandwich formed by two immunoglobulin folds by swapping their C-terminal halves. This is the first structure of CD19, which has no sequence homologs.
Collapse
Affiliation(s)
- Alexey Teplyakov
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| | - Galina Obmolova
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| | - Jinquan Luo
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| | - Gary L Gilliland
- Janssen Research and Development, LLC, 1400 McKean Road, Spring House, Pennsylvania, 19477
| |
Collapse
|
106
|
Marin AV, Cárdenas PP, Jiménez-Reinoso A, Muñoz-Ruiz M, Regueiro JR. Lymphocyte integration of complement cues. Semin Cell Dev Biol 2018; 85:132-142. [PMID: 29438807 DOI: 10.1016/j.semcdb.2018.02.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/08/2018] [Indexed: 12/17/2022]
Abstract
We address current data, views and puzzles on the emerging topic of regulation of lymphocytes by complement proteins or fragments. Such regulation is believed to take place through complement receptors (CR) and membrane complement regulators (CReg) involved in cell function or protection, respectively, including intracellular signalling. Original observations in B cells clearly support that complement cues through CR improve their performance. Other lymphocytes likely integrate complement-derived signals, as most lymphoid cells constitutively express or regulate CR and CReg upon activation. CR-induced signals, particularly by anaphylatoxins, clearly regulate lymphoid cell function. In contrast, data obtained by CReg crosslinking using antibodies are not always confirmed in human congenital deficiencies or knock-out mice, casting doubts on their physiological relevance. Unsurprisingly, human and mouse complement systems are not completely homologous, adding further complexity to our still fragmentary understanding of complement-lymphocyte interactions.
Collapse
Affiliation(s)
- Ana V Marin
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Paula P Cárdenas
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Anaïs Jiménez-Reinoso
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Miguel Muñoz-Ruiz
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Jose R Regueiro
- Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| |
Collapse
|
107
|
Gies V, Schickel JN, Jung S, Joublin A, Glauzy S, Knapp AM, Soley A, Poindron V, Guffroy A, Choi JY, Gottenberg JE, Anolik JH, Martin T, Soulas-Sprauel P, Meffre E, Korganow AS. Impaired TLR9 responses in B cells from patients with systemic lupus erythematosus. JCI Insight 2018. [PMID: 29515028 DOI: 10.1172/jci.insight.96795] [Citation(s) in RCA: 44] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
B cells play a central role in systemic lupus erythematosus (SLE) pathophysiology but dysregulated pathways leading to a break in B cell tolerance remain unclear. Since Toll-like receptor 9 (TLR9) favors the elimination of autoreactive B cells in the periphery, we assessed TLR9 function in SLE by analyzing the responses of B cells and plasmacytoid dendritic cells (pDCs) isolated from healthy donors and patients after stimulation with CpG, a TLR9 agonist. We found that SLE B cells from patients without hydroxychloroquine treatment displayed defective in vitro TLR9 responses, as illustrated by the impaired upregulation of B cell activation molecules and the diminished production of various cytokines including antiinflammatory IL-10. In agreement with CD19 controlling TLR9 responses in B cells, decreased expression of the CD19/CD21 complex on SLE B cells was detected as early as the transitional B cell stage. In contrast, TLR7 function was preserved in SLE B cells, whereas pDCs from SLE patients properly responded to TLR9 stimulation, thereby revealing that impaired TLR9 function in SLE was restricted to B cells. We conclude that abnormal CD19 expression and TLR9 tolerogenic function in SLE B cells may contribute to the break of B cell tolerance in these patients.
Collapse
Affiliation(s)
- Vincent Gies
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
| | - Jean-Nicolas Schickel
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sophie Jung
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,Reference Center for Oral Rare Diseases (O-Rares), "Pôle de Médecine et de Chirurgie Bucco-Dentaires, University Hospital - Faculty of Dentistry, University of Strasbourg, Strasbourg, France
| | - Aurélie Joublin
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
| | - Salomé Glauzy
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anne-Marie Knapp
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
| | - Anne Soley
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France
| | - Vincent Poindron
- Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
| | - Aurélien Guffroy
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France
| | - Jin-Young Choi
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jacques-Eric Gottenberg
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,UFR Medicine, University of Strasbourg, Strasbourg, France.,Department of Rheumatology, National Reference Center for Autoimmune Diseases, University Hospital, Strasbourg, France
| | - Jennifer H Anolik
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, University of Rochester Medical Center, Rochester, New York, USA
| | - Thierry Martin
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France.,UFR Medicine, University of Strasbourg, Strasbourg, France
| | - Pauline Soulas-Sprauel
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France.,UFR Pharmaceutical Sciences, Strasbourg University, Illkirch, France
| | - Eric Meffre
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Anne-Sophie Korganow
- CNRS UPR 3572 "Immunopathology and Therapeutic Chemistry"/Laboratory of Excellence Médalis, Institute of Molecular and Cellular Biology (IBMC), Strasbourg, France.,Department of Clinical Immunology and Internal Medicine, National Reference Center for Rare Autoimmune Diseases, University Hospital, Strasbourg, France.,UFR Medicine, University of Strasbourg, Strasbourg, France
| |
Collapse
|
108
|
Richardson AM, Moyer AM, Hasadsri L, Abraham RS. Diagnostic Tools for Inborn Errors of Human Immunity (Primary Immunodeficiencies and Immune Dysregulatory Diseases). Curr Allergy Asthma Rep 2018; 18:19. [PMID: 29470720 DOI: 10.1007/s11882-018-0770-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
PURPOSE OF REVIEW The purpose of this review is to provide an overview of diagnostic testing in primary immunodeficiency and immune dysregulatory disorders (PIDDs), particularly focusing on flow cytometry and genetic techniques, utilizing specific examples of PIDDs. RECENT FINDINGS Flow cytometry remains a vital tool in the diagnosis and monitoring of immunological diseases. Its utility ranges from cellular analysis and specific protein quantitation to functional assays and signaling pathway analysis. Mass cytometry combines flow cytometry and mass spectrometry to dramatically increase the throughput of multivariate single-cell analysis. Next-generation sequencing in combination with other molecular techniques and processing algorithms has become more widely available and identified the diverse and heterogeneous genetic underpinnings of these disorders. As the spectrum of disease is further clarified by increasing immunological, genetic, and epigenetic knowledge, the careful application of these diagnostic tools and bioinformatics will assist not only in our understanding of these complex disorders, but also enable the implementation of personalized therapeutic approaches for disease management.
Collapse
Affiliation(s)
- Annely M Richardson
- Division of Allergic Diseases, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ann M Moyer
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Linda Hasadsri
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA
| | - Roshini S Abraham
- Department of Laboratory Medicine and Pathology, Mayo Clinic, 200 1st St SW, Rochester, MN, 55905, USA.
| |
Collapse
|
109
|
Ameratunga R, Woon ST, Bryant VL, Steele R, Slade C, Leung EY, Lehnert K. Clinical Implications of Digenic Inheritance and Epistasis in Primary Immunodeficiency Disorders. Front Immunol 2018; 8:1965. [PMID: 29434582 PMCID: PMC5790765 DOI: 10.3389/fimmu.2017.01965] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2017] [Accepted: 12/19/2017] [Indexed: 12/16/2022] Open
Abstract
The existence of epistasis in humans was first predicted by Bateson in 1909. Epistasis describes the non-linear, synergistic interaction of two or more genetic loci, which can substantially modify disease severity or result in entirely new phenotypes. The concept has remained controversial in human genetics because of the lack of well-characterized examples. In humans, it is only possible to demonstrate epistasis if two or more genes are mutated. In most cases of epistasis, the mutated gene products are likely to be constituents of the same physiological pathway leading to severe disruption of a cellular function such as antibody production. We have recently described a digenic family, who carry mutations of TNFRSF13B/TACI as well as TCF3 genes. Both genes lie in tandem along the immunoglobulin isotype switching and secretion pathway. We have shown they interact in an epistatic way causing severe immunodeficiency and autoimmunity in the digenic proband. With the advent of next generation sequencing, it is likely other families with digenic inheritance will be identified. Since digenic inheritance does not always cause epistasis, we propose an epistasis index which may help quantify the effects of the two mutations. We also discuss the clinical implications of digenic inheritance and epistasis in humans with primary immunodeficiency disorders.
Collapse
Affiliation(s)
- Rohan Ameratunga
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand.,Department of Clinical Immunology, Auckland City Hospital, Auckland, New Zealand
| | - See-Tarn Woon
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand
| | - Vanessa L Bryant
- Department of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Richard Steele
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand
| | - Charlotte Slade
- Department of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Allergy and Clinical Immunology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Euphemia Yee Leung
- Auckland Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Klaus Lehnert
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| |
Collapse
|
110
|
Flow cytometry-based diagnosis of primary immunodeficiency diseases. Allergol Int 2018; 67:43-54. [PMID: 28684198 DOI: 10.1016/j.alit.2017.06.003] [Citation(s) in RCA: 69] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2017] [Revised: 05/09/2017] [Accepted: 05/26/2017] [Indexed: 10/19/2022] Open
Abstract
Primary immunodeficiencies (PIDs) are a heterogeneous group of inherited diseases of the immune system. The definite diagnosis of PID is ascertained by genetic analysis; however, this takes time and is costly. Flow cytometry provides a rapid and highly sensitive tool for diagnosis of PIDs. Flow cytometry can evaluate specific cell populations and subpopulations, cell surface, intracellular and intranuclear proteins, biologic effects associated with specific immune defects, and certain functional immune characteristics, each being useful for the diagnosis and evaluation of PIDs. Flow cytometry effectively identifies major forms of PIDs, including severe combined immunodeficiency, X-linked agammaglobulinemia, hyper IgM syndromes, Wiskott-Aldrich syndrome, X-linked lymphoproliferative syndrome, familial hemophagocytic lymphohistiocytosis, autoimmune lymphoproliferative syndrome, IPEX syndrome, CTLA 4 haploinsufficiency and LRBA deficiency, IRAK4 and MyD88 deficiencies, Mendelian susceptibility to mycobacterial disease, chronic mucocuneous candidiasis, and chronic granulomatous disease. While genetic analysis is the definitive approach to establish specific diagnoses of PIDs, flow cytometry provides a tool to effectively evaluate patients with PIDs at relatively low cost.
Collapse
|
111
|
Update: Vaccines in primary immunodeficiency. J Allergy Clin Immunol 2017; 141:474-481. [PMID: 29288077 DOI: 10.1016/j.jaci.2017.12.980] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2017] [Revised: 12/19/2017] [Accepted: 12/19/2017] [Indexed: 11/21/2022]
Abstract
Vaccines were originally developed to prevent or ameliorate infectious disease. As knowledge of immune function and appreciation of immunodeficiency has developed, researchers have used vaccine responses as a tool to characterize the phenotypes of patients exhibiting various syndromes. Thus it has become possible for a clinician to evaluate individual responses to vaccines to interrogate the immunocompetence of their patients. Although there have been many advances in these areas, we still have much to learn about the quantity and quality of humoral and cellular vaccine responses in healthy and immunodeficient subjects and how that knowledge can then be extrapolated to diagnostic purposes. Adverse effects of vaccines have been recognized for many years, especially the occurrence of infections caused by viable vaccine organisms in immunodeficient hosts. Nevertheless, vaccines are essential for disease prevention in immunodeficient patients, just as they are for healthy subjects. Clinicians must understand the appropriate and safe use of vaccines in patients with immunodeficiency. This review highlights some recent advances and ongoing challenges in application of vaccines for the diagnosis and treatment of immunodeficiencies.
Collapse
|
112
|
Ghraichy M, Galson JD, Kelly DF, Trück J. B-cell receptor repertoire sequencing in patients with primary immunodeficiency: a review. Immunology 2017; 153:145-160. [PMID: 29140551 DOI: 10.1111/imm.12865] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2017] [Revised: 10/25/2017] [Accepted: 11/07/2017] [Indexed: 12/17/2022] Open
Abstract
The advent of next-generation sequencing (NGS) now allows a detailed assessment of the adaptive immune system in health and disease. In particular, high-throughput B-cell receptor (BCR) repertoire sequencing provides detailed information about the functionality and abnormalities of the B-cell system. However, it is mostly unknown how the BCR repertoire is altered in the context of primary immunodeficiencies (PID) and whether findings are consistent throughout phenotypes and genotypes. We have performed an extensive literature search of the published work on BCR repertoire sequencing in PID patients, including several forms of predominantly antibody disorders and combined immunodeficiencies. It is somewhat surprising that BCR repertoires, even from severe clinical phenotypes, often show only mild abnormalities and that diversity or immunoglobulin gene segment usage is generally preserved to some extent. Despite the great variety of wet laboratory and analytical methods that were used in the different studies, several findings are common to most investigated PIDs, such as the increased usage of gene segments that are associated with self-reactivity. These findings suggest that BCR repertoire characteristics may be used to assess the functionality of the B-cell compartment irrespective of the underlying defect. With the use of NGS approaches, there is now the opportunity to apply BCR repertoire sequencing to multiple patients and explore the PID BCR repertoire in more detail. Ultimately, using BCR repertoire sequencing in translational research could aid the management of PID patients by improving diagnosis, estimating functionality of the immune system and improving assessment of prognosis.
Collapse
Affiliation(s)
- Marie Ghraichy
- Division of Immunology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland
| | - Jacob D Galson
- Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| | - Dominic F Kelly
- Oxford Vaccine Group, Department of Paediatrics, University of Oxford, Oxford, UK
| | - Johannes Trück
- Division of Immunology, University Children's Hospital Zurich, Zurich, Switzerland.,Children's Research Center, University Children's Hospital, University of Zurich, Zurich, Switzerland.,University of Zurich, Zurich, Switzerland
| |
Collapse
|
113
|
Bonagura VR, Kaplan B, Jongco AM. Management of primary antibody deficiency syndromes. Ann Allergy Asthma Immunol 2017; 117:620-626. [PMID: 27979019 DOI: 10.1016/j.anai.2016.08.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2016] [Revised: 08/16/2016] [Accepted: 08/16/2016] [Indexed: 11/17/2022]
Affiliation(s)
- Vincent R Bonagura
- Laboratory of Host Defense, Center for Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, New York; Division of Allergy and Immunology, Hofstra Northwell School of Medicine, Great Neck, New York.
| | - Blanka Kaplan
- Division of Allergy and Immunology, Hofstra Northwell School of Medicine, Great Neck, New York
| | - Artemio M Jongco
- Laboratory of Host Defense, Center for Immunology and Inflammation, Feinstein Institute for Medical Research, Manhasset, New York; Division of Allergy and Immunology, Hofstra Northwell School of Medicine, Great Neck, New York
| |
Collapse
|
114
|
Jiménez-Reinoso A, Marin AV, Subias M, López-Lera A, Román-Ortiz E, Payne K, Ma CS, Arbore G, Kolev M, Freeley SJ, Kemper C, Tangye SG, Fernández-Malavé E, Rodríguez de Córdoba S, López-Trascasa M, Regueiro JR. Human plasma C3 is essential for the development of memory B, but not T, lymphocytes. J Allergy Clin Immunol 2017; 141:1151-1154.e14. [PMID: 29113906 DOI: 10.1016/j.jaci.2017.09.037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 08/06/2017] [Accepted: 09/08/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Anaïs Jiménez-Reinoso
- Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Ana V Marin
- Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Marta Subias
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Alberto López-Lera
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain; Immunology Unit, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | | | - Kathryn Payne
- Immunology Research Program, Garvan Institute of Medical Research, Darlinghurst, Australia
| | - Cindy S Ma
- Immunology Research Program, Garvan Institute of Medical Research, Darlinghurst, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Giuseppina Arbore
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's, College London, London, United Kingdom
| | - Martin Kolev
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's, College London, London, United Kingdom
| | - Simon J Freeley
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's, College London, London, United Kingdom
| | - Claudia Kemper
- MRC Centre for Transplantation, Division of Transplant Immunology and Mucosal Biology, King's, College London, London, United Kingdom
| | - Stuart G Tangye
- Immunology Research Program, Garvan Institute of Medical Research, Darlinghurst, Australia; St Vincent's Clinical School, University of New South Wales, Sydney, Australia
| | - Edgar Fernández-Malavé
- Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain
| | - Santiago Rodríguez de Córdoba
- Centro de Investigaciones Biológicas (CSIC), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Margarita López-Trascasa
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain; Immunology Unit, Hospital Universitario La Paz, IdiPAZ, Madrid, Spain
| | - José R Regueiro
- Department of Immunology, Complutense University School of Medicine and 12 de Octubre Health Research Institute (imas12), Madrid, Spain.
| |
Collapse
|
115
|
Rosain J, Miot C, Lambert N, Rousselet MC, Pellier I, Picard C. CD21 deficiency in 2 siblings with recurrent respiratory infections and hypogammaglobulinemia. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 5:1765-1767.e3. [PMID: 28499783 DOI: 10.1016/j.jaip.2017.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2017] [Revised: 03/22/2017] [Accepted: 04/04/2017] [Indexed: 12/31/2022]
|
116
|
Jain MD, Davila ML. Concise Review: Emerging Principles from the Clinical Application of Chimeric Antigen Receptor T Cell Therapies for B Cell Malignancies. Stem Cells 2017; 36:36-44. [PMID: 29024301 DOI: 10.1002/stem.2715] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 08/27/2017] [Accepted: 09/21/2017] [Indexed: 11/07/2022]
Abstract
Gene-engineered T cell therapies are soon to be United States Food and Drug Administration (FDA) approved for at least two types of B cell malignancies in pediatric and adult patients, in the form of CD19 targeted chimeric antigen receptor T (CAR T) cell therapy. This represents a triumph of a true bench to bedside clinical translation of a therapy that was conceived of in the early 1990s. Clinical results have demonstrated efficacious responses in patients with the CD19 positive diseases B cell acute lymphoblastic leukemia and diffuse large B cell lymphoma. However, significant challenges have emerged, including worrisome immune-related toxicities, therapy resistance, and understanding how to administer CD19 CAR T cells in clinical practice. Although much remains to be learned, pioneering clinical trials have led to foundational insights about the clinical translation of this novel therapy. Here, we review the "lessons learned" from the pre-clinical and human experience with CAR T cell therapy. Stem Cells 2018;36:36-44.
Collapse
Affiliation(s)
- Michael D Jain
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, University of South Florida, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Marco L Davila
- Department of Blood and Marrow Transplantation and Cellular Immunotherapy, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
- Department of Oncologic Sciences, University of South Florida, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| |
Collapse
|
117
|
Ameratunga R, Koopmans W, Woon ST, Leung E, Lehnert K, Slade CA, Tempany JC, Enders A, Steele R, Browett P, Hodgkin PD, Bryant VL. Epistatic interactions between mutations of TACI ( TNFRSF13B) and TCF3 result in a severe primary immunodeficiency disorder and systemic lupus erythematosus. Clin Transl Immunology 2017; 6:e159. [PMID: 29114388 PMCID: PMC5671988 DOI: 10.1038/cti.2017.41] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2017] [Revised: 07/21/2017] [Accepted: 07/21/2017] [Indexed: 12/22/2022] Open
Abstract
Common variable immunodeficiency disorders (CVID) are a group of primary immunodeficiencies where monogenetic causes account for only a fraction of cases. On this evidence, CVID is potentially polygenic and epistatic although there are, as yet, no examples to support this hypothesis. We have identified a non-consanguineous family, who carry the C104R (c.310T>C) mutation of the Transmembrane Activator Calcium-modulator and cyclophilin ligand Interactor (TACI, TNFRSF13B) gene. Variants in TNFRSF13B/TACI are identified in up to 10% of CVID patients, and are associated with, but not solely causative of CVID. The proband is heterozygous for the TNFRSF13B/TACI C104R mutation and meets the Ameratunga et al. diagnostic criteria for CVID and the American College of Rheumatology criteria for systemic lupus erythematosus (SLE). Her son has type 1 diabetes, arthritis, reduced IgG levels and IgA deficiency, but has not inherited the TNFRSF13B/TACI mutation. Her brother, homozygous for the TNFRSF13B/TACI mutation, is in good health despite profound hypogammaglobulinemia and mild cytopenias. We hypothesised that a second unidentified mutation contributed to the symptomatic phenotype of the proband and her son. Whole-exome sequencing of the family revealed a de novo nonsense mutation (T168fsX191) in the Transcription Factor 3 (TCF3) gene encoding the E2A transcription factors, present only in the proband and her son. We demonstrate mutations of TNFRSF13B/TACI impair immunoglobulin isotype switching and antibody production predominantly via T-cell-independent signalling, while mutations of TCF3 impair both T-cell-dependent and -independent pathways of B-cell activation and differentiation. We conclude that epistatic interactions between mutations of the TNFRSF13B/TACI and TCF3 signalling networks lead to the severe CVID-like disorder and SLE in the proband.
Collapse
Affiliation(s)
- Rohan Ameratunga
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand.,Department of Clinical Immunology, Auckland City Hospital, Auckland, New Zealand
| | - Wikke Koopmans
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand
| | - See-Tarn Woon
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand
| | - Euphemia Leung
- Cancer Society Research Centre, University of Auckland, Auckland, New Zealand
| | - Klaus Lehnert
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Charlotte A Slade
- Department of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Allergy and Clinical Immunology, Royal Melbourne Hospital, Parkville, VIC, Australia
| | - Jessica C Tempany
- Department of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Anselm Enders
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research and Centre for Personalised Immunology, Australian National University, Canberra, ACT, Australia
| | - Richard Steele
- Department of Virology and Immunology, Auckland City Hospital, Auckland, New Zealand
| | - Peter Browett
- Department of Hematology, LabPlus, Auckland City Hospital, Auckland, New Zealand.,Department of Molecular Medicine, and Pathology University of Auckland, Auckland, New Zealand
| | - Philip D Hodgkin
- Department of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia
| | - Vanessa L Bryant
- Department of Immunology, Walter and Eliza Hall Institute of Medical Research, Parkville, VIC, Australia.,Department of Medical Biology, University of Melbourne, Parkville, VIC, Australia.,Department of Allergy and Clinical Immunology, Royal Melbourne Hospital, Parkville, VIC, Australia
| |
Collapse
|
118
|
Queudeville M, Handgretinger R, Ebinger M. Immunotargeting relapsed or refractory precursor B-cell acute lymphoblastic leukemia - role of blinatumomab. Onco Targets Ther 2017; 10:3567-3578. [PMID: 28790849 PMCID: PMC5530848 DOI: 10.2147/ott.s103470] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Patients with refractory or relapsed (R/R) acute lymphoblastic leukemia (ALL) have a dismal prognosis of around 5% long-term survival when treated with cytotoxic chemotherapy and allogenic stem cell transplantation. T-cell immunobased strategies open up new therapeutic perspectives. Blinatumomab is the first of a new class of antibody constructs that was labeled bispecific T-cell engager (BiTE): it consists of two single chain variable fragment connected with a flexible linker, one side binding CD3, the other CD19. The tight binding and the close proximity to the CD19-positive B-cells and leukemic cells leads to non-major histocompatibility complex-restricted T-cell activation, polyclonal T-cell expansion and direct target cell killing. Applied by continuous infusion, blinatumomab achieves morphological complete response rates ranging from 39% to 69% in R/R ALL patients (compared to 25% after second-line chemotherapy) with prolonged overall survival (blinatumomab median overall survival, 7.7 months vs chemotherapy, 4.0 months). In comparison to conventional cytotoxic second-line protocols blinatumomab has a favorable safety profile. The main adverse event is related to the mode of action of blinatumomab: the induction of a cytokine-release syndrome that can be managed by interruption and/or the application of steroids or tocilizumab. Another typical complication is the occurrence of neurological side effects, such as seizures and encephalopathy. This neurotoxicity is reversible after application of steroids and/or withdrawal of blinatumomab. Blinatumomab has proven to be a powerful therapeutic option in R/R ALL patients both adult and pediatric because of its efficacy and limited toxicity.
Collapse
Affiliation(s)
- Manon Queudeville
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University of Tübingen, Tübingen, Germany
| | - Rupert Handgretinger
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University of Tübingen, Tübingen, Germany
| | - Martin Ebinger
- Department of Pediatric Hematology and Oncology, University Children's Hospital, University of Tübingen, Tübingen, Germany
| |
Collapse
|
119
|
Abstract
Within a few years, the success of treatments based on the use of T-cells armed with a chimeric T-receptor for the CD19 molecule (CAR-T CD19) has revolutionized the perception of adoptive transfer approaches. The levels of responses observed in acute leukemias, of the order of 70-90 % are indeed unprecedented. The medical and financial enthusiasm aroused by these results has led to the current situation where more than 300 clinical trials are under way, against some thirty different antigens. This enthusiasm, well justified by the first successes, must however be tempered by the difficulties associated with the use of these cells. Indeed, the management of patients is made very complex both for medical reasons, because the toxicities associated with these treatments are important, and for technical reasons, because the preparation of T lymphocytes for therapeutic use requires dedicated structures. During this same period, knowledge of the mechanisms of regulation of T lymphocytes and the possibilities offered by synthetic biology and techniques of genome engineering have progressed considerably. Combined, they allow envisaging a true "programming" of the T lymphocytes, intended to improve the efficiency of the treatments and the safety of the patients. Medical and industrial perspectives and the role of these approaches in the arsenal of cancer therapies will depend largely on two conditions: the emergence of a robust demonstration of their effectiveness in solid tumors, and the establishment of an acceptable production and distribution model 1.
Collapse
Affiliation(s)
- H Vié
- Inserm U1232, institut de recherche en santé de l'université de Nantes, 8, quai Moncousu, 44007 Nantes, France; Établissement français du sang (EFS), Pays-de-la-Loire, 34, boulevard Jean-Monnet, BP 91115, 44000 Nantes, France.
| | - B Clémenceau
- Inserm U1232, institut de recherche en santé de l'université de Nantes, 8, quai Moncousu, 44007 Nantes, France; UTCG, CHU de Nantes, 9, quai Moncousu, 44093 Nantes, France
| |
Collapse
|
120
|
Pott MC, Frede N, Wanders J, Hammarström L, Glocker EO, Glocker C, Tahami F, Grimbacher B. Autoantibodies against BAFF, APRIL or IL21 - an alternative pathogenesis for antibody-deficiencies? BMC Immunol 2017. [PMID: 28651547 PMCID: PMC5485583 DOI: 10.1186/s12865-017-0217-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND The ability of anti-cytokine antibodies to play a disease-causing role in the pathogenesis of immunodeficiencies is widely accepted. The aim of this study was to investigate whether autoantibodies against BAFF (important B cell survival signal), APRIL (important plasma cell survival signal), or Interleukin-21 (important cytokine for immunoglobulin class switch) present an alternative mechanism for the development of the following primary antibody deficiencies (PADs): common variable immune deficiency (CVID) or selective IgA deficiency (sIgAD). RESULTS Two hundred thirty-two sera from patients with PADs were screened for autoantibodies against cytokines by ELISA. Statistical data analysis yielded a significant difference (p < 0.01) between the healthy donor sera and both PAD cohorts. The analysis was deepened by subdividing the patient collective into groups with distinct B cell phenotypes but no significant differences were found. For selected sera with notable high ELISA-read outs functional analysis ensued. Anti-BAFF and anti-APRIL antibodies were further examined by a B cell survival assay, whilst the functional relevance of putative anti-IL-21 autoantibodies was investigated by means of a STAT3 phosphorylation assay. However, the results of these experiments revealed no discernible functional effect. CONCLUSION Whilst statistical analysis of ELISA results showed significant differences between patients and healthy controls, in our set of patients functional tests yielded no evidence for an involvement of autoantibodies against BAFF, APRIL, or IL-21 in the pathogenesis of CVID or sIgAD.
Collapse
Affiliation(s)
- Marian-Christopher Pott
- Centre for Chronic Immunodeficiency, Medical Centre University Hospital, Medical Faculty of Freiburg, Freiburg, Germany
| | - Natalie Frede
- Centre for Chronic Immunodeficiency, Medical Centre University Hospital, Medical Faculty of Freiburg, Freiburg, Germany
| | - Jennifer Wanders
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, UK
| | | | - Erik-Oliver Glocker
- Institute of Medical Microbiology and Hygiene, University Medical Center Freiburg, Freiburg, Germany.,Institute of Laboratory Medicine, Brandenburg Hospital, Brandenburg Medical School, Brandenburg, Germany
| | - Cristina Glocker
- Centre for Chronic Immunodeficiency, Medical Centre University Hospital, Medical Faculty of Freiburg, Freiburg, Germany
| | - Fariba Tahami
- Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, UK
| | - Bodo Grimbacher
- Centre for Chronic Immunodeficiency, Medical Centre University Hospital, Medical Faculty of Freiburg, Freiburg, Germany. .,Institute of Immunity and Transplantation, Royal Free Hospital, University College London, London, UK.
| |
Collapse
|
121
|
Lim WA, June CH. The Principles of Engineering Immune Cells to Treat Cancer. Cell 2017; 168:724-740. [PMID: 28187291 DOI: 10.1016/j.cell.2017.01.016] [Citation(s) in RCA: 788] [Impact Index Per Article: 98.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Revised: 01/12/2017] [Accepted: 01/17/2017] [Indexed: 12/13/2022]
Abstract
Chimeric antigen receptor (CAR) T cells have proven that engineered immune cells can serve as a powerful new class of cancer therapeutics. Clinical experience has helped to define the major challenges that must be met to make engineered T cells a reliable, safe, and effective platform that can be deployed against a broad range of tumors. The emergence of synthetic biology approaches for cellular engineering is providing us with a broadly expanded set of tools for programming immune cells. We discuss how these tools could be used to design the next generation of smart T cell precision therapeutics.
Collapse
Affiliation(s)
- Wendell A Lim
- Howard Hughes Medical Institute, Department of Cellular and Molecular Pharmacology, UCSF Center for Systems and Synthetic Biology, University of California, San Francisco, San Francisco, CA 94158, USA.
| | - Carl H June
- Center for Cellular Immunotherapies, the Department of Pathology and Laboratory Medicine at the Perelman School of Medicine, and the Parker Institute for Cancer Immunotherapy, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
122
|
Abstract
Crosstalk between B and T cells in transplantation is increasingly recognized as being important in the alloimmune response. T cell activation of B cells occurs by a 3-stage pathway, culminating with costimulation signals. We review the distinct T cell subtypes required for B-cell activation and discuss the formation of the germinal center (GC) after transplantation, with particular reference to the repopulation of the GC after depletional induction, and the subsequent effect of immunosuppressive manipulation of T cell-B cell interactions. In addition, ectopic GCs are seen in transplantation, but their role is not fully understood. Therapeutic options to target T cell-B cell interactions are of considerable interest, both as immunosuppressive tools, and to aid in the further understanding of these important alloimmune mechanisms.
Collapse
|
123
|
Ryherd M, Plassmeyer M, Alexander C, Eugenio I, Kleschenko Y, Badger A, Gupta R, Alpan O, Sønder SU. Improved panels for clinical immune phenotyping: Utilization of the violet laser. CYTOMETRY PART B-CLINICAL CYTOMETRY 2017; 94:671-679. [DOI: 10.1002/cyto.b.21532] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/11/2017] [Accepted: 05/04/2017] [Indexed: 12/13/2022]
Affiliation(s)
| | | | | | | | | | | | - Raavi Gupta
- Amerimmune LLC; Fairfax Virginia
- Department of Pathology; SUNY; New York
| | | | | |
Collapse
|
124
|
Kienzler AK, Hargreaves CE, Patel SY. The role of genomics in common variable immunodeficiency disorders. Clin Exp Immunol 2017; 188:326-332. [PMID: 28236292 DOI: 10.1111/cei.12947] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/18/2017] [Indexed: 01/16/2023] Open
Abstract
The advent of next-generation sequencing (NGS) and 'omic' technologies has revolutionized the field of genetics, and its implementation in health care has the potential to realize precision medicine. Primary immunodeficiencies (PID) are a group of rare diseases which have benefited from NGS, with a massive increase in causative genes identified in the past few years. Common variable immunodeficiency disorders (CVID) are a heterogeneous form of PID and the most common form of antibody failure in children and adults. While a monogenic cause of disease has been identified in a small subset of CVID patients, a genomewide association study and whole genome sequencing have found that, in the majority, a polygenic cause is likely. Other NGS technologies such as RNA sequencing and epigenetic studies have contributed further to our understanding of the contribution of altered gene expression in CVID pathogenesis. We believe that to unravel further the complexities of CVID, a multi-omic approach, combining DNA sequencing with gene expression, methylation, proteomic and metabolomics data, will be essential to identify novel disease-associated pathways and therapeutic targets.
Collapse
Affiliation(s)
- A-K Kienzler
- NIHR Oxford Biomedical Research Centre, Clinical Immunology Group, Oxford, UK
| | - C E Hargreaves
- NIHR Oxford Biomedical Research Centre, Clinical Immunology Group, Oxford, UK
| | - S Y Patel
- NIHR Oxford Biomedical Research Centre, Clinical Immunology Group, Oxford, UK
| |
Collapse
|
125
|
Phan TG, Tangye SG. Memory B cells: total recall. Curr Opin Immunol 2017; 45:132-140. [PMID: 28363157 DOI: 10.1016/j.coi.2017.03.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 12/27/2016] [Accepted: 03/02/2017] [Indexed: 12/18/2022]
Abstract
Immunological memory is a cornerstone of adaptive immune responses in higher vertebrates. The remarkable ability to generate memory cells following Ag exposure, in the context of natural infection or immunization, provides long-lived protection against infectious diseases, often for the hosts' lifetime. Indeed, the generation of memory B cells and long-lived plasma cells underpins the success of most vaccines. The concept of immunological memory is not new-it was first proposed nearly 2500 years ago. While our understanding of the complexities of humoral and cell-mediated memory continues to evolve, important aspects of this process remain unresolved. Here, we will provide an overview of recent advances in B-cell memory in mice and humans, and in health and disease.
Collapse
Affiliation(s)
- Tri Giang Phan
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, University of NSW, Australia.
| | - Stuart G Tangye
- Immunology Division, Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; St Vincent's Clinical School, University of NSW, Australia.
| |
Collapse
|
126
|
Arduini S, Dunne J, Conlon N, Feighery C, Doherty DG. Mucosal-associated invariant T cells are depleted and functionally altered in patients with common variable immunodeficiency. Clin Immunol 2017; 176:23-30. [PMID: 28011187 DOI: 10.1016/j.clim.2016.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2016] [Revised: 12/01/2016] [Accepted: 12/12/2016] [Indexed: 12/15/2022]
Abstract
Common variable immunodeficiency (CVID) is a primary immunoglobulin deficiency characterized by recurrent infections and complications, including autoimmunity, enteropathy, polyclonal lymphocytic infiltration or lymphoid malignancy. Innate T cells can support B cell maturation and antibody production. We investigated the numbers, phenotypes and functions of circulating B cell, γδ T cell, invariant natural killer T (iNKT) cell and mucosal-associated invariant T (MAIT) cell subsets in 23 CVID patients and 27 healthy controls. Switched-memory B cells and plasmablasts were depleted in CVID patients (p<0.0001). γδ T cells were found at normal numbers, but iNKT and MAIT cells were depleted (p<0.0001 and p<0.002). MAIT cells were especially low in patients with complicated CVID (p<0.05). MAIT cells from patients appeared more activated and more frequently produced interleukin-17A, interleukin-22 and tumor necrosis factor-α than MAIT cells from healthy subjects in vitro. Thus, MAIT cell depletion and activation may contribute to immunodeficiency and complications associated with CVID.
Collapse
Affiliation(s)
- Serena Arduini
- Discipline of Immunology, School of Medicine, Trinity College Dublin, Dublin, Ireland
| | - Jean Dunne
- Department of Immunology, St. James's Hospital, Dublin, Ireland
| | - Niall Conlon
- Department of Immunology, St. James's Hospital, Dublin, Ireland
| | - Conleth Feighery
- Discipline of Immunology, School of Medicine, Trinity College Dublin, Dublin, Ireland; Department of Immunology, St. James's Hospital, Dublin, Ireland
| | - Derek G Doherty
- Discipline of Immunology, School of Medicine, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
127
|
Berbers RM, Nierkens S, van Laar JM, Bogaert D, Leavis HL. Microbial Dysbiosis in Common Variable Immune Deficiencies: Evidence, Causes, and Consequences. Trends Immunol 2017; 38:206-216. [DOI: 10.1016/j.it.2016.11.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/29/2016] [Accepted: 11/30/2016] [Indexed: 12/19/2022]
|
128
|
Li X, Ding Y, Zi M, Sun L, Zhang W, Chen S, Xu Y. CD19, from bench to bedside. Immunol Lett 2017; 183:86-95. [PMID: 28153605 DOI: 10.1016/j.imlet.2017.01.010] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2016] [Revised: 01/19/2017] [Accepted: 01/20/2017] [Indexed: 12/27/2022]
|
129
|
Age-related alterations of the CD19 complex and memory B cells in children with Down syndrome. Clin Exp Med 2017; 18:125-131. [PMID: 28197808 DOI: 10.1007/s10238-017-0457-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2016] [Accepted: 02/06/2017] [Indexed: 10/20/2022]
Abstract
Children with Down syndrome (DS) have a high incidence of recurrent respiratory tract infections, leukaemia and autoimmune disorders, suggesting immune dysfunction. The present study evaluated the role of the CD19 complex and memory B cells in the pathogenesis of immunodeficiency in children with DS. The expression levels (median fluorescein intensity-MFI) of CD19, CD21 and CD81 molecules on the surface of B cells and memory B cell subsets were studied in 37 patients and 39 healthy controls. Twenty-nine of the DS group had congenital cardiac disease. The B cell count was significantly low in children with DS compared with healthy age-matched controls for all three age groups (under 2 years; 2-6 years and older than 6 years). The MFI of CD19 was reduced in all the age groups, whereas that of CD21 was increased in those older than 2 years with DS. The expression level of CD81 was significantly increased in those older than 6 years. Age-related changes were also detected in memory B cell subsets. The frequency of CD27+IgD+IgM+ natural effector B cells was reduced in children with DS who had needed hospitalisation admission due to infections. The observed intrinsic defects in B cells may be responsible for the increased susceptibility of children with DS to severe respiratory tract infections.
Collapse
|
130
|
Perez EE, Orange JS, Bonilla F, Chinen J, Chinn IK, Dorsey M, El-Gamal Y, Harville TO, Hossny E, Mazer B, Nelson R, Secord E, Jordan SC, Stiehm ER, Vo AA, Ballow M. Update on the use of immunoglobulin in human disease: A review of evidence. J Allergy Clin Immunol 2016; 139:S1-S46. [PMID: 28041678 DOI: 10.1016/j.jaci.2016.09.023] [Citation(s) in RCA: 415] [Impact Index Per Article: 46.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Revised: 09/12/2016] [Accepted: 09/23/2016] [Indexed: 12/20/2022]
Abstract
Human immunoglobulin preparations for intravenous or subcutaneous administration are the cornerstone of treatment in patients with primary immunodeficiency diseases affecting the humoral immune system. Intravenous preparations have a number of important uses in the treatment of other diseases in humans as well, some for which acceptable treatment alternatives do not exist. We provide an update of the evidence-based guideline on immunoglobulin therapy, last published in 2006. Given the potential risks and inherent scarcity of human immunoglobulin, careful consideration of its indications and administration is warranted.
Collapse
Affiliation(s)
- Elena E Perez
- Allergy Associates of the Palm Beaches, North Palm Beach, Fla.
| | - Jordan S Orange
- Department of Pediatrics, Section of Immunology Allergy and Rheumatology, Center for Human Immunobiology, Texas Children's Hospital and Baylor College of Medicine, Houston, Tex
| | - Francisco Bonilla
- Department of Pediatrics, Clinical Immunology Program, Children's Hospital Boston and Harvard Medical School, Boston, Mass
| | - Javier Chinen
- Department of Pediatrics, Section of Immunology Allergy and Rheumatology, Center for Human Immunobiology, Texas Children's Hospital and Baylor College of Medicine, Houston, Tex
| | - Ivan K Chinn
- Department of Pediatrics, Section of Immunology Allergy and Rheumatology, Center for Human Immunobiology, Texas Children's Hospital and Baylor College of Medicine, Houston, Tex
| | - Morna Dorsey
- Department of Pediatrics, Allergy, Immunology and BMT Division, Benioff Children's Hospital and University of California, San Francisco, Calif
| | - Yehia El-Gamal
- Department of Pediatrics, Pediatric Allergy and Immunology Unit, Children's Hospital and Ain Shams University, Cairo, Egypt
| | - Terry O Harville
- Departments of Pathology and Laboratory Services and Pediatrics, University of Arkansas, Little Rock, Ark
| | - Elham Hossny
- Department of Pediatrics, Pediatric Allergy and Immunology Unit, Children's Hospital and Ain Shams University, Cairo, Egypt
| | - Bruce Mazer
- Department of Pediatrics, Allergy and Immunology, Montreal Children's Hospital and McGill University, Montreal, Quebec, Canada
| | - Robert Nelson
- Department of Medicine and Pediatrics, Division of Hematology and Oncology and Stem Cell Transplantation, Riley Hospital, Indiana University School of Medicine and the IU Melvin and Bren Simon Cancer Center, Indianapolis, Ind
| | - Elizabeth Secord
- Department of Pediatrics, Wayne State University, Children's Hospital of Michigan, Detroit, Mich
| | - Stanley C Jordan
- Nephrology & Transplant Immunology, Kidney Transplant Program, David Geffen School of Medicine at UCLA and Cedars-Sinai Medical Center, Los Angeles, Calif
| | - E Richard Stiehm
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, Calif
| | - Ashley A Vo
- Transplant Immunotherapy Program, Comprehensive Transplant Center, Kidney Transplant Program, Cedars-Sinai Medical Center, Los Angeles, Calif
| | - Mark Ballow
- Department of Pediatrics, Division of Allergy & Immunology, University of South Florida, Morsani College of Medicine, Johns Hopkins All Children's Hospital, St Petersburg, Fla
| |
Collapse
|
131
|
Kieckens E, Rybarczyk J, Li RW, Vanrompay D, Cox E. Potential immunosuppressive effects of Escherichia coli O157:H7 experimental infection on the bovine host. BMC Genomics 2016; 17:1049. [PMID: 28003017 PMCID: PMC5178093 DOI: 10.1186/s12864-016-3374-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Accepted: 12/05/2016] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Enterohaemorrhagic Escherichia coli (EHEC), like E. coli O157:H7 are frequently detected in bovine faecal samples at slaughter. Cattle do not show clinical symptoms upon infection, but for humans the consequences after consuming contaminated beef can be severe. The immune response against EHEC in cattle cannot always clear the infection as persistent colonization and shedding in infected animals over a period of months often occurs. In previous infection trials, we observed a primary immune response after infection which was unable to protect cattle from re-infection. These results may reflect a suppression of certain immune pathways, making cattle more prone to persistent colonization after re-infection. To test this, RNA-Seq was used for transcriptome analysis of recto-anal junction tissue and ileal Peyer's patches in nine Holstein-Friesian calves in response to a primary and secondary Escherichia coli O157:H7 infection with the Shiga toxin (Stx) negative NCTC12900 strain. Non-infected calves served as controls. RESULTS In tissue of the recto-anal junction, only 15 genes were found to be significantly affected by a first infection compared to 1159 genes in the ileal Peyer's patches. Whereas, re-infection significantly changed the expression of 10 and 17 genes in the recto-anal junction tissue and the Peyer's patches, respectively. A significant downregulation of 69 immunostimulatory genes and a significant upregulation of seven immune suppressing genes was observed. CONCLUSIONS Although the recto-anal junction is a major site of colonization, this area does not seem to be modulated upon infection to the same extent as ileal Peyer's patches as the changes in gene expression were remarkably higher in the ileal Peyer's patches than in the recto-anal junction during a primary but not a secondary infection. We can conclude that the main effect on the transcriptome was immunosuppression by E. coli O157:H7 (Stx-) due to an upregulation of immune suppressive effects (7/12 genes) or a downregulation of immunostimulatory effects (69/94 genes) in the ileal Peyer's patches. These data might indicate that a primary infection promotes a re-infection with EHEC by suppressing the immune function.
Collapse
Affiliation(s)
- E. Kieckens
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
- Laboratory of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - J. Rybarczyk
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
- Laboratory of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - R. W. Li
- USDA-ARS, Bovine Functional Genomics Laboratory, Beltsville, MD USA
| | - D. Vanrompay
- Laboratory of Immunology and Animal Biotechnology, Faculty of Bioscience Engineering, Ghent University, Coupure Links 653, 9000 Ghent, Belgium
| | - E. Cox
- Laboratory of Immunology, Faculty of Veterinary Medicine, Ghent University, Salisburylaan 133, 9820 Merelbeke, Belgium
| |
Collapse
|
132
|
Timmermans WMC, van Laar JAM, van Hagen PM, van Zelm MC. Immunopathogenesis of granulomas in chronic autoinflammatory diseases. Clin Transl Immunology 2016; 5:e118. [PMID: 28090320 PMCID: PMC5192066 DOI: 10.1038/cti.2016.75] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2016] [Revised: 11/11/2016] [Accepted: 11/12/2016] [Indexed: 12/23/2022] Open
Abstract
Granulomas are clusters of immune cells. These structures can be formed in reaction to infection and display signs of necrosis, such as in tuberculosis. Alternatively, in several immune disorders, such as sarcoidosis, Crohn's disease and common variable immunodeficiency, non-caseating granulomas are formed without an obvious infectious trigger. Despite advances in our understanding of the human immune system, the pathogenesis underlying these non-caseating granulomas in chronic inflammatory diseases is still poorly understood. Here, we review the current knowledge about the immunopathogenesis of granulomas, and we discuss how the involved immune cells can be targeted with novel therapeutics.
Collapse
Affiliation(s)
- Wilhelmina Maria Cornelia Timmermans
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| | - Jan Alexander Michael van Laar
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Petrus Martinus van Hagen
- Department of Internal Medicine, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
- Department of Immunology, Erasmus MC, University Medical Center, Rotterdam, The Netherlands
| | - Menno Cornelis van Zelm
- Department of Immunology and Pathology, Central Clinical School, Monash University, Melbourne, VIC, Australia
| |
Collapse
|
133
|
Guffroy A, Gies V, Martin M, Korganow AS. [Primary immunodeficiency and autoimmunity]. Rev Med Interne 2016; 38:383-392. [PMID: 27889323 DOI: 10.1016/j.revmed.2016.10.388] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2016] [Revised: 10/03/2016] [Accepted: 10/20/2016] [Indexed: 12/15/2022]
Abstract
Many evidences highlight that immunodeficiency and autoimmunity are two sides of a same coin. Primary immune deficiencies (PIDs), which are rare mono- or multigenic defects of innate or adaptative immunity, frequently associate with autoimmunity. Analyses of single-gene defects in immune pathways of families with PIDs, by new tools of molecular biology (next genome sequencing technologies), allowed a better understanding of the ways that could both drive immune defect with immune deficiency and autoimmunity. Moreover, genes implicated in rare single-gene defects are now known to be also involved in polygenic conventional autoimmune diseases. Here, we describe the main autoimmune symptoms occurring in PIDs and the underlying mechanisms that lead to autoimmunity in immunodeficiency. We review the links between autoimmunity and immunodeficiency and purpose some principles of care for patients with PIDs and autoimmunity.
Collapse
Affiliation(s)
- A Guffroy
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France.
| | - V Gies
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - M Martin
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| | - A-S Korganow
- CNRS UPR 3572 « immunopathologie et chimie thérapeutique », laboratoire d'excellence Medalis, institut de biologie moléculaire et cellulaire (IBMC), 67000 Strasbourg, France; Service d'immunologie clinique et de médecine interne, Centre national de référence des maladies auto-immunes rares, hôpitaux universitaires de Strasbourg, 1, place de l'Hôpital, 67091 Strasbourg, France
| |
Collapse
|
134
|
Sobh A, Bonilla FA. Vaccination in Primary Immunodeficiency Disorders. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2016; 4:1066-1075. [DOI: 10.1016/j.jaip.2016.09.012] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 09/21/2016] [Accepted: 09/21/2016] [Indexed: 02/07/2023]
|
135
|
Berrón-Ruíz L, López-Herrera G, Ávalos-Martínez CE, Valenzuela-Ponce C, Ramírez-SanJuan E, Santoyo-Sánchez G, Mújica Guzmán F, Espinosa-Rosales FJ, Santos-Argumedo L. Variations of B cell subpopulations in peripheral blood of healthy Mexican population according to age: Relevance for diagnosis of primary immunodeficiencies. Allergol Immunopathol (Madr) 2016; 44:571-579. [PMID: 27780620 DOI: 10.1016/j.aller.2016.05.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2016] [Revised: 04/25/2016] [Accepted: 05/04/2016] [Indexed: 11/28/2022]
Abstract
BACKGROUND Peripheral blood B cells include lymphocytes at various stages of differentiation, each with a specific function in the immune response. All these stages show variations in percentage and absolute number throughout human life. The numbers and proportions of B subpopulation are influenced by factors such as gender, age, ethnicity, and lifestyle. This study establishes reference values according to age of peripheral blood B cell subtypes in healthy Mexican population. METHODS Peripheral blood from healthy new-borns and adults were analysed for total B cell subpopulations, using surface markers such as CD19, IgM, IgD, CD21, CD24, CD27, and CD38, to identify naïve, memory with and without isotype switch, double-negative, transitional, and plasmablast cells. RESULTS We observed a significant variation in terms of frequency and absolute counts between all groups analysed. Values from each B cell subpopulation show variations according to age. CONCLUSIONS In order to attempt to elucidate reference values for B cell subpopulation, the present study evaluated a population sample of healthy blood donors from this region. Values reported here can also be used as a tool for diagnosis of diseases in which B cell maturation is affected.
Collapse
Affiliation(s)
- L Berrón-Ruíz
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados-IPN, México, D.F., Mexico; Unidad de Investigación en Inmunodeficiencias, Instituto Nacional de Pediatría-SSa, México, D.F., Mexico
| | - G López-Herrera
- Unidad de Investigación en Inmunodeficiencias, Instituto Nacional de Pediatría-SSa, México, D.F., Mexico
| | - C E Ávalos-Martínez
- Laboratorio de Inmunoquímica, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, D.F., Mexico
| | - C Valenzuela-Ponce
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados-IPN, México, D.F., Mexico
| | - E Ramírez-SanJuan
- Laboratorio de Farmacología, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, México, D.F., Mexico
| | - G Santoyo-Sánchez
- Programa de Posgrado en Ciencias Médicas, Odontológicas y de la Salud, Universidad Nacional Autónoma de México, México, D.F., Mexico
| | - F Mújica Guzmán
- Laboratorio de Hematología, Instituto Nacional de Pediatría-SSa, México, D.F., Mexico
| | - F J Espinosa-Rosales
- Unidad de Investigación en Inmunodeficiencias, Instituto Nacional de Pediatría-SSa, México, D.F., Mexico
| | - L Santos-Argumedo
- Departamento de Biomedicina Molecular, Centro de Investigación y de Estudios Avanzados-IPN, México, D.F., Mexico.
| |
Collapse
|
136
|
Successful Treatment of ANCA-Associated Vasculitis in the Setting of Common Variable Immunodeficiency Using Rituximab. Am J Ther 2016; 23:e1239-45. [DOI: 10.1097/mjt.0000000000000323] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
|
137
|
Abstract
PURPOSE OF REVIEW Autoimmune and inflammatory manifestations are the biggest clinical challenge in the care of patients with common variable immunodeficiency (CVID). The increasing pathogenic knowledge and potential therapeutic implications require a new evaluation of the status quo. (Figure is included in full-text article.) RECENT FINDINGS The conundrum of the simultaneous manifestation of primary immunodeficiency and autoimmune disease (AID) is increasingly elucidated by newly discovered genetic defects. Thus, cytotoxic T lymphocyte-associated antigen 4 or caspase-9 deficiency presenting with CVID-like phenotypes reiterate concepts of immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome and autoimmune lymphoproliferative syndrome. Activating signaling defects downstream of antigen or cytokine receptors are often associated with loss-of-tolerance in the affected patients. Increasingly, forms of combined immunodeficiency are discovered among CVID-like patients. Although different autoimmune manifestations often coincide in the same patient their immunopathology varies. Treatment of AID in CVID remains a challenge, but based on a better definition of the immunopathology first attempts of targeted treatment have been made. SUMMARY The increasing comprehension of immunological concepts promoting AID in CVID will allow better and in some cases possibly even targeted treatment. A genetic diagnosis therefore becomes important information in this group of patients, especially in light of the fact that some patients might require hematopoietic stem cell transplantation because of their underlying immunodeficiency.
Collapse
|
138
|
Indications to Epigenetic Dysfunction in the Pathogenesis of Common Variable Immunodeficiency. Arch Immunol Ther Exp (Warsz) 2016; 65:101-110. [DOI: 10.1007/s00005-016-0414-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 06/10/2016] [Indexed: 12/12/2022]
|
139
|
Aryan Z, Aghamohammadi A, Rezaei N. Toward the stratification and personalization of common variable immunodeficiency treatment. Expert Opin Orphan Drugs 2016. [DOI: 10.1080/21678707.2016.1205480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
140
|
Boysen J, Nelson M, Magzoub G, Maiti GP, Sinha S, Goswami M, Vesely SK, Shanafelt TD, Kay NE, Ghosh AK. Dynamics of microvesicle generation in B-cell chronic lymphocytic leukemia: implication in disease progression. Leukemia 2016; 31:350-360. [PMID: 27480387 PMCID: PMC5288303 DOI: 10.1038/leu.2016.217] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2016] [Revised: 06/16/2016] [Accepted: 07/15/2016] [Indexed: 12/11/2022]
Abstract
Previously, we reported that B-cell chronic lymphocytic leukemia (CLL) patients contained elevated levels of microvesicles (MVs). However, given the quiescent nature of CLL B-cells and the relative indolence of the disease, the dynamics of MV generation and their unique phenotypes are not clearly defined. In this study, we find that CLL B-cells generate MVs spontaneously and can be further induced by B-cell receptor-ligation. Most interestingly, CLL B-cells predominantly generate CD52+ MVs, but not CD19+ MVs in vitro, suggesting preferential usage of CD52 into leukemic-MVs and that the CLL plasma MV phenotypes corroborate well with the in vitro findings. Importantly, we detected increased accumulation of CD52+ MVs in previously untreated CLL patients with progressive disease. Finally, sequential studies on MVs in pre- and post-therapy CLL patients demonstrate that while the plasma CD52+ MV levels drop significantly after therapy in most and remain at low levels in some patients, a trend of increased accumulation of CD52+ MVs was detected in majority of post-therapy CLL patients (25 of 33). In total this study emphasizes that dynamic accumulation of CD52+ MVs in plasma can be used to study CLL progression and may be a useful biomarker for patients as they progress and require therapy.
Collapse
Affiliation(s)
- J Boysen
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - M Nelson
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - G Magzoub
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - G P Maiti
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - S Sinha
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - M Goswami
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - S K Vesely
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - T D Shanafelt
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - N E Kay
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - A K Ghosh
- Stephenson Cancer Center, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA.,Department of Pathology, The University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
141
|
Abstract
BACKGROUND Common variable immunodeficiency (CVID) is characterized by hypogammaglobulinemia, defective antibody production and recurrent upper and lower respiratory tract infections. The diagnosis in adult patients is often thought to be rare, and thus misdiagnosis often occurs. A limited number of cases of adult-onset CVID have been reported in China, and the features of the syndrome remain unclear. The objective of this study was to describe the main characteristics of CVID, and evaluate the treatment of adult patients who present with CVID. MATERIALS AND METHODS This was a retrospective analysis of 8 patients with CVID from different departments in 1 center in China. Patients were diagnosed according to the diagnostic criteria of the European Society for Immunodeficiency Diseases. Demographics, clinical and immunological data from each patient were collected and a statistical analysis was undertaken. RESULTS The mean age at diagnosis was 43 ± 13.7 years, whereas the mean duration of diagnostic delay was 10.5 years. The median total serum levels of immunoglobulin (Ig) G, IgA and IgM at diagnosis were 2.5 ± 0.59, 0.23 ± 0.05 and 0.17 ± 0.05g/L, respectively. A total of 7 patients also had a low CD4(+)/CD8(+) ratio. All patients presented with recurrent respiratory infections. Regular infusions of intravenous immunoglobulin every 3 weeks substantially reduced pneumonic episodes. CONCLUSIONS Diagnosis is often delayed in adult CVID. Pulmonary infections and diseases were the most frequent presentations at onset of the disease. Regular intravenous immunoglobulin infusions were beneficial in controlling recurrent infections.
Collapse
|
142
|
Autoimmunity and infection in common variable immunodeficiency (CVID). Autoimmun Rev 2016; 15:877-82. [PMID: 27392505 DOI: 10.1016/j.autrev.2016.07.011] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 06/05/2016] [Indexed: 11/23/2022]
Abstract
Common variable immunodeficiency (CVID) is a heterogeneous group of diseases, characterized by primary hypogammaglobulinemia. B and T cell abnormalities have been described in CVID. Typical clinical features of CVID are recurrent airway infections; lymphoproliferative, autoinflammatory, or neoplastic disorders; and autoimmune diseases among which autoimmune thrombocytopenia (ITP) is the most common. The coexistence of immunodeficiency and autoimmunity appears paradoxical, since one represents a hypoimmune state and the other a hyperimmune state. Considering both innate and adaptive immune response abnormalities in CVID, it is easier to understand the mechanisms that lead to a breakdown of self-tolerance. CD21(low) B cells derive from mature B cells that have undergone chronic immune stimulation; they are increased in CVID patients. The expansion of CD21(low) B cells is also observed in certain autoimmune diseases. We have studied CD21(low) B cells in patients with CVID, CVID, and ITP and with ITP only. We observed a statistically significant increase in the CD21(low) population in the three pathological groups. Moreover, we found statistical differences between the two groups of CVID patients: patients with ITP had a higher percentage of CD21(low) cells. Our data suggest that CD21(low) cells are related to autoimmunity and may represent a link between infection and autoimmunity.
Collapse
|
143
|
The early activation of memory B cells from Wiskott-Aldrich syndrome patients is suppressed by CD19 downregulation. Blood 2016; 128:1723-34. [PMID: 27330000 DOI: 10.1182/blood-2016-03-703579] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 06/06/2016] [Indexed: 11/20/2022] Open
Abstract
Wiskott-Aldrich syndrome (WAS) pediatric patients exhibit a deficiency in humoral immune memory. However, the mechanism by which Wiskott-Aldrich syndrome protein (WASP) regulates the differentiation and activation of memory B cells remains elusive. Here we examine the early activation events of memory B cells from the peripheral blood mononuclear cells of WAS patients and age-matched healthy controls (HCs) using total internal reflection fluorescence microscopy. In response to stimulation through the B-cell receptor (BCR), memory B cells from HCs showed significantly higher magnitudes of BCR clustering and cell spreading than naive B cells from the same individuals. This was associated with increases in CD19 recruitment to the BCR and the activation of its downstream signaling molecule Btk and decreases in FcγRIIB recruitment and the activation of its downstream molecule Src homology 2-containing inositol 5' phosphatase (SHIP). However, these enhanced signaling activities mediated by CD19 and Btk are blocked in memory B cells from WAS patients, whereas the activation of FcγRIIB and SHIP was increased. Although the expression levels of CD19, Btk, and FcγRIIB did not change between CD27(-) and CD27(+) B cells of HCs, the protein and mRNA levels of CD19 but not Btk and FcγRIIB were significantly reduced in both CD27(-) and CD27(+) B cells of WAS patients, compared with those of HCs. Overall, our study suggests that WASP is required for memory B-cell activation, promoting the activation by positive regulating CD19 transcription and CD19 recruitment to the BCR.
Collapse
|
144
|
Understanding the genetic and epigenetic basis of common variable immunodeficiency disorder through omics approaches. Biochim Biophys Acta Gen Subj 2016; 1860:2656-63. [PMID: 27316315 DOI: 10.1016/j.bbagen.2016.06.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2016] [Revised: 05/26/2016] [Accepted: 06/09/2016] [Indexed: 11/20/2022]
Abstract
BACKGROUND Common variable immunodeficiency disorder (CVID) is the most frequently encountered symptomatic primary immunodeficiency, characterized by highly heterogeneous immunological features and clinical presentations. As better targeted therapies are importantly needed for CVID, improved understanding of the genetic and epigenetic basis for the development of CVID presents the most promising venue for improvement. SCOPE OF REVIEW Several genomic and epigenomic studies of CVID have recently been carried out on cohorts of sporadic cases of CVID. Using high-throughput array and sequencing technologies, these studies identified several loci associated with the disease. Here, we review the omics approaches used in these studies and resulting discoveries. We also discuss how these findings lead to improved understanding of the molecular basis of CVID and possible future directions to pursue. MAJOR CONCLUSIONS High-throughput omics approaches have been productive in genetic and epigenetic studies of CVID, leading to the identifications of several significantly associated loci of different variant types, as well as genes and pathways elucidating the shared genetic basis of CVID and autoimmunity. Complex polygenic model of inheritance together with interplay between genetic components and environmental factors may account for the etiology of CVID and various associated comorbidities. GENERAL SIGNIFICANCE The genetic and epigenetic basis of CVID when further translated through functional studies will allow for improved understanding of the CVID etiology and will provide new insights into the development of potential new therapeutic approaches for this devastating condition. This article is part of a Special Issue entitled "System Genetics" Guest Editor: Dr. Yudong Cai and Dr. Tao Huang.
Collapse
|
145
|
Bogaert DJA, Dullaers M, Lambrecht BN, Vermaelen KY, De Baere E, Haerynck F. Genes associated with common variable immunodeficiency: one diagnosis to rule them all? J Med Genet 2016; 53:575-90. [PMID: 27250108 DOI: 10.1136/jmedgenet-2015-103690] [Citation(s) in RCA: 196] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2016] [Accepted: 05/10/2016] [Indexed: 12/15/2022]
Abstract
Common variable immunodeficiency (CVID) is a primary antibody deficiency characterised by hypogammaglobulinaemia, impaired production of specific antibodies after immunisation and increased susceptibility to infections. CVID shows a considerable phenotypical and genetic heterogeneity. In contrast to many other primary immunodeficiencies, monogenic forms count for only 2-10% of patients with CVID. Genes that have been implicated in monogenic CVID include ICOS, TNFRSF13B (TACI), TNFRSF13C (BAFF-R), TNFSF12 (TWEAK), CD19, CD81, CR2 (CD21), MS4A1 (CD20), TNFRSF7 (CD27), IL21, IL21R, LRBA, CTLA4, PRKCD, PLCG2, NFKB1, NFKB2, PIK3CD, PIK3R1, VAV1, RAC2, BLK, IKZF1 (IKAROS) and IRF2BP2 With the increasing number of disease genes identified in CVID, it has become clear that CVID is an umbrella diagnosis and that many of these genetic defects cause distinct disease entities. Moreover, there is accumulating evidence that at least a subgroup of patients with CVID has a complex rather than a monogenic inheritance. This review aims to discuss current knowledge regarding the molecular genetic basis of CVID with an emphasis on the relationship with the clinical and immunological phenotype.
Collapse
Affiliation(s)
- Delfien J A Bogaert
- Clinical Immunology Research Lab, Department of Pulmonary Medicine, Ghent University Hospital, Ghent, Belgium Department of Pediatric Immunology and Pulmonology, Centre for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium
| | - Melissa Dullaers
- Clinical Immunology Research Lab, Department of Pulmonary Medicine, Ghent University Hospital, Ghent, Belgium Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Bart N Lambrecht
- Laboratory of Immunoregulation, VIB Inflammation Research Center, Ghent, Belgium Department of Internal Medicine, Ghent University, Ghent, Belgium
| | - Karim Y Vermaelen
- Clinical Immunology Research Lab, Department of Pulmonary Medicine, Ghent University Hospital, Ghent, Belgium Department of Internal Medicine, Ghent University, Ghent, Belgium Tumor Immunology Laboratory, Department of Pulmonary Medicine, Ghent University Hospital, Ghent, Belgium
| | - Elfride De Baere
- Center for Medical Genetics, Ghent University and Ghent University Hospital, Ghent, Belgium
| | - Filomeen Haerynck
- Clinical Immunology Research Lab, Department of Pulmonary Medicine, Ghent University Hospital, Ghent, Belgium Department of Pediatric Immunology and Pulmonology, Centre for Primary Immunodeficiency, Jeffrey Modell Diagnosis and Research Centre, Ghent University Hospital, Ghent, Belgium
| |
Collapse
|
146
|
Abstract
Autoimmune diseases represent a heterogeneous group of common disorders defined by complex trait genetics and environmental effects. The genetic variants usually align in immune and metabolic pathways that affect cell survival or apoptosis and modulate leukocyte function. Nevertheless, the exact triggers of disease development remain poorly understood and the current therapeutic interventions only modify the disease course. Both the prevention and the cure of autoimmune disorders are beyond our present medical capabilities. In contrast, a growing number of single gene autoimmune disorders have also been identified and characterized in the last few decades. Mutations and other gene alterations exert significant effects in these conditions, and often affect genes involved in central or peripheral immunologic tolerance induction. Even though a single genetic abnormality may be the disease trigger, it usually upsets a number of interactions among immune cells, and the biological developments of these monogenic disorders are also complex. Nevertheless, identification of the triggering molecular abnormalities greatly contributes to our understanding of the pathogenesis of autoimmunity and facilitates the development of newer and more effective treatment strategies.
Collapse
Affiliation(s)
- Mark Plander
- a Markusovszky University Teaching Hospital , Szombathely , Hungary and
| | - Bernadette Kalman
- a Markusovszky University Teaching Hospital , Szombathely , Hungary and.,b University of Pecs , Pecs , Hungary
| |
Collapse
|
147
|
Gallagher S, Turman S, Yusuf I, Akhgar A, Wu Y, Roskos LK, Herbst R, Wang Y. Pharmacological profile of MEDI-551, a novel anti-CD19 antibody, in human CD19 transgenic mice. Int Immunopharmacol 2016; 36:205-212. [PMID: 27163209 DOI: 10.1016/j.intimp.2016.04.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2016] [Revised: 04/15/2016] [Accepted: 04/21/2016] [Indexed: 10/21/2022]
Abstract
B cell depletion therapy is beneficial for patients with B cell malignancies and autoimmune diseases. CD19, a transmembrane protein, is expressed on a vast majority of normal and neoplastic B cells, making it a suitable target for monoclonal antibody (MAb) mediated immunotherapy. We have developed MEDI-551, an affinity optimized and afucosylated IgG1 MAb targeting human CD19 for B cell depletion. MEDI-551 is currently under investigation in multiple clinical trials. Because MEDI-551 does not cross react with rodent and non-human primate CD19, the pharmacological characteristics of the MAb were evaluated in human CD19 transgenic mice (hCD19 Tg). Here we show that MEDI-551 potently depletes tissue and circulating B cells in hCD19 Tg mice and is more efficacious than the anti-CD19 MAb with intact fucose. The length of B cell depletion depends on MEDI-551 dose; and, B cell recovery in the circulation follows stepwise phenotypic maturation. Furthermore, intravenous (IV) and subcutaneous (SC) administration of MEDI-551 results in comparable efficacy. Lastly, the combination of MEDI-551 with the anti-CD20 MAb, rituximab, further prolongs the duration of B cell depletion. In summary, the pharmacological profile of MEDI-551 presented in hCD19 Tg mice supports further testing of MEDI-551 in clinical trials involving B cell malignancies and autoimmune diseases.
Collapse
Affiliation(s)
- Sandra Gallagher
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Sean Turman
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Isharat Yusuf
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Ahmad Akhgar
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, United States
| | - Yuling Wu
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, United States
| | - Lorin K Roskos
- Translational Sciences, MedImmune LLC, Gaithersburg, MD, United States
| | - Ronald Herbst
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States
| | - Yue Wang
- Respiratory, Inflammation and Autoimmune Research, Gaithersburg, MD, United States.
| |
Collapse
|
148
|
Abstract
B cells have diverse functions during immune responses, including antibody production, antigen presentation, and cytokine secretion. Multiple lymphomas and leukemias derive from malignant B cells, so therapies that deplete B cells are clinically important, particularly antibodies targeting the B cell-specific surface molecules CD19 and CD20. Macrophages are the principal mediators of CD19 and CD20 monoclonal antibody-dependent B-cell and lymphoma depletion in mice through Fcγ receptor-dependent phagocytosis. Thereby, the extent of CD19 or CD20 antibody-induced B cell and tumor depletion in vivo is influenced by molecular changes within tumors and genetic variations between individuals. In addition to Fcγ receptor polymorphisms, lymphoma- and regulatory B cell-derived cytokine production and macrophage localization and function within tumor microenvironments influence tumor clearance. Given the dynamic interactions of these factors, the identification of effector cell and tumor microenvironment genetic alterations will identify molecular targets that enhance immunotherapies for the treatment of human diseases.
Collapse
|
149
|
Berrón-Ruiz L, López-Herrera G, Vargas-Hernández A, Santos-Argumedo L, López-Macías C, Isibasi A, Segura-Méndez NH, Bonifaz L. Impaired selective cytokine production by CD4+ T cells in Common Variable Immunodeficiency associated with the absence of memory B cells. Clin Immunol 2016; 166-167:19-26. [DOI: 10.1016/j.clim.2016.03.013] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Revised: 03/18/2016] [Accepted: 03/28/2016] [Indexed: 11/28/2022]
|
150
|
Keller MD, Pandey R, Li D, Glessner J, Tian L, Henrickson SE, Chinn IK, Monaco-Shawver L, Heimall J, Hou C, Otieno FG, Jyonouchi S, Calabrese L, van Montfrans J, Orange JS, Hakonarson H. Mutation in IRF2BP2 is responsible for a familial form of common variable immunodeficiency disorder. J Allergy Clin Immunol 2016; 138:544-550.e4. [PMID: 27016798 DOI: 10.1016/j.jaci.2016.01.018] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Revised: 01/01/2016] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
BACKGROUND Genome-wide association studies have shown a pattern of rare copy number variations and single nucleotide polymorphisms in patients with common variable immunodeficiency disorder (CVID), which was recognizable by a support vector machine (SVM) algorithm. However, rare monogenic causes of CVID might lack such a genetic fingerprint. OBJECTIVE We sought to identify a unique monogenic cause of familial immunodeficiency and evaluate the use of SVM to identify patients with possible monogenic disorders. METHODS A family with multiple members with a diagnosis of CVID was screened by using whole-exome sequencing. The proband and other subjects with mutations associated with CVID-like phenotypes were screened through the SVM algorithm from our recent CVID genome-wide association study. RT-PCR, protein immunoblots, and in vitro plasmablast differentiation assays were performed on patient and control EBV lymphoblastoids cell lines. RESULTS Exome sequencing identified a novel heterozygous mutation in IRF2BP2 (c.1652G>A:p.[S551N]) in affected family members. Transduction of the mutant gene into control human B cells decreased production of plasmablasts in vitro, and IRF2BP2 transcripts and protein expression were increased in proband versus control EBV-immortalized lymphoblastoid cell lines. The SVM algorithm categorized the proband and subjects with other immunodeficiency-associated gene variants in TACI, BAFFR, ICOS, CD21, LRBA, and CD27 as genetically dissimilar from polygenic CVID. CONCLUSION A novel IRFBP2 mutation was identified in a family with autosomal dominant CVID. Transduction experiments suggest that the mutant protein has an effect on B-cell differentiation and is likely a monogenic cause of the family's CVID phenotype. Successful grouping by the SVM algorithm suggests that our family and other subjects with rare immunodeficiency disorders cluster separately and lack the genetic pattern present in polygenic CVID cases.
Collapse
Affiliation(s)
- Michael D Keller
- Division of Allergy and Immunology, Children's National Medical Center, Washington, DC
| | - Rahul Pandey
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Dong Li
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Joseph Glessner
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Lifeng Tian
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Sarah E Henrickson
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Ivan K Chinn
- Division of Immunology, Allergy, and Rheumatology, Texas Children's Hospital, Houston, Tex; Baylor Genomics Institute, Baylor College of Medicine, Houston, Tex
| | - Linda Monaco-Shawver
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa; Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Jennifer Heimall
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Cuiping Hou
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Frederick G Otieno
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Soma Jyonouchi
- Division of Allergy and Immunology, Children's Hospital of Philadelphia, Philadelphia, Pa
| | - Leonard Calabrese
- Department of Rheumatologic and Immunologic Disease, Cleveland Clinic, Cleveland, Ohio
| | - Joris van Montfrans
- Department of Pediatric Immunology and Infectious Diseases, Wilhelmina Children's Hospital, University Medical Center, Utrecht, The Netherlands
| | - Jordan S Orange
- Division of Immunology, Allergy, and Rheumatology, Texas Children's Hospital, Houston, Tex.
| | - Hakon Hakonarson
- Center for Applied Genomics, Children's Hospital of Philadelphia, Philadelphia, Pa.
| |
Collapse
|