1
|
Aranda CS, Gouveia-Pereira MP, da Silva CJM, Rizzo MCFV, Ishizuka E, de Oliveira EB, Condino-Neto A. Severe combined immunodeficiency diagnosis and genetic defects. Immunol Rev 2024; 322:138-147. [PMID: 38287514 DOI: 10.1111/imr.13310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/02/2024] [Indexed: 01/31/2024]
Abstract
Severe combined immunodeficiency (SCID) is a rare and life-threatening genetic disorder that severely impairs the immune system's ability to defend the body against infections. Often referred to as the "bubble boy" disease, SCID gained widespread recognition due to the case of David Vetter, a young boy who lived in a sterile plastic bubble to protect him from germs. SCID is typically present at birth, and it results from genetic mutations that affect the development and function of immune cells, particularly T cells and B cells. These immune cells are essential for identifying and fighting off infections caused by viruses, bacteria, and fungi. In SCID patients, the immune system is virtually non-existent, leaving them highly susceptible to recurrent, severe infections. There are several forms of SCID, with varying degrees of severity, but all share common features. Newborns with SCID often exhibit symptoms such as chronic diarrhea, thrush, skin rashes, and persistent infections that do not respond to standard treatments. Without prompt diagnosis and intervention, SCID can lead to life-threatening complications and a high risk of mortality. There are over 20 possible affected genes. Treatment options for SCID primarily involve immune reconstitution, with the most well-known approach being hematopoietic stem cell transplantation (HSCT). Alternatively, gene therapy is also available for some forms of SCID. Once treated successfully, SCID patients can lead relatively normal lives, but they may still require vigilant infection control measures and lifelong medical follow-up to manage potential complications. In conclusion, severe combined immunodeficiency is a rare but life-threatening genetic disorder that severely compromises the immune system's function, rendering affected individuals highly vulnerable to infections. Early diagnosis and appropriate treatment are fundamental. With this respect, newborn screening is progressively and dramatically improving the prognosis of SCID.
Collapse
Affiliation(s)
- Carolina Sanchez Aranda
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Federal University of São Paulo Medical School-UNIFESP, São Paulo, Brazil
| | - Mariana Pimentel Gouveia-Pereira
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Federal University of São Paulo Medical School-UNIFESP, São Paulo, Brazil
| | - Celso Jose Mendanha da Silva
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Federal University of São Paulo Medical School-UNIFESP, São Paulo, Brazil
| | - Maria Candida Faria Varanda Rizzo
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics, Federal University of São Paulo Medical School-UNIFESP, São Paulo, Brazil
| | | | | | - Antonio Condino-Neto
- Department of Immunology, Institute of Biomedical Sciences, University of Sao Paulo, Instituto Jo Clemente, and Immunogenic Laboratories, São Paulo, Brazil
| |
Collapse
|
2
|
Shah I, Chiang S, Yang L, Akeno N, Kelly A, White J, Caywood E, Hwang S, Le T. γδ CD8+ T cells and novel genetic variants in ZAP70 deficiency. Pediatr Allergy Immunol 2023; 34:e14035. [PMID: 37877847 DOI: 10.1111/pai.14035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/20/2023] [Accepted: 09/26/2023] [Indexed: 10/26/2023]
Affiliation(s)
- Isma Shah
- Division of Allergy & Immunology, Nemours Children's Hospital, Wilmington, Delaware, USA
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Samuel Chiang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Li Yang
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Nagako Akeno
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, USA
| | - Allison Kelly
- Division of Allergy & Immunology, Nemours Children's Hospital, Wilmington, Delaware, USA
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Jason White
- Department of Pediatrics, University of Cincinnati, Cincinnati, Ohio, USA
| | - Emi Caywood
- Division of Pediatric Hematology/Oncology, Nemours Children's Hospital, Wilmington, Delaware, USA
| | - Sharon Hwang
- Division of Allergy & Immunology, Nemours Children's Hospital, Wilmington, Delaware, USA
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| | - Trong Le
- Division of Allergy & Immunology, Nemours Children's Hospital, Wilmington, Delaware, USA
- Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
3
|
Staudacher O, Klein J, Thee S, Ullrich J, Wahn V, Unterwalder N, Kölsch U, Lankes E, Stittrich A, Dedieu C, Dinges S, Völler M, Schuetz C, Schulte J, Boztug K, Meisel C, Kuehl JS, Krüger R, Blankenstein O, von Bernuth H. Screening Newborns for Low T Cell Receptor Excision Circles (TRECs) Fails to Detect Immunodeficiency, Centromeric Instability, and Facial Anomalies Syndrome. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:2872-2883. [PMID: 37302792 DOI: 10.1016/j.jaip.2023.06.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/30/2023] [Accepted: 06/01/2023] [Indexed: 06/13/2023]
Abstract
BACKGROUND Assessment of T-cell receptor excision circles (TRECs) in dried blood spots of newborns allows the detection of severe combined immunodeficiency (SCID) (T cells <300/μL at birth) with a presumed sensitivity of 100%. TREC screening also identifies patients with selected combined immunodeficiency (CID) (T cells >300/μL, yet <1500/μL at birth). Nevertheless, relevant CIDs that would benefit from early recognition and curative treatment pass undetected. OBJECTIVE We hypothesized that TREC screening at birth cannot identify CIDs that develop with age. METHODS We analyzed the number of TRECs in dried blood spots in archived Guthrie cards of 22 children who had been born in the Berlin-Brandenburg area between January 2006 and November 2018 and who had undergone hematopoietic stem-cell transplantation (HSCT) for inborn errors of immunity. RESULTS All patients with SCID would have been identified by TREC screening, but only 4 of 6 with CID. One of these patients had immunodeficiency, centromeric instability, and facial anomalies syndrome type 2 (ICF2). Two of 3 patients with ICF whom we have been following up at our institution had TREC numbers above the cutoff value suggestive of SCID at birth. Yet all patients with ICF had a severe clinical course that would have justified earlier HSCT. CONCLUSIONS In ICF, naïve T cells may be present at birth, yet they decline with age. Therefore, TREC screening cannot identify these patients. Early recognition is nevertheless crucial, as patients with ICF benefit from HSCT early in life.
Collapse
Affiliation(s)
- Olga Staudacher
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Jeanette Klein
- Newborn Screening Laboratory, Charité Universitätsmedizin, Berlin, Germany
| | - Stephanie Thee
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Jan Ullrich
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Volker Wahn
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Nadine Unterwalder
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Uwe Kölsch
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Erwin Lankes
- Newborn Screening Laboratory, Charité Universitätsmedizin, Berlin, Germany; Department of Pediatric Endocrinology, Charité-Uninrsitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Anna Stittrich
- Department of Human Genetics, Labor Berlin Charité-Vivantes, Berlin, Germany
| | - Cinzia Dedieu
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Sarah Dinges
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Mirjam Völler
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Johannes Schulte
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; St. Anna Children's Cancer Research Institute, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, St. Anna Children's Hospital, Medical University of Vienna, Vienna, Austria
| | - Christian Meisel
- Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany; Institute of Medical Immunology, Charité-Universitätsmedizin Berlin, Berlin, Germany
| | - Jörn-Sven Kuehl
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Pediatric Oncology, Hematology and Hemostaseology, University of Leipzig, Leipzig, Germany
| | - Renate Krüger
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany
| | | | - Horst von Bernuth
- Department of Pediatric Respiratory Medicine, Immunology and Critical Care Medicine, Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany; Department of Immunology, Labor Berlin Charité-Vivantes, Berlin, Germany; Berlin Institute of Health (BIH), Charité-Universitätsmedizin Berlin, Berlin, Germany; Berlin-Brandenburg Center for Regenerative Therapies (BCRT), Charité-Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health (BIH), Berlin, Germany.
| |
Collapse
|
4
|
Infections in Inborn Errors of Immunity with Combined Immune Deficiency: A Review. Pathogens 2023; 12:pathogens12020272. [PMID: 36839544 PMCID: PMC9958715 DOI: 10.3390/pathogens12020272] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2022] [Revised: 01/13/2023] [Accepted: 02/04/2023] [Indexed: 02/10/2023] Open
Abstract
Enhanced susceptibility to microbes, often resulting in severe, intractable and frequent infections due to usually innocuous organisms at uncommon sites, is the most striking feature in individuals with an inborn error of immunity. In this narrative review, based on the International Union of Immunological Societies' 2022 (IUIS 2022) Update on phenotypic classification of human inborn errors of immunity, the focus is on commonly encountered Combined Immunodeficiency Disorders (CIDs) with susceptibility to infections. Combined immune deficiency disorders are usually commensurate with survival beyond infancy unlike Severe Combined Immune Deficiency (SCID) and are often associated with clinical features of a syndromic nature. Defective humoral and cellular immune responses result in susceptibility to a broad range of microbial infections. Although disease onset is usually in early childhood, mild defects may present in late childhood or even in adulthood. A precise diagnosis is imperative not only for determining management strategies, but also for providing accurate genetic counseling, including prenatal diagnosis, and also in deciding empiric treatment of infections upfront before investigation reports are available.
Collapse
|
5
|
Gupta S, Agrawal A. Dendritic cells in inborn errors of immunity. Front Immunol 2023; 14:1080129. [PMID: 36756122 PMCID: PMC9899832 DOI: 10.3389/fimmu.2023.1080129] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 01/06/2023] [Indexed: 01/24/2023] Open
Abstract
Dendritic cells (DCs) are crucial cells for initiating and maintaining immune response. They play critical role in homeostasis, inflammation, and autoimmunity. A number of molecules regulate their functions including synapse formation, migration, immunity, and induction of tolerance. A number of IEI are characterized by mutations in genes encoding several of these molecules resulting in immunodeficiency, inflammation, and autoimmunity in IEI. Currently, there are 465 Inborn errors of immunity (IEI) that have been grouped in 10 different categories. However, comprehensive studies of DCs have been reported in only few IEI. Here we have reviewed biology of DCs in IEI classified according to recently published IUIS classification. We have reviewed DCs in selected IEI in each group category and discussed in depth changes in DCs where significant data are available regarding role of DCs in clinical and immunological manifestations. These include severe immunodeficiency diseases, antibody deficiencies, combined immunodeficiency with associated and syndromic features, especially disorders of synapse formation, and disorders of immune regulation.
Collapse
Affiliation(s)
- Sudhir Gupta
- Division of Basic and Clinical Immunology, University of California, Irvine, CA, United States
| | | |
Collapse
|
6
|
FASCIA Method in the Assessment of Lymphocyte Mitogen Responses in the Laboratory Diagnostics of Primary Immunodeficiencies. J Clin Immunol 2023; 43:653-661. [PMID: 36512178 PMCID: PMC9958160 DOI: 10.1007/s10875-022-01417-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022]
Abstract
Lymphocyte responses to mitogens constitute a key part of the diagnostics of combined immunodeficiency (CID). Currently, mostly radioactive thymidine incorporation and carboxyfluorescein diacetate succinimidyl ester (CFSE) dilution methods are used. Flow-cytometric assay for specific cell-mediated immune-response in activated whole blood (FASCIA) has been put forth as an easy-to-perform option for the measurement of lymphocyte responses with the advantage of recognizing different lymphocyte subtypes and avoiding the use of radioactive reagents. Our aim was to analyze retrospectively the usefulness of FASCIA in the diagnostics of CID. We included all lymphocyte stimulation tests done with FASCIA in HUSLAB (Helsinki, Finland) between February 2015 and September 2018 in our analysis. The cohort was divided into two groups according to the patients' final diagnoses: CID (n = 30) or non-CID (n = 159). We evaluated the stimulation responses with a combined FASCIA score (the average of all mitogen responses). The FASCIA score was significantly lower among the CID group compared to the other patients (p = 0.002), and in the ROC analysis, the AUC was 0.75 (p < 0.001) for the FASCIA score. When the three mitogens were analyzed separately, phytohemagglutinin (PHA) was best in separating patients with CID from non-CID (in the ROC analysis AUC 0.71, p = 0.001). Immunosuppressive medication affected the FASCIA result significantly and needs to be considered when evaluating the results. In conclusion, FASCIA can reliably detect the CID patients in the absence of immunosuppressive medication. It emerges as a method with many benefits compared to tests requiring radioactive reagents or the complicated CFSE staining.
Collapse
|
7
|
Damoiseaux M, Damoiseaux J, Pico-Knijnenburg I, van der Burg M, Bredius R, van Well G. Lessons learned from the diagnostic work-up of a patient with the bare lymphocyte syndrome type II. Clin Immunol 2022; 235:108932. [DOI: 10.1016/j.clim.2022.108932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2021] [Accepted: 01/15/2022] [Indexed: 11/03/2022]
|
8
|
Reynolds A, Gaughan M, Holden D, Redenbaugh V, Dunne J, Redmond J, Conlon N. The effects of dimethyl fumarate and fingolimod on T-cell lymphocyte proliferation in patients with multiple sclerosis. Ir J Med Sci 2022; 191:2759-2762. [PMID: 35028898 DOI: 10.1007/s11845-021-02913-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2021] [Accepted: 12/25/2021] [Indexed: 11/28/2022]
Abstract
OBJECTIVE The disease-modifying therapies (DMT), dimethyl fumarate (DMF) and fingolimod (FTY) improve the outcomes in multiple sclerosis (MS) by reducing relapses and numbers and volume of lesions. They mediate their effects through reduction of immune reactivation, which may potentially lead to lymphopaenia and increased risk of infections. Previous studies have examined the effects of these therapies on lymphocyte subsets; however, the in vivo effects on circulating lymphocyte proliferation require further elucidation. The aim of this study was to determine the effects of DMF and FTY on T-cell proliferation in patients with MS. METHOD We examined T-cell lymphocyte proliferation and lymphocyte subsets in ten patients (five on DMF, five on FTY) before starting DMT and again 4 to 11 months after being maintained on DMT. RESULTS In the FTY-treated group, the mean percentage proliferation was significantly lower using both assays (PHA assay mean percentage change - 51.2 ± 25.97, p < 0.05; anti-CD3/CD28 assay mean percentage change - 39.74 ± 27.85, p < 0.05). There was no statistical difference in T-cell lymphocyte proliferation in the DMF-treated group for either assay (PHA, p = 0.316; anti-CD3/CD28, p = 0.373). CONCLUSIONS This pilot study suggests that the T-lymphocytes of patients on FTY have an abnormal proliferation response as well as being reduced in the circulation.
Collapse
Affiliation(s)
- Audrey Reynolds
- Department of Neurology, St James's Hospital, Dublin 8, Ireland.
| | - Maria Gaughan
- Department of Neurology, St James's Hospital, Dublin 8, Ireland
| | - Dean Holden
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| | | | - Jean Dunne
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| | - Janice Redmond
- Department of Neurology, St James's Hospital, Dublin 8, Ireland
| | - Niall Conlon
- Department of Immunology, St James's Hospital, Dublin 8, Ireland
| |
Collapse
|
9
|
El Allam A, El Fakihi S, Tahoune H, Sahmoudi K, Bousserhane H, Bakri Y, El Hafidi N, Seghrouchni F. Cytometric analysis and clinical features in a Moroccan cohort with severe combined immunodeficiency. Hum Antibodies 2022; 30:67-77. [PMID: 35094990 DOI: 10.3233/hab-211510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Severe combined immunodeficiency (SCID) is a form of primary immunodeficiency disease (PID). It is characterized by a serious abnormality of the cellular and sometimes humoral system due to a deficiency in development of T cells, B cells and/or NK cells. The early diagnosis of SCID improves the prognosis. Typically, the initial consideration of SCID is made based on low lymphocyte counts. Notwithstanding, the heterogeneity of lymphocyte count presentation makes the diagnosis of SCID a significant challenge. The objective of this cross-sectional retrospective study was to analyze the lymphocyte subpopulation counts along with clinical manifestations within a Moroccan cohort diagnosed as SCID compared to children diagnosed with non-PID diseases. Thirty-five SCID confirmed patients were selected in the period between 2008 and 2018 and compared with non-PID patients. Results of peripheral blood T, B, and NK lymphocyte subpopulation counts were measured by flow cytometry for each SCID subtype. As expected, T cell count was less than 300 cells/μL in most patients with SCID (85.5%). Unexpectedly, significantly higher T cell counts were detected in some patients with a confirmed clinical diagnosis and family history of SCID. 5.7% of our SCID Moroccan cohort had T cell numbers in the range between 300 and 500 cells/μL. 8.7% of our SCID Moroccan cohort had T cell numbers higher than 500 cells/μL. Of the SCID subtypes, the proportion of SCID with B cell deficiencies was highly represented in our cohort. 71.4% of Moroccan SCID patients (25 out of 35 patients) were of T-B-subtype. Furthermore, 40% of the patients (14 out of 35 patients) had a T-B-NK+ profile and 31.4% had a T-B-NK- profile (11 out of 35 patients). The most common clinical manifestations observed in our SCID cohort were pneumonia, failure to thrive, candidiasis, diarrhea, bronchitis and urinary tract infections. Our results not only highlight the relatively frequent presence of atypical SCID in the Moroccan population with unexpectedly high T cell numbers, but also describes the incidence pattern of common SCID subtypes in Morocco. Physicians in Morocco may find this local region-specific difference in SCID important for making improved early diagnosis of this disease.
Collapse
Affiliation(s)
- Aicha El Allam
- Laboratory of Cellular Immunology, National Institute of Hygiene, Rabat, Morocco
- Laboratory of Biology and Human Pathology, Faculty of Sciences, University Mohammed V, Rabat, Morocco
| | - Sara El Fakihi
- Laboratory of Cellular Immunology, National Institute of Hygiene, Rabat, Morocco
- Med Biotech Laboratory, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Hicham Tahoune
- Department of Biology, Faculty of Sciences, University Ibn Tofail, Kenitra, Morocco
| | - Karima Sahmoudi
- Laboratory of Cellular Immunology, National Institute of Hygiene, Rabat, Morocco
- Department of Biology, Faculty of Sciences, University Mohammed V, Rabat, Morocco
| | - Houria Bousserhane
- Laboratory of Cellular Immunology, National Institute of Hygiene, Rabat, Morocco
- Med Biotech Laboratory, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Youssef Bakri
- Laboratory of Biology and Human Pathology, Faculty of Sciences, University Mohammed V, Rabat, Morocco
- Centre of Human Pathology Genomic, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| | - Naima El Hafidi
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Immunology, Allergic and Respiratory Diseases Unit, Children's Hospital of Rabat, Ibn Sina University Hospital Centre, Rabat, Morocco
| | - Fouad Seghrouchni
- Laboratory of Cellular Immunology, National Institute of Hygiene, Rabat, Morocco
- Med Biotech Laboratory, Faculty of Medicine and Pharmacy, University Mohammed V, Rabat, Morocco
| |
Collapse
|
10
|
Belaid B, Lamara Mahammed L, Mohand Oussaid A, Migaud M, Khadri Y, Casanova JL, Puel A, Ben Halla N, Djidjik R. Case Report: Interleukin-2 Receptor Common Gamma Chain Defect Presented as a Hyper-IgE Syndrome. Front Immunol 2021; 12:696350. [PMID: 34248995 PMCID: PMC8264782 DOI: 10.3389/fimmu.2021.696350] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2021] [Accepted: 06/07/2021] [Indexed: 11/16/2022] Open
Abstract
X-linked severe combined immunodeficiency (X-SCID) is caused by mutations of IL2RG, the gene encoding the interleukin common gamma chain (IL-2Rγ or γc) of cytokine receptors for interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15, and IL-21. Hypomorphic mutations of IL2RG may cause combined immunodeficiencies with atypical clinical and immunological presentations. Here, we report a clinical, immunological, and functional characterization of a missense mutation in exon 1 (c.115G>A; p. Asp39Asn) of IL2RG in a 7-year-old boy. The patient suffered from recurrent sinopulmonary infections and refractory eczema. His total lymphocyte counts have remained normal despite skewed T cell subsets, with a pronounced serum IgE elevation. Surface expression of IL-2Rγ was reduced on his lymphocytes. Signal transducer and activator of transcription (STAT) phosphorylation in response to IL-2, IL-4, and IL-7 showed a partially preserved receptor function. T-cell proliferation in response to mitogens and anti-CD3/anti-CD28 monoclonal antibodies was significantly reduced. Further analysis revealed a decreased percentage of CD4+ T cells capable of secreting IFN-γ, but not IL-4 or IL-17. Studies on the functional consequences of IL-2Rγ variants are important to get more insight into the pathogenesis of atypical phenotypes which may lay the ground for novel therapeutic strategies.
Collapse
Affiliation(s)
- Brahim Belaid
- Department of Medical Immunology, Beni-Messous University Hospital Center, Algiers, Algeria.,Faculty of Medicine, Benyoucef Benkhedda University of Algiers 1, Algiers, Algeria
| | - Lydia Lamara Mahammed
- Department of Medical Immunology, Beni-Messous University Hospital Center, Algiers, Algeria.,Faculty of Medicine, Benyoucef Benkhedda University of Algiers 1, Algiers, Algeria
| | - Aida Mohand Oussaid
- Faculty of Medicine, Benyoucef Benkhedda University of Algiers 1, Algiers, Algeria.,Department of Pediatrics A, Beni-Messous University Hospital Center, Algiers, Algeria
| | - Melanie Migaud
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, INSERM UMR 1163, Paris, France.,Imagine Institute, University of Paris, Paris, France
| | - Yasmine Khadri
- Department of Pediatrics A, Beni-Messous University Hospital Center, Algiers, Algeria
| | - Jean Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, INSERM UMR 1163, Paris, France.,Imagine Institute, University of Paris, Paris, France.,St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University, New York, NY, United States.,Howard Hughes Medical Institute, New York, NY, United States
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Hospital for Sick Children, INSERM UMR 1163, Paris, France.,Imagine Institute, University of Paris, Paris, France.,St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University, New York, NY, United States
| | - Nafissa Ben Halla
- Faculty of Medicine, Benyoucef Benkhedda University of Algiers 1, Algiers, Algeria.,Department of Pediatrics A, Beni-Messous University Hospital Center, Algiers, Algeria
| | - Reda Djidjik
- Department of Medical Immunology, Beni-Messous University Hospital Center, Algiers, Algeria.,Faculty of Medicine, Benyoucef Benkhedda University of Algiers 1, Algiers, Algeria
| |
Collapse
|
11
|
Chaudhry IUH, Alshaer A, Al Jassas B, Alkhunizi A, Alsaiary M, AlMubayaedh TA, AlMalki AA, Almesfer A. Bronchopleural fistula in a 5- years old child with novel CARMIL 2 mutation: A rare disease and a rare case. Ann Med Surg (Lond) 2021; 66:102443. [PMID: 34150204 PMCID: PMC8193081 DOI: 10.1016/j.amsu.2021.102443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 11/24/2022] Open
Abstract
A five year girl had eczema and allergic rhinitis in the past, presented with a history of cough, shortness of breath for the last one month. Her chest -X-ray showed a left side pleural effusion, and a computed tomographic scan (CT) of the chest showed left side hydropneumothorax. Left side 21 Fr drain was inserted. Her clinical condition deteriorated despite antimicrobial therapy, and she required mechanical ventilatory support due to respiratory distress. She also developed a right-sided pneumothorax that was managed by inserting a 21 Fr chest drain. A video-assisted thoracoscopic VATS procedure was done to staple the lung bullae and drain the empyema. Her post-operative chest X-ray showed good lung expansion. Pleural fluid culture report was positive for candida. She was commenced on antifungal microbial therapy. Two days later, she developed again left side pneumothorax, which was again managed by left intercostal drain. We were unable to wean her off from mechanical ventilatory support due to a significant air leak due to bronchopleural fistula. A posterolateral thoracotomy was performed, and the bronchopleural fistula was closed. She was extubated the next day, and intercostal drains were removed on the 4th post-operative day.
Collapse
Affiliation(s)
- Ikram ul Haq Chaudhry
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Ahmed Alshaer
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Burair Al Jassas
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Amal Alkhunizi
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Mohammad Alsaiary
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Tasneem A. AlMubayaedh
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Abass A. AlMalki
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| | - Ahmed Almesfer
- Department of Pediatric Thoracic Surgery and Intensive Care Medicine, Infectious Disease, and Immunology, Maternity and Children Hospital, Dammam, Saudi Arabia
| |
Collapse
|
12
|
Cagdas D, Mayr D, Baris S, Worley L, Langley DB, Metin A, Aytekin ES, Atan R, Kasap N, Bal SK, Dmytrus J, Heredia RJ, Karasu G, Torun SH, Toyran M, Karakoc-Aydiner E, Christ D, Kuskonmaz B, Uçkan-Çetinkaya D, Uner A, Oberndorfer F, Schiefer AI, Uzel G, Deenick EK, Keller B, Warnatz K, Neven B, Durandy A, Sanal O, Ma CS, Özen A, Stepensky P, Tezcan I, Boztug K, Tangye SG. Genomic Spectrum and Phenotypic Heterogeneity of Human IL-21 Receptor Deficiency. J Clin Immunol 2021; 41:1272-1290. [PMID: 33929673 PMCID: PMC8086229 DOI: 10.1007/s10875-021-01031-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/29/2021] [Indexed: 01/04/2023]
Abstract
Biallelic inactivating mutations in IL21R causes a combined immunodeficiency that is often complicated by cryptosporidium infections. While eight IL-21R-deficient patients have been reported previously, the natural course, immune characteristics of disease, and response to hematopoietic stem cell transplantation (HSCT) remain to be comprehensively examined. In our study, we have collected clinical histories of 13 patients with IL-21R deficiency from eight families across seven centers worldwide, including five novel patients identified by exome or NGS panel sequencing. Eight unique mutations in IL21R were identified in these patients, including two novel mutations. Median age at disease onset was 2.5 years (0.5–7 years). The main clinical manifestations were recurrent bacterial (84.6%), fungal (46.2%), and viral (38.5%) infections; cryptosporidiosis-associated cholangitis (46.2%); and asthma (23.1%). Inflammatory skin diseases (15.3%) and recurrent anaphylaxis (7.9%) constitute novel phenotypes of this combined immunodeficiency. Most patients exhibited hypogammaglobulinemia and reduced proportions of memory B cells, circulating T follicular helper cells, MAIT cells and terminally differentiated NK cells. However, IgE levels were elevated in 50% of IL-21R-deficient patients. Overall survival following HSCT (6 patients, mean follow-up 1.8 year) was 33.3%, with pre-existing organ damage constituting a negative prognostic factor. Mortality of non-transplanted patients (n = 7) was 57.1%. Our detailed analysis of the largest cohort of IL-21R-deficient patients to date provides in-depth clinical, immunological and immunophenotypic features of these patients, thereby establishing critical non-redundant functions of IL-21/IL-21R signaling in lymphocyte differentiation, humoral immunity and host defense against infection, and mechanisms of disease pathogenesis due to IL-21R deficiency. Outcome following HSCT depends on prior chronic infections and organ damage, which should thus be considered as early as possible following molecular diagnosis.
Collapse
Affiliation(s)
- Deniz Cagdas
- Division of Pediatric Immunology, Department of Pediatrics, İhsan Doğramacı Children's Hospital, Hacettepe University Medical Faculty, Ankara, Turkey.
- Section of Pediatric Immunology, Institutes of Child Health, Health Science Institute, Hacettepe University, Ankara, Turkey.
| | - Daniel Mayr
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Safa Baris
- Department of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, İstanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Lisa Worley
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - David B Langley
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Ayse Metin
- Department of Pediatric Immunology, Ankara City Hospital, University of Health Sciences, Ankara, Turkey
| | - Elif Soyak Aytekin
- Division of Pediatric Immunology, Department of Pediatrics, İhsan Doğramacı Children's Hospital, Hacettepe University Medical Faculty, Ankara, Turkey
| | - Raziye Atan
- Department of Pediatrics, Hacettepe University Medical Faculty, 1031, Ankara, Turkey
| | - Nurhan Kasap
- Department of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, İstanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Sevgi Köstel Bal
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Jasmin Dmytrus
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Raul Jimenez Heredia
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Gulsun Karasu
- School of Medicine, Goztepe Medicalpark Hospital, Pediatric stem Cell Transplantation Unit, İstinye University, İstanbul, Turkey
| | - Selda Hancerli Torun
- İstanbul Medical Faculty, Pediatric Infectious Disease, Istanbul University, İstanbul, Turkey
| | - Muge Toyran
- Department of Pediatric Immunology, Ankara City Hospital, University of Health Sciences, Ankara, Turkey
| | - Elif Karakoc-Aydiner
- Department of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, İstanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Daniel Christ
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Baris Kuskonmaz
- Department of Pediatrics, Division of Pediatric Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Duygu Uçkan-Çetinkaya
- Department of Pediatrics, Division of Pediatric Hematology, Hacettepe University Medical School, Ankara, Turkey
| | - Aysegul Uner
- Department of Pathology, Hacettepe University Medical School, Ankara, Turkey
| | | | - Ana-Iris Schiefer
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Elissa K Deenick
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Baerbel Keller
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Department of Rheumatology and Clinical Immunology, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
- Center for Chronic Immunodeficiency (CCI), Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Bénédicte Neven
- Department of Pediatric Immunology, Hematology and Rheumatology, Necker Children Hospital, Paris, France
| | - Anne Durandy
- Laboratory of Human Lymphohematopoiesis, Imagine Institute, INSERM UMR 1163, Université de Paris, Paris, France
| | - Ozden Sanal
- Division of Pediatric Immunology, Department of Pediatrics, İhsan Doğramacı Children's Hospital, Hacettepe University Medical Faculty, Ankara, Turkey
- Section of Pediatric Immunology, Institutes of Child Health, Health Science Institute, Hacettepe University, Ankara, Turkey
| | - Cindy S Ma
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia
| | - Ahmet Özen
- Department of Pediatric Allergy and Immunology, Marmara University Faculty of Medicine, İstanbul, Turkey
- Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkey
- The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Polina Stepensky
- Department of Bone Marrow Transplantation, Hadassah Medical Center and Faculty of Medicine Hebrew University of Jerusalem, Jerusalem, Israel
| | - Ilhan Tezcan
- Division of Pediatric Immunology, Department of Pediatrics, İhsan Doğramacı Children's Hospital, Hacettepe University Medical Faculty, Ankara, Turkey
- Section of Pediatric Immunology, Institutes of Child Health, Health Science Institute, Hacettepe University, Ankara, Turkey
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria.
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria.
- Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.
- St. Anna Children's Hospital, Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria.
| | - Stuart G Tangye
- Garvan Institute of Medical Research, 384 Victoria Street, Darlinghurst, NSW, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine, UNSW, Sydney, Australia.
| |
Collapse
|
13
|
Ling E, Broides A, Ling G, Shubinsky G, Hadad N, Nahum A, Simon AJ, Lev A, Somech R. A novel zeta-associated protein 70 homozygous mutation causing combined immunodeficiency presenting as neonatal autoimmune hemolytic anemia. Immunol Res 2021; 69:100-106. [PMID: 33484432 DOI: 10.1007/s12026-021-09172-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Accepted: 01/07/2021] [Indexed: 10/22/2022]
Abstract
Biallelic mutations in the zeta-associated protein 70 (ZAP70) gene cause combined immunodeficiency (CID). Neonatal screening for severe CID in Israel is implemented since 2015. We report on clinical, flow cytometry, and genetic data of an unusual ZAP70 deficiency patient. A 10-week-old Bedouin female presented with severe autoimmune hemolytic anemia. Cytomegalovirus (CMV) negative packed cell therapy was given without improvement; indexes of hemolysis worsened. At this time, thrombocytopenia was noted. The patient was treated with single dose of 1 g/kg intravenous immunoglobulin with rapid resolution of hemolysis. Serum immunoglobulin concentrations were normal; flow cytometry revealed severe CD8 lymphocytopenia. Lymphocyte proliferation test demonstrated reduced response to concanavalin A and phytohemagglutinin. Gated T cells were negative for intracellular ZAP70. A genetic analysis revealed a missense homozygous c.1388C > T (p.A463V) mutation, confirming the diagnosis of ZAP70 deficiency. She later on developed urinary tract infection due to ESBL producing E. coli treated with amikacin and severe CMV infection that partially responded to ganciclovir therapy and at 7 months of age, she successfully underwent allogeneic hematopoietic stem cell transplantation. Neonatal screening by T cell receptor excision circles (TRECs) for SCID was normal, yet very low TRECs were recorded at the time of CID diagnosis. Normal neonatal screening for SCID does not rule out the diagnosis of CID due to ZAP70 deficiency. This type of CID can present with autoimmunity as the sole initial manifestation of the disease.
Collapse
Affiliation(s)
- Eduard Ling
- Department of Pediatrics B and Pediatric Rheumatology Clinic, Soroka University Medical Center, Rager Avenue, Beer Sheva, Israel. .,Soroka University Medical Center, Beer Sheva, Israel. .,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Arnon Broides
- Soroka University Medical Center, Beer Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Pediatric Immunology Clinic, Beer Sheva, Israel
| | - Galina Ling
- Soroka University Medical Center, Beer Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Pediatric Gastroenterology and Nutrition Unit and Pediatric Ambulatory Service, Tel Aviv, Israel
| | - George Shubinsky
- Soroka University Medical Center, Beer Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Flow Cytometry Unit, Beer Sheva, Israel
| | - Nurit Hadad
- Soroka University Medical Center, Beer Sheva, Israel.,Infectious Disease Laboratory, Tel Aviv, Israel
| | - Amit Nahum
- Soroka University Medical Center, Beer Sheva, Israel.,Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Pediatric Immunology Clinic, Beer Sheva, Israel
| | - Amos J Simon
- Pediatric Immunology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Atar Lev
- Pediatric Immunology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Raz Somech
- Pediatric Immunology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
14
|
NaserEddin A, Dinur-Schejter Y, Shadur B, Zaidman I, Even-Or E, Averbuch D, Shamriz O, Tal Y, Shaag A, Warnatz K, Elpeleg O, Stepensky P. Bacillus Calmette-Guerin (BCG) Vaccine-associated Complications in Immunodeficient Patients Following Stem Cell Transplantation. J Clin Immunol 2020; 41:147-162. [PMID: 33111199 PMCID: PMC7591244 DOI: 10.1007/s10875-020-00892-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 10/07/2020] [Indexed: 11/28/2022]
Abstract
PURPOSE Bacillus Calmette-Guerin (BCG) is a live attenuated vaccine with the potential of causing severe iatrogenic complications in patients with primary immunodeficiency diseases (PID) before and after hematopoietic stem cell transplantation (HSCT). We aim to investigate risk factors of post-HSCT BCG-related complications in PID patients. METHODS A retrospective analysis of pediatric PID patients who had received the BCG vaccine and underwent HSCT at Hadassah-Hebrew University Medical Center, between 2007 and 2019. RESULTS We found 15/36 (41.67%) patients who developed post-HSCT BCG-related complications. The most significant risk factor for developing BCG-related complications was T cell deficiency (47.6% of the non-complicated vs 83.3% of the BCGitis and 100% of the BCGosis groups had T cell lymphopenia, p = 0.013). None of the chronic granulomatous patients developed BCG-related manifestation post-transplant. Among T cell-deficient patients, lower NK (127 vs 698 cells/μl, p = 0.04) cell counts and NK-SCID were risk factors for ongoing post-HSCT BCGosis, as was pretransplant disseminated BCGosis (33.3% of patients with BCGosis vs none of the non-BCGosis patients, p = 0.04). Immune reconstitution inflammatory syndrome (IRIS) was observed in 3/5 patients with Omenn syndrome. Prophylactic antimycobacterial treatment was not proven effective. CONCLUSION BCG vaccination can cause significant morbidity and mortality in the post-transplant T cell-deficient patient, especially in the presence of pre-transplant disease. Taking a detailed medical history prior to administering, the BCG vaccine is crucial for prevention of this complication.
Collapse
Affiliation(s)
- Adeeb NaserEddin
- Bone Marrow Transplantation Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel. .,Hadassah Medical Organization, POB 12000, Kiryat Hadassah, 91120, Jerusalem, Israel.
| | - Yael Dinur-Schejter
- Bone Marrow Transplantation Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel.,Allergy & Clinical Immunology Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Bella Shadur
- Garvan Institute of Medical Research, Sydney, Australia.,Graduate Research School, University of New South Wales, Kensington, Australia
| | - Irina Zaidman
- Bone Marrow Transplantation Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Ehud Even-Or
- Bone Marrow Transplantation Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Diana Averbuch
- Pediatric Infectious Diseases Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Oded Shamriz
- Allergy & Clinical Immunology Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Yuval Tal
- Allergy & Clinical Immunology Unit, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Avraham Shaag
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Klaus Warnatz
- Department for Rheumatology and Clinical Immunology and Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, Freiburg im Breisgau, Germany.,Center for Chronic Immunodeficiency (CCI), Medical Center, University of Freiburg, Faculty of Medicine, Freiburg im Breisgau, Germany
| | - Orly Elpeleg
- Monique and Jacques Roboh Department of Genetic Research, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Polina Stepensky
- Bone Marrow Transplantation Department, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| |
Collapse
|
15
|
Chitty-Lopez M, Westermann-Clark E, Dawson I, Ujhazi B, Csomos K, Dobbs K, Le K, Yamazaki Y, Sadighi Akha AA, Chellapandian D, Oshrine B, Notarangelo LD, Sunkersett G, Leiding JW, Walter JE. Asymptomatic Infant With Atypical SCID and Novel Hypomorphic RAG Variant Identified by Newborn Screening: A Diagnostic and Treatment Dilemma. Front Immunol 2020; 11:1954. [PMID: 33117328 PMCID: PMC7552884 DOI: 10.3389/fimmu.2020.01954] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 07/20/2020] [Indexed: 12/11/2022] Open
Abstract
The T-cell receptor excision circle (TREC) assay detects T-cell lymphopenia (TCL) in newborns and is especially important to identify severe combined immunodeficiency (SCID). A spectrum of SCID variants and non-SCID conditions that present with TCL are being discovered with increasing frequency by newborn screening (NBS). Recombination-activating gene (RAG) deficiency is one the most common causes of classical and atypical SCID and other conditions with immune dysregulation. We present the case of an asymptomatic male with undetectable TRECs on NBS at 1 week of age. The asymptomatic newborn was found to have severe TCL, but normal B cell quantities and lymphocyte proliferation upon mitogen stimulation. Next generation sequencing revealed compound heterozygous hypomorphic RAG variants, one of which was novel. The moderately decreased recombinase activity of the RAG variants (16 and 40%) resulted in abnormal T and B-cell receptor repertoires, decreased fraction of CD3+ TCRVα7.2+ T cells and an immune phenotype consistent with the RAG hypomorphic variants. The patient underwent successful treatment with hematopoietic stem cell transplantation (HSCT) at 5 months of age. This case illustrates how after identification of a novel RAG variant, in vitro studies are important to confirm the pathogenicity of the variant. This confirmation allows the clinician to expedite definitive treatment with HSCT in an asymptomatic phase, mitigating the risk of serious infectious and non-infectious complications.
Collapse
Affiliation(s)
- Maria Chitty-Lopez
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Emma Westermann-Clark
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Irina Dawson
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Boglarka Ujhazi
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Krisztian Csomos
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Khuong Le
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Yasuhiro Yamazaki
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Amir A Sadighi Akha
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, United States
| | - Deepak Chellapandian
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Ben Oshrine
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Gauri Sunkersett
- Cancer and Blood Disorders Institute, Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Jennifer W Leiding
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Jolan E Walter
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States.,Division of Pediatric Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA, United States
| |
Collapse
|
16
|
Al-Rasheed B, Alazami AM, Al-Mousa H. Phenoidentical HLA-Related Hematopoietic Stem Cell Transplant Without Conditioning to Reconstitute a Patient with a Putative Loss-of-Function CARD11 Mutation. J Clin Immunol 2020; 40:1163-1165. [PMID: 32815076 DOI: 10.1007/s10875-020-00846-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 08/13/2020] [Indexed: 02/07/2023]
Affiliation(s)
- Bashayer Al-Rasheed
- Pediatric Allergy & Clinical Immunology, Department of Pediatrics, King Faisal Specialist Hospital and Research Centre, MBC 58, P.O.Box 3354, Riyadh, 11211, Saudi Arabia
| | - Anas M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hamoud Al-Mousa
- Pediatric Allergy & Clinical Immunology, Department of Pediatrics, King Faisal Specialist Hospital and Research Centre, MBC 58, P.O.Box 3354, Riyadh, 11211, Saudi Arabia. .,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| |
Collapse
|
17
|
Shinwari K, Bolkov M, Tuzankina IA, Chereshnev VA. Newborn Screening through TREC, TREC/KREC System for Primary Immunodeficiency with limitation of TREC/KREC. Comprehensive Review. Antiinflamm Antiallergy Agents Med Chem 2020; 20:132-149. [PMID: 32748762 DOI: 10.2174/1871523019999200730171600] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Revised: 06/11/2020] [Accepted: 06/21/2020] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Newborn screening (NBS) by quantifying T cell receptor excision circles (TRECs) and Kappa receptor excision circles in neonatal dried blood spots (DBS) enables early diagnosis of different types of primary immune deficiencies. Global newborn screening for PID, using an assay to detect T-cell receptor excision circles (TREC) in dried blood spots (DBS), is now being performed in all states in the United States. In this review, we discuss the development and outcomes of TREC, TREC/KREC combines screening, and continued challenges to implementation. OBJECTIVE To review the diagnostic performance of published articles for TREC and TREC/ KREC based NBS for PID and its different types. METHODS Different research resources were used to get an approach for the published data of TREС and KREC based NBS for PID like PubMed, Scopus, Google Scholar, Research gate EMBASE. We extracted TREC and KREC screening Publisher with years of publication, content and cut-off values, and a number of retests, repeat DBS, and referrals from the different published pilot, pilot cohort, Case series, and cohort studies. RESULTS We included the results of TREC, combined TREC/KREC system based NBS screening from different research articles, and divided these results between the Pilot studies, case series, and cohort. For each of these studies, different parameter data are excluded from different articles. Thirteen studies were included, re-confirming 89 known SCID cases in case series and reporting 53 new SCID cases in 3.15 million newborns. Individual TREC contents in all SCID patients were <25 TRECs/μl (except in those evaluated with the New York State assay). CONCLUSION TREC and KREC sensitivity for typical SCID and other types of PID was 100 %. It shows its importance and anticipating the significance of implementation in different undeveloped and developed countries in the NBS program in upcoming years. Data adapting the screening algorithm for pre-term/ill infants reduce the amount of false-positive test results.
Collapse
Affiliation(s)
- Khyber Shinwari
- Department of Immunochemistry, Institute of Chemical Engineering, Ural Federal University, Yekaterinburg, Russian Federation
| | - Mikhail Bolkov
- Department of Immunochemistry, Institute of Chemical Engineering, Ural Federal University, Yekaterinburg, Russian Federation
| | - Irina A Tuzankina
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russian Federation
| | - Valery A Chereshnev
- Institute of Immunology and Physiology of the Ural Branch of the Russian Academy of Sciences, Yekaterinburg, Russian Federation
| |
Collapse
|
18
|
Dasouki M, Jabr A, AlDakheel G, Elbadaoui F, Alazami AM, Al-Saud B, Arnaout R, Aldhekri H, Alotaibi I, Al-Mousa H, Hawwari A. TREC and KREC profiling as a representative of thymus and bone marrow output in patients with various inborn errors of immunity. Clin Exp Immunol 2020; 202:60-71. [PMID: 32691468 DOI: 10.1111/cei.13484] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2020] [Revised: 06/11/2020] [Accepted: 06/14/2020] [Indexed: 12/13/2022] Open
Abstract
Primary immune deficiency (PID) disorders are clinically and molecularly heterogeneous diseases. T cell receptor excision circles (TRECs) and κ (kappa)-deleting excision circles (KRECs) are markers of T and B cell development, respectively. They are useful tools to assess T and B cell function and immune reconstitution and have been used for newborn screening for severe combined immunodeficiency disease (SCID) and agammaglobulinemia, respectively. Their profiles in several genetically confirmed PIDs are still lacking. The objective of this study was to determine TREC and KREC genomic profiling among various molecularly confirmed PIDs. We used real-time-quantitative polymerase chain reaction (RT-qPCR)-based triplex analysis of TRECs, KRECs and β-actin (ACTB) in whole blood genomic DNA isolated from 108 patients with molecularly confirmed PIDs. All agammaglobulinemia patients had low KREC counts. All SCIDs and Omenn syndrome patients secondary to mutations in RAG1, RAG2, DCLRE1C and NHEJ1 had low TREC and KREC counts. JAK3-deficient patients had normal KREC and the TREC count was influenced by the type of mutation. Early-onset ADA patients had low TREC and KREC counts. Four patients with zeta-chain-associated protein kinase 70 (ZAP70) had low TREC. All purine nucleoside phosphorylase (PNP) patients had low TREC. Combined immunodeficiency (CID) patients secondary to AK2, PTPRC, CD247, DCLREC1 and STAT1 had normal TREC and KREC counts. Most patients with ataxia-telangiectasia (AT) patients had low TREC and KREC, while most DOCK8-deficient patients had low TRECs only. Two of five patients with Wiskott-Aldrich syndrome (WAS) had low TREC counts as well as one patient each with bare lymphocyte syndrome (BLS) and chronic granulomatous disease. All patients with Griscelli disease, Chediak-Higashi syndrome, hyper-immunoglobulin (Ig)M syndrome and IFNGR2 had normal TREC and KREC counts. These data suggest that, in addition to classical SCID and agammaglobulinemia, TREC/KREC assay may identify ZAP70 patients and secondary target PIDs, including dedicator of cytokinesis 8 (DOCK8) deficiency, AT and some individuals with WAS and BLS.
Collapse
Affiliation(s)
- M Dasouki
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - A Jabr
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - G AlDakheel
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - F Elbadaoui
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - A M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - B Al-Saud
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - R Arnaout
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - H Aldhekri
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - I Alotaibi
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - H Al-Mousa
- Department of Pediatrics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia
| | - A Hawwari
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdulaziz University for Health Sciences, King Abdulaziz Medical City Hospital, Ministry of National Guard Health Affairs, Al-Ahsa, Saudi Arabia
| |
Collapse
|
19
|
Sharapova SO, Skomska-Pawliszak M, Rodina YA, Wolska-Kuśnierz B, Dabrowska-Leonik N, Mikołuć B, Pashchenko OE, Pasic S, Freiberger T, Milota T, Formánková R, Szaflarska A, Siedlar M, Avčin T, Markelj G, Ciznar P, Kalwak K, Kołtan S, Jackowska T, Drabko K, Gagro A, Pac M, Naumova E, Kandilarova S, Babol-Pokora K, Varabyou DS, Barendregt BH, Raykina EV, Varlamova TV, Pavlova AV, Grombirikova H, Debeljak M, Mersiyanova IV, Bondarenko AV, Chernyshova LI, Kostyuchenko LV, Guseva MN, Rascon J, Muleviciene A, Preiksaitiene E, Geier CB, Leiss-Piller A, Yamazaki Y, Kawai T, Walter JE, Kondratenko IV, Šedivá A, van der Burg M, Kuzmenko NB, Notarangelo LD, Bernatowska E, Aleinikova OV. The Clinical and Genetic Spectrum of 82 Patients With RAG Deficiency Including a c.256_257delAA Founder Variant in Slavic Countries. Front Immunol 2020; 11:900. [PMID: 32655540 PMCID: PMC7325958 DOI: 10.3389/fimmu.2020.00900] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Accepted: 04/20/2020] [Indexed: 11/13/2022] Open
Abstract
Background: Variants in recombination-activating genes (RAG) are common genetic causes of autosomal recessive forms of combined immunodeficiencies (CID) ranging from severe combined immunodeficiency (SCID), Omenn syndrome (OS), leaky SCID, and CID with granulomas and/or autoimmunity (CID-G/AI), and even milder presentation with antibody deficiency. Objective: We aim to estimate the incidence, clinical presentation, genetic variability, and treatment outcome with geographic distribution of patients with the RAG defects in populations inhabiting South, West, and East Slavic countries. Methods: Demographic, clinical, and laboratory data were collected from RAG-deficient patients of Slavic origin via chart review, retrospectively. Recombinase activity was determined in vitro by flow cytometry-based assay. Results: Based on the clinical and immunologic phenotype, our cohort of 82 patients from 68 families represented a wide spectrum of RAG deficiencies, including SCID (n = 20), OS (n = 37), and LS/CID (n = 25) phenotypes. Sixty-seven (81.7%) patients carried RAG1 and 15 patients (18.3%) carried RAG2 biallelic variants. We estimate that the minimal annual incidence of RAG deficiency in Slavic countries varies between 1 in 180,000 and 1 in 300,000 live births, and it may vary secondary to health care disparities in these regions. In our cohort, 70% (n = 47) of patients with RAG1 variants carried p.K86Vfs*33 (c.256_257delAA) allele, either in homozygous (n = 18, 27%) or in compound heterozygous (n = 29, 43%) form. The majority (77%) of patients with homozygous RAG1 p.K86Vfs*33 variant originated from Vistula watershed area in Central and Eastern Poland, and compound heterozygote cases were distributed among all Slavic countries except Bulgaria. Clinical and immunological presentation of homozygous RAG1 p.K86Vfs*33 cases was highly diverse (SCID, OS, and AS/CID) suggestive of strong influence of additional genetic and/or epigenetic factors in shaping the final phenotype. Conclusion: We propose that RAG1 p.K86Vfs*33 is a founder variant originating from the Vistula watershed region in Poland, which may explain a high proportion of homozygous cases from Central and Eastern Poland and the presence of the variant in all Slavs. Our studies in this cohort of RAG1 founder variants confirm that clinical and immunological phenotypes only partially depend on the underlying genetic defect. As access to HSCT is improving among RAG-deficient patients in Eastern Europe, we anticipate improvements in survival.
Collapse
Affiliation(s)
- Svetlana O. Sharapova
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk Region, Belarus
| | | | - Yulia A. Rodina
- Department of Immunology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | | | | | - Bozena Mikołuć
- Department of Pediatrics, Rheumatology, Immunology and Metabolic Bone Diseases, Medical University of Bialystok, Bialystok, Poland
| | - Olga E. Pashchenko
- Immunology Department, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Srdjan Pasic
- Pediatric Immunology, Medical Faculty, Mother and Child Health Institute, University of Belgrade, Belgrade, Serbia
| | - Tomáš Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Tomáš Milota
- Department of Immunology, University Hospital Motol, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Renata Formánková
- Department of Pediatric Hematology and Oncology, University Hospital Motol, Prague, Czechia
- Faculty of Medicine, Charles University, Prague, Czechia
| | - Anna Szaflarska
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
| | - Maciej Siedlar
- Department of Clinical Immunology, Institute of Pediatrics, Jagiellonian University Medical College, Krakow, Poland
- Department of Clinical Immunology, University Children's Hospital, Krakow, Poland
| | - Tadej Avčin
- University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Gašper Markelj
- University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Peter Ciznar
- Pediatric Department, Faculty of Medicine, Comenius University, Bratislava, Slovakia
| | - Krzysztof Kalwak
- Department of Pediatric Hematology/Oncology and BMT, Wroclaw Medical University, Wroclaw, Poland
| | - Sylwia Kołtan
- Department of Pediatrics, Hematology and Oncology Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland
- Nicolaus Copernicus University in Torun, Torun, Poland
| | - Teresa Jackowska
- Department of Pediatrics, Medical Center of Postgraduate Education, Warsaw, Poland
| | - Katarzyna Drabko
- Department of Pediatric Hematology, Oncology and Transplantology, Medical University of Lublin, Lublin, Poland
| | - Alenka Gagro
- Department of Pediatrics, School of Medicine, Zagreb Children's Hospital, University of Zagreb, Zagreb, Croatia
- Faculty of Medicine, Josip Juraj Strossmayer University of Osijek, Osijek, Croatia
| | - Małgorzata Pac
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - Elissaveta Naumova
- Department of Clinical Immunology, University Hospital Alexandrovska, Sofia, Bulgaria
| | - Snezhina Kandilarova
- Department of Clinical Immunology, University Hospital Alexandrovska, Sofia, Bulgaria
| | - Katarzyna Babol-Pokora
- Department of Pediatrics, Oncology and Hematology, Medical University of Lodz, Lodz, Poland
| | - Dzmitry S. Varabyou
- Department of Geographical Ecology, Belarusian State University, Minsk, Belarus
| | - Barbara H. Barendregt
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Elena V. Raykina
- Laboratory of Molecular Biology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Tatiana V. Varlamova
- Laboratory of Molecular Biology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Anna V. Pavlova
- Laboratory of Molecular Biology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Hana Grombirikova
- Centre for Cardiovascular Surgery and Transplantation, Brno, Czechia
- Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Maruša Debeljak
- University Children's Hospital, University Medical Centre Ljubljana, Ljubljana, Slovenia
- Faculty of Medicine, University of Ljubljana, Ljubljana, Slovenia
| | - Irina V. Mersiyanova
- Laboratory of Molecular Biology, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Anastasiia V. Bondarenko
- Department of Pediatric Infectious Diseases and Pediatric Immunology, Shupyk National Medical Academy for Postgraduate Education, Kiev, Ukraine
| | - Liudmyla I. Chernyshova
- Department of Pediatric Infectious Diseases and Pediatric Immunology, Shupyk National Medical Academy for Postgraduate Education, Kiev, Ukraine
| | - Larysa V. Kostyuchenko
- Pediatric Department, West-Ukrainian Specialized Children's Medical Center, Lviv, Ukraine
| | - Marina N. Guseva
- Consulting Center of Pediatric Medical Academy, St. Petersburg, Russia
| | - Jelena Rascon
- Center for Pediatric Oncology and Hematology, Vilnius University, Vilnius, Lithuania
| | - Audrone Muleviciene
- Center for Pediatric Oncology and Hematology, Vilnius University, Vilnius, Lithuania
| | - Egle Preiksaitiene
- Hematology, Oncology and Transfusion Medicine Center, Vilnius University, Vilnius, Lithuania
| | | | | | - Yasuhiro Yamazaki
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Tomoki Kawai
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Jolan E. Walter
- University of South Florida at Johns Hopkins All Children's Hospital, Saint Petersburg, FL, United States
- Massachusetts General Hospital for Children, Boston, MA, United States
| | - Irina V. Kondratenko
- Department of Clinical Immunology, Russian Clinical Children's Hospital by Pirogov Russian National Research Medical University, Moscow, Russia
| | - Anna Šedivá
- Department of Immunology, University Hospital Motol, Prague, Czechia
- Second Faculty of Medicine, Charles University, Prague, Czechia
| | - Mirjam van der Burg
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
- Department of Pediatric, Laboratory for Pediatric Immunology, Willem Alexander Children's Hospital, LUMC, Leiden, Netherlands
| | - Natalia B. Kuzmenko
- Department of Epidemiology and Monitoring of Primary Immunodeficiencies, Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russia
| | - Luigi D. Notarangelo
- Laboratory of Clinical Immunology and Microbiology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, United States
| | - Ewa Bernatowska
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - Olga V. Aleinikova
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk Region, Belarus
| |
Collapse
|
20
|
Novel heterozygous PIK3CD mutation presenting with only laboratory markers of combined immunodeficiency. LYMPHOSIGN JOURNAL 2020. [DOI: 10.14785/lymphosign-2020-0003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Introduction: Phosphatidylinositol-4,5-Bisphosphate 3-Kinase Catalytic Subunit Delta (PIK3CD) is one part of a heterodimer forming the enzyme phosphoinositide 3-kinase (PI3K), found primarily in leukocytes. PIK3CD generates phosphatidyl-inositol 3,4,5-trisphosphate (PIP3), and is involved in cell growth, survival, proliferation, motility, and morphology. An increasing number of patients have been described with heterozygous PIK3CD gain-of-function (GOF) mutations, leading to combined immunodeficiency with both B- and T-cell dysfunction. Patients suffer recurrent respiratory infections, often associated with bronchiectasis and ear and sinus damage, as well as severe recurrent or persistent infections by herpesviruses, including EBV-induced lymphoproliferation. Aim: To present the clinical phenotypic variability of a novel PI3KCD mutation within a family. Methods: Patient information was collected prospectively and retrospectively from medical records. Comprehensive immune work up, genetic, and signaling evaluation was performed. Results: We describe here 2 patients, daughter and mother, with heterozygous PIK3CD mutation identified by whole exome sequencing and Sanger confirmation. The child was screen-positive by newborn screening for severe combined immunodeficiency (SCID). Cellular assays revealed an increase in the baseline phosphorylation of T cells in the patient. Furthermore, both patients had hyper-activation of the catalytic domain, resulting in increased phosphorylation of AKT upon activation. Discussion: GOF mutations affecting the PIK3CD gene are associated with an increased risk for lymphoproliferation leading to Activated PIK3-delta syndrome (APDS). The clinical course of APDS is highly variable, ranging from combined immunodeficiency with recurrent infections, autoimmune complications, and requiring stem cell transplantation, through isolated antibody deficiency, to asymptomatic adults. Our patient is the first to be identified by newborn screening for SCID. Surprisingly, the clinical course has so far been unremarkable, as well, the mother appears to be completely asymptomatic. Nevertheless, the persistent lymphopenia indicates PIK3CD dysfunction. Because of the wide gap between laboratory findings and clinical manifestations, this kindred poses both a diagnostic as well treatment challenge. Statement of novelty: We report here a novel PIK3CD mutation diagnosed due to abnormal newborn screen for SCID.
Collapse
|
21
|
Kalina T, Bakardjieva M, Blom M, Perez-Andres M, Barendregt B, Kanderová V, Bonroy C, Philippé J, Blanco E, Pico-Knijnenburg I, Paping JHMP, Wolska-Kuśnierz B, Pac M, Tkazcyk J, Haerynck F, Akar HH, Formánková R, Freiberger T, Svatoň M, Šedivá A, Arriba-Méndez S, Orfao A, van Dongen JJM, van der Burg M. EuroFlow Standardized Approach to Diagnostic Immunopheneotyping of Severe PID in Newborns and Young Children. Front Immunol 2020; 11:371. [PMID: 32265901 PMCID: PMC7096355 DOI: 10.3389/fimmu.2020.00371] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2019] [Accepted: 02/17/2020] [Indexed: 12/13/2022] Open
Abstract
The EuroFlow PID consortium developed a set of flow cytometry tests for evaluation of patients with suspicion of primary immunodeficiency (PID). In this technical report we evaluate the performance of the SCID-RTE tube that explores the presence of recent thymic emigrants (RTE) together with T-cell activation status and maturation stages and discuss its applicability in the context of the broader EuroFlow PID flow cytometry testing algorithm for diagnostic orientation of PID of the lymphoid system. We have analyzed peripheral blood cells of 26 patients diagnosed between birth and 2 years of age with a genetically defined primary immunodeficiency disorder: 15 severe combined immunodeficiency (SCID) patients had disease-causing mutations in RAG1 or RAG2 (n = 4, two of them presented with Omenn syndrome), IL2RG (n = 4, one of them with confirmed maternal engraftment), NHEJ1 (n = 1), CD3E (n = 1), ADA (n = 1), JAK3 (n = 3, two of them with maternal engraftment) and DCLRE1C (n = 1) and 11 other PID patients had diverse molecular defects [ZAP70 (n = 1), WAS (n = 2), PNP (n = 1), FOXP3 (n = 1), del22q11.2 (DiGeorge n = 4), CDC42 (n = 1) and FAS (n = 1)]. In addition, 44 healthy controls in the same age group were analyzed using the SCID-RTE tube in four EuroFlow laboratories using a standardized 8-color approach. RTE were defined as CD62L+CD45RO-HLA-DR-CD31+ and the activation status was assessed by the expression of HLA-DR+. Naïve CD8+ T-lymphocytes and naïve CD4+ T-lymphocytes were defined as CD62L+CD45RO-HLA-DR-. With the SCID-RTE tube, we identified patients with PID by low levels or absence of RTE in comparison to controls as well as low levels of naïve CD4+ and naïve CD8+ lymphocytes. These parameters yielded 100% sensitivity for SCID. All SCID patients had absence of RTE, including the patients with confirmed maternal engraftment or oligoclonally expanded T-cells characteristic for Omenn syndrome. Another dominant finding was the increased numbers of activated CD4+HLA-DR+ and CD8+HLA-DR+ lymphocytes. Therefore, the EuroFlow SCID-RTE tube together with the previously published PIDOT tube form a sensitive and complete cytometric diagnostic test suitable for patients suspected of severe PID (SCID or CID) as well as for children identified via newborn screening programs for SCID with low or absent T-cell receptor excision circles (TRECs).
Collapse
Affiliation(s)
- Tomas Kalina
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Marina Bakardjieva
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Maartje Blom
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Martin Perez-Andres
- Department of Medicine-Serv. Cytometry, Cancer Research Center (IBMCC-CSIC/USAL), University of Salamanca, Salamanca, Spain
| | - Barbara Barendregt
- Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Veronika Kanderová
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Carolien Bonroy
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Jan Philippé
- Department of Diagnostic Sciences, Ghent University, Ghent, Belgium.,Department of Laboratory Medicine, Ghent University Hospital, Ghent, Belgium
| | - Elena Blanco
- Department of Medicine-Serv. Cytometry, Cancer Research Center (IBMCC-CSIC/USAL), University of Salamanca, Salamanca, Spain
| | - Ingrid Pico-Knijnenburg
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| | - Jitse H M P Paping
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands
| | | | - Malgorzata Pac
- Department of Immunology, Children's Memorial Health Institute, Warsaw, Poland
| | - Jakub Tkazcyk
- Department of Pediatrics, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Filomeen Haerynck
- PID Research Lab, Department of Pediatric Pulmonology and Immunology, Ghent University Hospital, Ghent, Belgium
| | - Himmet Haluk Akar
- Department of Pediatric Immunology and Allergy, Kanuni Sultan Süleyman Training and Research Hospital, Istanbul Health Sciences University, Istanbul, Turkey
| | - Renata Formánková
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Tomáš Freiberger
- Centre for Cardiovascular Surgery and Transplantation, Brno, Czechia.,Medical Faculty, Masaryk University, Brno, Czechia
| | - Michael Svatoň
- Department of Paediatric Haematology and Oncology, Second Faculty of Medicine, Charles University and University Hospital Motol, Prague, Czechia
| | - Anna Šedivá
- Department of Immunology, University Hospital Motol, Prague, Czechia
| | - Sonia Arriba-Méndez
- Servicio de Pediatría, Hospital Universitario de Salamanca, Salamanca, Spain
| | - Alberto Orfao
- Department of Medicine-Serv. Cytometry, Cancer Research Center (IBMCC-CSIC/USAL), University of Salamanca, Salamanca, Spain
| | - Jacques J M van Dongen
- Department of Immunohematology and Blood Transfusion (IHB), Leiden University Medical Center (LUMC), Leiden, Netherlands
| | - Mirjam van der Burg
- Laboratory for Immunology, Department of Pediatrics, Leiden University Medical Center (LUMC), Leiden, Netherlands.,Department of Immunology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, Netherlands
| |
Collapse
|
22
|
Tuovinen EA, Grönholm J, Öhman T, Pöysti S, Toivonen R, Kreutzman A, Heiskanen K, Trotta L, Toiviainen-Salo S, Routes JM, Verbsky J, Mustjoki S, Saarela J, Kere J, Varjosalo M, Hänninen A, Seppänen MRJ. Novel Hemizygous IL2RG p.(Pro58Ser) Mutation Impairs IL-2 Receptor Complex Expression on Lymphocytes Causing X-Linked Combined Immunodeficiency. J Clin Immunol 2020; 40:503-514. [PMID: 32072341 PMCID: PMC7142052 DOI: 10.1007/s10875-020-00745-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 01/06/2020] [Indexed: 11/30/2022]
Abstract
Hypomorphic IL2RG mutations may lead to milder phenotypes than X-SCID, named variably as atypical X-SCID or X-CID. We report an 11-year-old boy with a novel c. 172C>T;p.(Pro58Ser) mutation in IL2RG, presenting with atypical X-SCID phenotype. We also review the growing number of hypomorphic IL2RG mutations causing atypical X-SCID. We studied the patient's clinical phenotype, B, T, NK, and dendritic cell phenotypes, IL2RG and CD25 cell surface expression, and IL-2 target gene expression, STAT tyrosine phosphorylation, PBMC proliferation, and blast formation in response to IL-2 stimulation, as well as protein-protein interactions of the mutated IL2RG by BioID proximity labeling. The patient suffered from recurrent upper and lower respiratory tract infections, bronchiectasis, and reactive arthritis. His total lymphocyte counts have remained normal despite skewed T and B cells subpopulations, with very low numbers of plasmacytoid dendritic cells. Surface expression of IL2RG was reduced on his lymphocytes. This led to impaired STAT tyrosine phosphorylation in response to IL-2 and IL-21, reduced expression of IL-2 target genes in patient CD4+ T cells, and reduced cell proliferation in response to IL-2 stimulation. BioID proximity labeling showed aberrant interactions between mutated IL2RG and ER/Golgi proteins causing mislocalization of the mutated IL2RG to the ER/Golgi interface. In conclusion, IL2RG p.(Pro58Ser) causes X-CID. Failure of IL2RG plasma membrane targeting may lead to atypical X-SCID. We further identified another carrier of this mutation from newborn SCID screening, lost to closer scrutiny.
Collapse
Affiliation(s)
- Elina A Tuovinen
- Folkhälsan Research Center, Helsinki, Finland.,Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Rare Diseases Center and Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Juha Grönholm
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland. .,Rare Diseases Center and Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland.
| | - Tiina Öhman
- Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sakari Pöysti
- Department of Clinical Microbiology and Immunology, Turku University Hospital and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Raine Toivonen
- Department of Clinical Microbiology and Immunology, Turku University Hospital and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Anna Kreutzman
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Kaarina Heiskanen
- Rare Diseases Center and Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - Luca Trotta
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Sanna Toiviainen-Salo
- Department of Pediatric Radiology, HUS Medical Imaging Center, Radiology, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| | - John M Routes
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - James Verbsky
- Department of Pediatrics, Medical College of Wisconsin, Milwaukee, WI, USA
| | - Satu Mustjoki
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Hematology Research Unit Helsinki, Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland.,Department of Clinical Chemistry and Hematology, University of Helsinki, Helsinki, Finland
| | - Janna Saarela
- Institute for Molecular Medicine Finland, HiLIFE, University of Helsinki, Helsinki, Finland.,Department of Medical Genetics, Helsinki Central University Hospital, Helsinki, Finland.,Centre for Molecular Medicine Norway, University of Oslo, Oslo, Norway
| | - Juha Kere
- Folkhälsan Research Center, Helsinki, Finland.,Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden.,Stem Cells and Metabolism Research Program, University of Helsinki, Helsinki, Finland
| | - Markku Varjosalo
- Systems Biology Research Group and Proteomics Unit, Institute of Biotechnology, HiLIFE, University of Helsinki, Helsinki, Finland
| | - Arno Hänninen
- Department of Clinical Microbiology and Immunology, Turku University Hospital and Institute of Biomedicine, University of Turku, Turku, Finland
| | - Mikko R J Seppänen
- Translational Immunology Research Program, University of Helsinki, Helsinki, Finland.,Rare Diseases Center and Pediatric Research Center, New Children's Hospital, University of Helsinki and HUS Helsinki University Hospital, Helsinki, Finland
| |
Collapse
|
23
|
Al-Saud B, Alajlan H, Sabar H, Anwar S, Alruwaili H, Al-Hussain T, Alamri N, Alazami AM. STK4 Deficiency in a Patient with Immune Complex Glomerulonephritis, Salt-Losing Tubulopathy, and Castleman's-Like Disease. J Clin Immunol 2019; 39:823-826. [PMID: 31444685 DOI: 10.1007/s10875-019-00682-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Accepted: 08/16/2019] [Indexed: 01/08/2023]
Affiliation(s)
- Bandar Al-Saud
- Section of Pediatric Allergy/Immunology, Department of Pediatrics, King Faisal Specialist Hospital & Research Center, MBC-58, PO Box 3354, Riyadh, 11211, Saudi Arabia. .,College of Medicine, Alfaisal University, Riyadh, Saudi Arabia.
| | - Huda Alajlan
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, MBC-03, PO Box 3354, Riyadh, 11211, Saudi Arabia
| | - Haneen Sabar
- Section of Pediatric Allergy/Immunology, Department of Pediatrics, King Faisal Specialist Hospital & Research Center, MBC-58, PO Box 3354, Riyadh, 11211, Saudi Arabia
| | - Siddiq Anwar
- Section of Nephrology, Department of Medicine, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Hibah Alruwaili
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, MBC-03, PO Box 3354, Riyadh, 11211, Saudi Arabia
| | - Turki Al-Hussain
- Section of Anatomic Pathology, Department of Pathology and Laboratory Medicine, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Nada Alamri
- Section of Nephrology, Department of Medicine, King Faisal Specialist Hospital & Research Center, Riyadh, Saudi Arabia
| | - Anas M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, MBC-03, PO Box 3354, Riyadh, 11211, Saudi Arabia. .,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia.
| |
Collapse
|
24
|
Suresh S, Dadi H, Reid B, Vong L, Bulman DE, Roifman CM. Time-dependent decline of T-cell receptor excision circle levels in ZAP-70 deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2019; 8:806-808.e2. [PMID: 31449923 DOI: 10.1016/j.jaip.2019.08.018] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/08/2019] [Accepted: 08/10/2019] [Indexed: 10/26/2022]
Affiliation(s)
- Sneha Suresh
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada; Division of IHOPE, Department of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Harjit Dadi
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada; The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON, Canada
| | - Brenda Reid
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada
| | - Linda Vong
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada; The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON, Canada
| | - Dennis E Bulman
- CHEO Research Institute and Newborn Screening Ontario, Department of Pediatrics, University of Ottawa, Ottawa, ON, Canada
| | - Chaim M Roifman
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children and the University of Toronto, Toronto, ON, Canada; The Canadian Centre for Primary Immunodeficiency and The Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, The Hospital for Sick Children, Toronto, ON, Canada.
| |
Collapse
|
25
|
Marcus N, Stauber T, Lev A, Simon AJ, Stein J, Broides A, Somekh I, Almashanu S, Somech R. MHC II deficient infant identified by newborn screening program for SCID. Immunol Res 2019; 66:537-542. [PMID: 30084052 DOI: 10.1007/s12026-018-9019-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Newborn screening (NBS) programs for severe combined immunodeficiency (SCID), using the TREC-based assay, have enabled early diagnosis, prompt treatment, and eventually changed the natural history of affected infants. Nevertheless, it was believed that some affected infants with residual T cell, such as patients with MHC II deficiency, will be misdiagnosed by this assay. A full immune workup and genetic analysis using direct Sanger sequencing and whole exome sequencing have been performed to a patient that was identified by the Israeli NBS program for SCID. The patient was found to have severe CD4 lymphopenia with an inverted CD4/CD8 ratio, low TREC levels in peripheral blood, abnormal response to mitogen stimulation, and a skewed T cell receptor repertoire. HLA-DR expression on peripheral blood lymphocytes was undetectable suggesting a diagnosis of MHC II deficiency. Direct sequencing of the RFX5 gene revealed a stop codon change (p. R239X, c. C715T), which could cause the patient's immune phenotype. His parents were found to be heterozygote carriers for the mutation. Whole exome sequencing could not identify other potential mutations to explain his immunodeficiency. The patient underwent successful conditioned hematopoietic stem cell transplantation from healthy matched unrelated donor and is currently well and alive with full chimerism. Infants with MHC class II deficiency can potentially be identified by the TREC-based assay NBS for SCID. Therefore, MHC II molecules (e.g., HLA-DR) measurement should be part of the confirmatory immune-phenotyping for patients with positive screening results. This will make the diagnosis of such patients straightforward.
Collapse
Affiliation(s)
- Nufar Marcus
- Allergy and Immunology Unit, Felsenstein Medical Research Center, Kipper Institute of Immunology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Tali Stauber
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel
| | - Atar Lev
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel
| | - Amos J Simon
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel
| | - Jerry Stein
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Department for Hemato-Oncology, Schneider Children's Medical Center of Israel, Petach Tikva, Israel
| | - Arnon Broides
- Pediatric Immunology Clinic, Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ido Somekh
- Dr. von Hauner Children's Hospital, University Hospital, LMU Munich, Munich, Germany
| | - Shlomo Almashanu
- The National Center for Newborn Screening, Ministry of Health, 52621, Tel HaShomer, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.
- Pediatric Department A, Pediatric Immunology Service, Jeffrey Modell Foundation Center, 52621, Tel Hashomer, Israel.
- Edmond and Lily Safra Children's Hospital, Sheba Medical Center, 52621, Tel Hashomer, Israel.
- The National Lab for Confirming Primary Immunodeficiency in Newborn Screening Center for Newborn Screening, Ministry of Health, Tel HaShomer, Israel.
| |
Collapse
|
26
|
Frizinsky S, Rechavi E, Barel O, Najeeb RH, Greenberger S, Lee YN, Simon AJ, Lev A, Ma CA, Sun G, Blackstone SA, Milner JD, Somech R, Stauber T. Novel MALT1 Mutation Linked to Immunodeficiency, Immune Dysregulation, and an Abnormal T Cell Receptor Repertoire. J Clin Immunol 2019; 39:401-413. [DOI: 10.1007/s10875-019-00629-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2018] [Accepted: 04/09/2019] [Indexed: 02/06/2023]
|
27
|
Fatal Pneumocystis jirovecii and Cytomegalovirus Infections in an Infant With Normal TRECs Count: Pitfalls of Newborn Screening for Severe Combined Immunodeficiency. Pediatr Infect Dis J 2019; 38:157-160. [PMID: 29613974 DOI: 10.1097/inf.0000000000002058] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Newborn screening for severe combined immunodeficiency using T-cell receptor excision circles allows prompt diagnosis and initiation of supportive and curative therapy thereby reducing morbidity and mortality. However, profound combined immunodeficiencies with normal numbers of nonfunctional T cells will go undetected. We present a patient with calcium release-activated calcium channel gene (ORAI1) deficiency and normal T-cell receptor excision circle numbers observed after diagnosis at the age of 14 months who suffered from disseminated fatal cytomegalovirus and Pneumocystis jirovecii infection, demonstrating a potential pitfall of the current newborn screening program.
Collapse
|
28
|
|
29
|
Alizadeh Z, Mazinani M, Houshmand M, Shakerian L, Nourizadeh M, Pourpak Z, Fazlollahi MR. Genetic Analysis of Patients with Two Different Types of Hyper IgM Syndrome. Immunol Invest 2018; 47:745-753. [PMID: 30081731 DOI: 10.1080/08820139.2018.1493052] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
BACKGROUND Hyper IgM Syndrome (HIGM) is a rare primary immunodeficiency in which impairment of class switching recombination (CSR) and somatic hyper-mutation (SHM) leads to recurrent infections. OBJECTIVES The aim of this study is to report the clinical and genetic features of six Iranian HIGM patients. METHODS Six patients, who suspected to have HIGM based on two clinical findings, including recurrent infections and low levels of IgG and IgA and normal or elevated levels of IgM, were entered this study to undergo genetic studies. Sanger sequencing was applied to detect pathogenic mutations in CD40L and AID genes causing two most common forms of HIGM, which known as HIGM type 1 and 2, respectively. RESULTS All patients who entered the study were males from unrelated families with a median age of 3.8 years. The most frequent clinical manifestation was recurrent pneumonia. Genetic studies of the patients revealed six different mutations, including five mutations in CD40L besides one mutation in AID. Two mutations in CD40L (p.F31fsX5 and p.C84S) were novel and three mutations (p. G219R, p.D62fsX18, and p.Q186X) have been previously reported. The mutation found in AID (p.E122X) was also previously described. CONCLUSION The study results may provide valuable information for prenatal diagnosis and also for genetic counseling especially for those who have a history of primary immunodeficiency in their family.
Collapse
Affiliation(s)
- Zahra Alizadeh
- a Immunology, Asthma & Allergy Research Institute , Tehran University of Medical Sciences , Tehran , Iran
| | - Marzieh Mazinani
- a Immunology, Asthma & Allergy Research Institute , Tehran University of Medical Sciences , Tehran , Iran
| | - Masoud Houshmand
- b Department of Medical Genetics , National Institute of Genetic Engineering and Biotechnology (NIGEB) , Tehran , Iran
| | - Leila Shakerian
- a Immunology, Asthma & Allergy Research Institute , Tehran University of Medical Sciences , Tehran , Iran
| | - Maryam Nourizadeh
- a Immunology, Asthma & Allergy Research Institute , Tehran University of Medical Sciences , Tehran , Iran
| | - Zahra Pourpak
- a Immunology, Asthma & Allergy Research Institute , Tehran University of Medical Sciences , Tehran , Iran
| | - Mohammad Reza Fazlollahi
- a Immunology, Asthma & Allergy Research Institute , Tehran University of Medical Sciences , Tehran , Iran
| |
Collapse
|
30
|
Dual loss of p110δ PI3-kinase and SKAP (KNSTRN) expression leads to combined immunodeficiency and multisystem syndromic features. J Allergy Clin Immunol 2018; 142:618-629. [DOI: 10.1016/j.jaci.2017.10.033] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 10/11/2017] [Accepted: 10/18/2017] [Indexed: 11/17/2022]
|
31
|
Nourizadeh M, Shakerian L, Borte S, Fazlollahi M, Badalzadeh M, Houshmand M, Alizadeh Z, Dalili H, Rashidi-Nezhad A, Kazemnejad A, Moin M, Hammarström L, Pourpak Z. Newborn screening using TREC/KREC assay for severe T and B cell lymphopenia in Iran. Scand J Immunol 2018; 88:e12699. [PMID: 29943473 DOI: 10.1111/sji.12699] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 06/11/2018] [Accepted: 06/20/2018] [Indexed: 01/01/2023]
Abstract
T-cell receptor excision circles (TRECs) and κ-deleting recombination excision circles (KRECs) are recently used for detection of T or B cell lymphopenia in neonates based on region-specific cutoff levels. Here, we report cutoffs for TREC and KREC copies useful for newborn screening and/or diagnosis of primary immunodeficiency diseases (PID) in Iran. DNA was extracted from a single 3.2 mm punch of dried blood spots collected from 2160 anonymized newborns referred to two major referral health centres between 2014 and 2016. For refinement of the cutoffs, 51 patients with a definite diagnosis of severe combined immunodeficiency, X-linked agammaglobulinaemia and combined immunodeficiency, including ataxia telangiectasia, human phosphoglucomutase 3 and Janus kinase-3 deficiency, as well as 47 healthy controls were included. Samples from patients with an X-linked hyper-IgM-syndrome, Wiskott-Aldrich syndrome and DNA ligase 4 deficiency were considered as disease controls. Triplex-quantitative real-time PCR was used. Cutoffs were calculated as TRECs < 11 and KRECs < 6 copies with an ACTB > 700 copies with sensitivity of 100% for TREC and 97% for KREC. Among thirty anonymized newborn samples (1.5%) with abnormal results for TREC and/or KREC, only twenty-one available cases were retested and shown to be in the normal range except for three samples (0.15%). All of the patients with a definitive diagnosis were correctly identified based on our established TREC/KREC copy numbers. Determining cutoffs for TREC/KREC is essential for correctly identifying children with PID in newborn screening. Early diagnosis of PID patients enables appropriate measures and therapies like stem cell transplantation.
Collapse
Affiliation(s)
- Maryam Nourizadeh
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Shakerian
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Stephan Borte
- ImmunoDeficiencyCenter Leipzig (IDCL), Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Municipal Hospital, Leipzig, Germany
- Department of Laboratory Medicine, Division of Clinical Immunology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Mohammadreza Fazlollahi
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohsen Badalzadeh
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Massoud Houshmand
- Medical Genetics Department, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | - Zahra Alizadeh
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Dalili
- Breastfeeding Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Rashidi-Nezhad
- Maternal, Fetal and Neonatal Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | | | - Mostafa Moin
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology and Allergy, Children Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Lennart Hammarström
- Department of Laboratory Medicine, Division of Clinical Immunology, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Zahra Pourpak
- Immunology, Asthma and Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
- Department of Immunology and Allergy, Children Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
32
|
Alazami AM, Al-Helale M, Alhissi S, Al-Saud B, Alajlan H, Monies D, Shah Z, Abouelhoda M, Arnaout R, Al-Dhekri H, Al-Numair NS, Ghebeh H, Sheikh F, Al-Mousa H. Novel CARMIL2 Mutations in Patients with Variable Clinical Dermatitis, Infections, and Combined Immunodeficiency. Front Immunol 2018; 9:203. [PMID: 29479355 PMCID: PMC5811477 DOI: 10.3389/fimmu.2018.00203] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022] Open
Abstract
Combined immunodeficiencies are a heterogeneous collection of primary immune disorders that exhibit defects in T cell development or function, along with impaired B cell activity even in light of normal B cell maturation. CARMIL2 (RLTPR) is a protein involved in cytoskeletal organization and cell migration, which also plays a role in CD28 co-signaling of T cells. Mutations in this protein have recently been reported to cause a novel primary immunodeficiency disorder with variable phenotypic presentations. Here, we describe seven patients from three unrelated, consanguineous multiplex families that presented with dermatitis, esophagitis, and recurrent skin and chest infections with evidence of combined immunodeficiency. Through the use of whole exome sequencing and autozygome-guided analysis, we uncovered two mutations not previously reported (p.R50T and p.L846Sfs) in CARMIL2. Real-time PCR analysis revealed that the biallelic frameshift mutation is under negative selection, likely due to nonsense-mediated RNA decay and leading to loss of detectable protein upon immunoblotting. Protein loss was also observed for the missense mutation, and 3D modeling suggested a disturbance in structural stability due to an increase in the electrostatic energy for the affected amino acid and surrounding residues. Immunophenotyping revealed that patient Treg counts were significantly depressed, and that CD4+ T cells were heavily skewed towards the naïve status. CD3/CD28 signaling impairment was evidenced by reduced proliferative response to stimulation. This work broadens the allelic heterogeneity associated with CARMIL2 and highlights a deleterious missense alteration located outside the leucine-rich repeat of the protein, where all other missense mutations have been reported to date.
Collapse
Affiliation(s)
- Anas M Alazami
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Maryam Al-Helale
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Safa Alhissi
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Bandar Al-Saud
- Department of Pediatrics, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Huda Alajlan
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Dorota Monies
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Zeeshan Shah
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Mohamed Abouelhoda
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Saudi Human Genome Program, King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Rand Arnaout
- Department of Medicine, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Alfaisal University, Riyadh, Saudi Arabia
| | - Hasan Al-Dhekri
- Department of Pediatrics, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Nouf S Al-Numair
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hazem Ghebeh
- Alfaisal University, Riyadh, Saudi Arabia.,Stem Cell & Tissue Re-Engineering Program, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Farrukh Sheikh
- Department of Medicine, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Hamoud Al-Mousa
- Department of Genetics, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Department of Pediatrics, Allergy and Immunology Section, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.,Alfaisal University, Riyadh, Saudi Arabia
| |
Collapse
|
33
|
Martire B, Azzari C, Badolato R, Canessa C, Cirillo E, Gallo V, Graziani S, Lorenzini T, Milito C, Panza R, Moschese V. Vaccination in immunocompromised host: Recommendations of Italian Primary Immunodeficiency Network Centers (IPINET). Vaccine 2018; 36:3541-3554. [PMID: 29426658 DOI: 10.1016/j.vaccine.2018.01.061] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2017] [Revised: 12/29/2017] [Accepted: 01/24/2018] [Indexed: 12/13/2022]
Abstract
Infectious complications are a major cause of morbidity and mortality in patients with primary or secondary immunodeficiency. Prevention of infectious diseases by vaccines is among the most effective healthcare measures mainly for these subjects. However immunocompromised people vary in their degree of immunosuppression and susceptibility to infection and, therefore, represent a heterogeneous population with regard to immunization. To date there is no well- established evidence for use of vaccines in immunodeficient patients, and indications are not clearly defined even in high-quality reviews and in most of the guidelines prepared to provide recommendations for the active vaccination of immunocompromised hosts. The aim of this document is to issue recommendations based on published literature and the collective experience of the Italian primary immunodeficiency centers, about how and when vaccines can be used in immunocompromised patients, in order to facilitate physician decisions and to ensure the best immune protection with the lowest risk to the health of the patient.
Collapse
Affiliation(s)
- Baldassarre Martire
- Paediatric Hematology Oncology Unit, "Policlinico-Giovanni XXII" Hospital, University of Bari, Italy.
| | - Chiara Azzari
- Pediatric Immunology Unit "Anna Meyer" Hospital University of Florence, Italy
| | - Raffaele Badolato
- Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Clementina Canessa
- Pediatric Immunology Unit "Anna Meyer" Hospital University of Florence, Italy
| | - Emilia Cirillo
- Department of Translational Medical Sciences, Pediatric section, Federico II University, Naples, Italy
| | - Vera Gallo
- Department of Translational Medical Sciences, Pediatric section, Federico II University, Naples, Italy
| | - Simona Graziani
- Paediatric Allergology and Immunology Unit, Policlinico Tor Vergata, University of Rome Tor, Vergata, Italy
| | - Tiziana Lorenzini
- Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - Cinzia Milito
- Department of Molecular Medicine, Sapienza University of Rome, Italy
| | - Raffaella Panza
- Paediatric Hematology Oncology Unit, "Policlinico-Giovanni XXII" Hospital, University of Bari, Italy
| | - Viviana Moschese
- Paediatric Allergology and Immunology Unit, Policlinico Tor Vergata, University of Rome Tor, Vergata, Italy
| | | |
Collapse
|
34
|
Biggs CM, Haddad E, Issekutz TB, Roifman CM, Turvey SE. Newborn screening for severe combined immunodeficiency: a primer for clinicians. CMAJ 2017; 189:E1551-E1557. [PMID: 29255099 PMCID: PMC5738248 DOI: 10.1503/cmaj.170561] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Affiliation(s)
- Catherine M Biggs
- Department of Pediatrics (Biggs, Turvey), British Columbia Children's Hospital, University of British Columbia, Vancouver, BC; Departments of Pediatrics, and Microbiology, Infection and Immunology (Haddad), University of Montreal, CHU Sainte-Justine, Montréal, Que.; Department of Pediatrics (Issekutz), IWK Health Centre, Dalhousie University, Halifax, NS; Division of Immunology and Allergy (Roifman), Hospital for Sick Children; Department of Pediatrics (Roifman), University of Toronto, Toronto, Ont
| | - Elie Haddad
- Department of Pediatrics (Biggs, Turvey), British Columbia Children's Hospital, University of British Columbia, Vancouver, BC; Departments of Pediatrics, and Microbiology, Infection and Immunology (Haddad), University of Montreal, CHU Sainte-Justine, Montréal, Que.; Department of Pediatrics (Issekutz), IWK Health Centre, Dalhousie University, Halifax, NS; Division of Immunology and Allergy (Roifman), Hospital for Sick Children; Department of Pediatrics (Roifman), University of Toronto, Toronto, Ont
| | - Thomas B Issekutz
- Department of Pediatrics (Biggs, Turvey), British Columbia Children's Hospital, University of British Columbia, Vancouver, BC; Departments of Pediatrics, and Microbiology, Infection and Immunology (Haddad), University of Montreal, CHU Sainte-Justine, Montréal, Que.; Department of Pediatrics (Issekutz), IWK Health Centre, Dalhousie University, Halifax, NS; Division of Immunology and Allergy (Roifman), Hospital for Sick Children; Department of Pediatrics (Roifman), University of Toronto, Toronto, Ont
| | - Chaim M Roifman
- Department of Pediatrics (Biggs, Turvey), British Columbia Children's Hospital, University of British Columbia, Vancouver, BC; Departments of Pediatrics, and Microbiology, Infection and Immunology (Haddad), University of Montreal, CHU Sainte-Justine, Montréal, Que.; Department of Pediatrics (Issekutz), IWK Health Centre, Dalhousie University, Halifax, NS; Division of Immunology and Allergy (Roifman), Hospital for Sick Children; Department of Pediatrics (Roifman), University of Toronto, Toronto, Ont
| | - Stuart E Turvey
- Department of Pediatrics (Biggs, Turvey), British Columbia Children's Hospital, University of British Columbia, Vancouver, BC; Departments of Pediatrics, and Microbiology, Infection and Immunology (Haddad), University of Montreal, CHU Sainte-Justine, Montréal, Que.; Department of Pediatrics (Issekutz), IWK Health Centre, Dalhousie University, Halifax, NS; Division of Immunology and Allergy (Roifman), Hospital for Sick Children; Department of Pediatrics (Roifman), University of Toronto, Toronto, Ont.
| |
Collapse
|
35
|
Erdle S, Ellis AK, Upton JEM. Advanced clinical testing of the adaptive immune system. Ann Allergy Asthma Immunol 2017; 118:655-663. [PMID: 28583259 DOI: 10.1016/j.anai.2017.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 04/05/2017] [Accepted: 04/06/2017] [Indexed: 11/19/2022]
Affiliation(s)
- Stephanie Erdle
- Department of Paediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Anne K Ellis
- Division of Allergy and Immunology, Department of Medicine, Queen's University, Kingston, Ontario, Canada
| | - Julia E M Upton
- Division of Immunology and Allergy, Department of Pediatrics, The Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
36
|
Alizadeh Z, Mazinani M, Shakerian L, Nabavi M, Fazlollahi MR. DOCK2 Deficiency in a Patient with Hyper IgM Phenotype. J Clin Immunol 2017; 38:10-12. [PMID: 29204803 DOI: 10.1007/s10875-017-0468-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2017] [Accepted: 11/27/2017] [Indexed: 10/18/2022]
Affiliation(s)
- Zahra Alizadeh
- Immunology, Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Mazinani
- Immunology, Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Leila Shakerian
- Immunology, Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Nabavi
- Department of Allergy and Clinical Immunology, Rasoul-e-Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Mohammad R Fazlollahi
- Immunology, Asthma & Allergy Research Institute, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
37
|
Peacock ME, Arce RM, Cutler CW. Periodontal and other oral manifestations of immunodeficiency diseases. Oral Dis 2017; 23:866-888. [PMID: 27630012 PMCID: PMC5352551 DOI: 10.1111/odi.12584] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Revised: 08/31/2016] [Accepted: 09/08/2016] [Indexed: 12/14/2022]
Abstract
The list of immunodeficiency diseases grows each year as novel disorders are discovered, classified, and sometimes reclassified due to our ever-increasing knowledge of immune system function. Although the number of patients with secondary immunodeficiencies (SIDs) greatly exceeds those with primary immunodeficiencies (PIDs), the prevalence of both appears to be on the rise probably because of scientific breakthroughs that facilitate earlier and more accurate diagnosis. Primary immunodeficiencies in adults are not as rare as once thought. Globally, the main causes of secondary immunodeficiency are HIV infection and nutritional insufficiencies. Persons with acquired immune disorders such as AIDS caused by the human immunodeficiency virus (HIV) are now living long and fulfilling lives as a result of highly active antiretroviral therapy (HAART). Irrespective of whether the patient's immune-deficient state is a consequence of a genetic defect or is secondary in nature, dental and medical practitioners must be aware of the constant potential for infections and/or expressions of autoimmunity in these individuals. The purpose of this review was to study the most common conditions resulting from primary and secondary immunodeficiency states, how they are classified, and the detrimental manifestations of these disorders on the periodontal and oral tissues.
Collapse
Affiliation(s)
- Mark E Peacock
- Associate Professor, Departments of Periodontics, Oral Biology
| | - Roger M. Arce
- Assistant Professor, Departments of Periodontics, Oral Biology
| | - Christopher W Cutler
- Professor, Departments of Periodontics, Oral Biology; Chair, Department of Periodontics, Associate Dean for Research, The Dental College of Georgia at Augusta University
| |
Collapse
|
38
|
Yáñez L, Lama P, Rivacoba C, Zamorano J, Marinovic MA. [Primary immunodeficiencies in seriously ill children: Report of 3 clinical cases]. ACTA ACUST UNITED AC 2017; 88:136-141. [PMID: 28288231 DOI: 10.1016/j.rchipe.2016.07.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2015] [Accepted: 07/25/2016] [Indexed: 10/20/2022]
Abstract
Primary immunodeficiency diseases (PID) are congenital disorders secondary to an impaired immune response. Infections, autoimmune disorders, atopy, and lymphoproliferative syndromes are commonly associated with this disorder. OBJECTIVE To present and discuss 3 infants diagnosed with PID. CLINICAL CASES The cases are presented of three patients with PID diagnosed during their first admission to a Paediatric Intensive Critical Care Unit. The first patient, a 4-month-old infant affected by a severe pneumonia, and was diagnosed as a Severe Combined Immunodeficiency Disease. The second patient was an 8-month-old infant with Candida lusitaniae mesenteric adenitis, and diagnosed with a Chronic Granulomatous Disease. The last patient, a 6-month-old infant presented with ecthyma gangrenosum and X-linked agammaglobulinaemia. CONCLUSION PID should be suspected when an infectious disease does not responde to the appropriate therapy within the expected period. An update of each disease is presented.
Collapse
Affiliation(s)
- Leticia Yáñez
- Unidad de Paciente Crítico Pediátrico, Clínica Santa María, Santiago, Chile
| | - Pamela Lama
- Unidad de Paciente Crítico Pediátrico, Clínica Santa María, Santiago, Chile
| | - Carolina Rivacoba
- Unidad de Paciente Crítico Pediátrico, Clínica Santa María, Santiago, Chile
| | - Juanita Zamorano
- Unidad de Paciente Crítico Pediátrico, Clínica Santa María, Santiago, Chile
| | | |
Collapse
|
39
|
Mauracher AA, Pagliarulo F, Faes L, Vavassori S, Güngör T, Bachmann LM, Pachlopnik Schmid J. Causes of low neonatal T-cell receptor excision circles: A systematic review. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY-IN PRACTICE 2017; 5:1457-1460.e22. [DOI: 10.1016/j.jaip.2017.02.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 01/03/2017] [Accepted: 02/03/2017] [Indexed: 10/19/2022]
|
40
|
Dadi H, Jones TA, Merico D, Sharfe N, Ovadia A, Schejter Y, Reid B, Sun M, Vong L, Atkinson A, Lavi S, Pomerantz JL, Roifman CM. Combined immunodeficiency and atopy caused by a dominant negative mutation in caspase activation and recruitment domain family member 11 (CARD11). J Allergy Clin Immunol 2017; 141:1818-1830.e2. [PMID: 28826773 DOI: 10.1016/j.jaci.2017.06.047] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Revised: 06/27/2017] [Accepted: 06/30/2017] [Indexed: 11/18/2022]
Abstract
BACKGROUND Combined immunodeficiency (CID) is a T-cell defect frequently presenting with recurrent infections, as well as associated immune dysregulation manifesting as autoimmunity or allergic inflammation. OBJECTIVE We sought to identify the genetic aberration in 4 related patients with CID, early-onset asthma, eczema, and food allergies, as well as autoimmunity. METHODS We performed whole-exome sequencing, followed by Sanger confirmation, assessment of the genetic variant effect on cell signaling, and evaluation of the resultant immune function. RESULTS A heterozygous novel c.C88T 1-bp substitution resulting in amino acid change R30W in caspase activation and recruitment domain family member 11 (CARD11) was identified by using whole-exome sequencing and segregated perfectly to family members with severe atopy only but was not found in healthy subjects. We demonstrate that the R30W mutation results in loss of function while also exerting a dominant negative effect on wild-type CARD11. The CARD11 defect altered the classical nuclear factor κB pathway, resulting in poor in vitro T-cell responses to mitogens and antigens caused by reduced secretion of IFN-γ and IL-2. CONCLUSION Unlike patients with biallelic mutations in CARD11 causing severe CID, the R30W defect results in a less profound yet prominent susceptibility to infections, as well as multiorgan atopy and autoimmunity.
Collapse
Affiliation(s)
- Harjit Dadi
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Tyler A Jones
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Md
| | | | - Nigel Sharfe
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Adi Ovadia
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Yael Schejter
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Brenda Reid
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Mark Sun
- Deep Genomics, Toronto, Ontario, Canada
| | - Linda Vong
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Adelle Atkinson
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada
| | - Sasson Lavi
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada
| | - Joel L Pomerantz
- Department of Biological Chemistry and Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, Md
| | - Chaim M Roifman
- Division of Immunology and Allergy, Department of Pediatrics, Hospital for Sick Children and the University of Toronto, Toronto, Ontario, Canada; Canadian Centre for Primary Immunodeficiency and the Jeffrey Modell Research Laboratory for the Diagnosis of Primary Immunodeficiency, Hospital for Sick Children, Toronto, Ontario, Canada.
| |
Collapse
|
41
|
Azarsiz E, Karaca N, Ergun B, Durmuscan M, Kutukculer N, Aksu G. In vitro T lymphocyte proliferation by carboxyfluorescein diacetate succinimidyl ester method is helpful in diagnosing and managing primary immunodeficiencies. J Clin Lab Anal 2017; 32. [PMID: 28383134 DOI: 10.1002/jcla.22216] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 02/28/2017] [Indexed: 01/16/2023] Open
Abstract
BACKGROUND Functional studies besides routine laboratory tests for the definitive diagnosis of T lymphocyte disorders with isolated T or combined T/B-cell immunodeficiencies are important. We hereby summarized our experience with a carboxyfluorescein diacetate succinimidyl ester (CFSE)-based assay for the assessment of mitogenic T-cell proliferation responses in primary immunodeficiency (PID) patients who have not been diagnosed yet or genetically analyzed, but classified as probably having T-cell defects. METHODS Unclassified patients (n=46) and controls (n=25) were evaluated for T-cell disorders with CFSE-based assay. RESULTS CD3+ blast cells after PHA-L stimulation were significantly lower in patients (31.1±28.8) than controls (67.9±8.79; P<.001). Nine patients with low and four patients with normal CD3 values had severely decreased blastic transformation. The proliferation response decreased mostly in combined immunodeficiency group. Sixteen of them had impaired proliferation responses. Appropriate molecular genetical analyses were planned after thorough evaluation of each patient. CONCLUSIONS In vitro lymphocyte cell proliferation analysis by CFSE method is a reliable and practical choice for the assessment of mitogenic T lymphocyte responses in yet unclassified PID patients for targeting further genetical analyses.
Collapse
Affiliation(s)
- Elif Azarsiz
- Department of Pediatric Immunology, Ege University, Izmir, Turkey
| | - Neslihan Karaca
- Department of Pediatric Immunology, Ege University, Izmir, Turkey
| | - Birgul Ergun
- Department of Pediatric Immunology, Ege University, Izmir, Turkey
| | - Mehmet Durmuscan
- Department of Pediatric Immunology, Ege University, Izmir, Turkey
| | - Necil Kutukculer
- Department of Pediatric Immunology, Ege University, Izmir, Turkey
| | - Guzide Aksu
- Department of Pediatric Immunology, Ege University, Izmir, Turkey
| |
Collapse
|
42
|
Aluri J, Italia K, Gupta M, Dalvi A, Bavdekar A, Madkaikar M. Low T cell receptor excision circles (TRECs) in a case of ZAP 70 deficient severe combined immunodeficiency (SCID) with a novel mutation from India. Blood Cells Mol Dis 2016; 65:95-96. [PMID: 27839984 DOI: 10.1016/j.bcmd.2016.10.022] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Accepted: 10/29/2016] [Indexed: 12/13/2022]
Affiliation(s)
- Jahnavi Aluri
- Department of Pediatric Immunology and Leukocyte Biology, National Institute of Immunohematology (ICMR), 13th Floor, NMS Building, KEM Hospital, Parel, Mumbai 400012, India
| | - Khushnuma Italia
- Department of Pediatric Immunology and Leukocyte Biology, National Institute of Immunohematology (ICMR), 13th Floor, NMS Building, KEM Hospital, Parel, Mumbai 400012, India
| | - Maya Gupta
- Department of Pediatric Immunology and Leukocyte Biology, National Institute of Immunohematology (ICMR), 13th Floor, NMS Building, KEM Hospital, Parel, Mumbai 400012, India
| | - Aparna Dalvi
- Department of Pediatric Immunology and Leukocyte Biology, National Institute of Immunohematology (ICMR), 13th Floor, NMS Building, KEM Hospital, Parel, Mumbai 400012, India
| | - Ashish Bavdekar
- KEM hospital, 489, Rasta Peth, Sardar Moodilar Road, Pune 411011, India
| | - Manisha Madkaikar
- Department of Pediatric Immunology and Leukocyte Biology, National Institute of Immunohematology (ICMR), 13th Floor, NMS Building, KEM Hospital, Parel, Mumbai 400012, India.
| |
Collapse
|
43
|
Speckmann C, Doerken S, Aiuti A, Albert MH, Al-Herz W, Allende LM, Scarselli A, Avcin T, Perez-Becker R, Cancrini C, Cant A, Di Cesare S, Finocchi A, Fischer A, Gaspar HB, Ghosh S, Gennery A, Gilmour K, González-Granado LI, Martinez-Gallo M, Hambleton S, Hauck F, Hoenig M, Moshous D, Neven B, Niehues T, Notarangelo L, Picard C, Rieber N, Schulz A, Schwarz K, Seidel MG, Soler-Palacin P, Stepensky P, Strahm B, Vraetz T, Warnatz K, Winterhalter C, Worth A, Fuchs S, Uhlmann A, Ehl S. A prospective study on the natural history of patients with profound combined immunodeficiency: An interim analysis. J Allergy Clin Immunol 2016; 139:1302-1310.e4. [PMID: 27658761 DOI: 10.1016/j.jaci.2016.07.040] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2015] [Revised: 07/21/2016] [Accepted: 07/28/2016] [Indexed: 12/14/2022]
Abstract
BACKGROUND Absent T-cell immunity resulting in life-threatening infections provides a clear rationale for hematopoetic stem cell transplantation (HSCT) in patients with severe combined immunodeficiency (SCID). Combined immunodeficiencies (CIDs) and "atypical" SCID show reduced, not absent T-cell immunity. If associated with infections or autoimmunity, they represent profound combined immunodeficiency (P-CID), for which outcome data are insufficient for unambiguous early transplant decisions. OBJECTIVES We sought to compare natural histories of severity-matched patients with/without subsequent transplantation and to determine whether immunologic and/or clinical parameters may be predictive for outcome. METHODS In this prospective and retrospective observational study, we recruited nontransplanted patients with P-CID aged 1 to 16 years to compare natural histories of severity-matched patients with/without subsequent transplantation and to determine whether immunologic and/or clinical parameters may be predictive for outcome. RESULTS A total of 51 patients were recruited (median age, 9.6 years). Thirteen of 51 had a genetic diagnosis of "atypical" SCID and 14 of 51 of CID. About half of the patients had less than 10% naive T cells, reduced/absent T-cell proliferation, and at least 1 significant clinical event/year, demonstrating their profound immunodeficiency. Nineteen patients (37%) underwent transplantation within 1 year of enrolment, and 5 of 51 patients died. Analysis of the HSCT decisions revealed the anticipated heterogeneity, favoring an ongoing prospective matched-pair analysis of patients with similar disease severity with or without transplantation. Importantly, so far neither the genetic diagnosis nor basic measurements of T-cell immunity were good predictors of disease evolution. CONCLUSIONS The P-CID study for the first time characterizes a group of patients with nontypical SCID T-cell deficiencies from a therapeutic perspective. Because genetic and basic T-cell parameters provide limited guidance, prospective data from this study will be a helpful resource for guiding the difficult HSCT decisions in patients with P-CID.
Collapse
Affiliation(s)
- Carsten Speckmann
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Sam Doerken
- Institute for Medical Biometry and Statistics, Center for Medical Biometry and Medical Informatics, Medical Center - University of Freiburg, Freiburg, Germany
| | - Alessandro Aiuti
- Pediatric Immunohematology and Bone Marrow Transplant Unit, San Raffaele Scientific Institute, Milan, Italy; Department of Pediatrics, Ospedale Pediatrico Bambino Gesù and University of Rome "Tor Vergata," Rome, Italy
| | - Michael H Albert
- Dr von Hauner Children's Hospital, Ludwig-Maximilians-University, Munich, Germany
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Luis M Allende
- Servicio de Inmunología, Hospital Universitario 12 de Octubre and Instituto de Investigación i+12, Madrid, Spain
| | - Alessia Scarselli
- Department of Pediatrics, Ospedale Pediatrico Bambino Gesù and University of Rome "Tor Vergata," Rome, Italy
| | - Tadej Avcin
- Department of Allergology, Rheumatology and Clinical Immunology, University Children's Hospital, University Medical Center, Ljubljana, Slovenia
| | - Ruy Perez-Becker
- Center for Pediatrics and Adolescent Medicine, Helios Hospital Krefeld, Krefeld, Germany
| | - Caterina Cancrini
- Department of Pediatrics, Ospedale Pediatrico Bambino Gesù and University of Rome "Tor Vergata," Rome, Italy
| | - Andrew Cant
- Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Silvia Di Cesare
- Department of Pediatrics, Ospedale Pediatrico Bambino Gesù and University of Rome "Tor Vergata," Rome, Italy
| | - Andrea Finocchi
- Department of Pediatrics, Ospedale Pediatrico Bambino Gesù and University of Rome "Tor Vergata," Rome, Italy
| | - Alain Fischer
- AP-HP, Hôpital Necker-Enfants Malades, Immunologie et Hématologie Pédiatriques, Paris, France
| | - H Bobby Gaspar
- Department of Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Sujal Ghosh
- Department of Pediatric Oncology, Hematology and Clinical Immunology, Medical Faculty, Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany
| | - Andrew Gennery
- Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Kimberly Gilmour
- Department of Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Luis I González-Granado
- Servicio de Inmunología, Hospital Universitario 12 de Octubre and Instituto de Investigación i+12, Madrid, Spain; Immunodeficiencies Unit, Hematology & Oncology Unit, Pediatrics, Hospital 12 Octubre, Madrid, Spain
| | - Monica Martinez-Gallo
- Immunology Division, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Sophie Hambleton
- Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Fabian Hauck
- Immunodeficiency Unit and Immunological Diagnostics Laboratory, Dr von Hauner Children's Hospital Ludwig-Maximilians-University, Munich, Germany
| | - Manfred Hoenig
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Despina Moshous
- AP-HP, Hôpital Necker-Enfants Malades, Immunologie et Hématologie Pédiatriques, Paris, France; INSERM UMR1163, Genome Dynamics in the Immune System, Université Paris Descartes - Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Benedicte Neven
- AP-HP, Hôpital Necker-Enfants Malades, Immunologie et Hématologie Pédiatriques, Paris, France
| | - Tim Niehues
- Center for Pediatrics and Adolescent Medicine, Helios Hospital Krefeld, Krefeld, Germany
| | - Luigi Notarangelo
- Division of Immunology, Boston Children's Hospital, and Department of Pediatrics, Harvard Medical School, Boston, Mass
| | - Capucine Picard
- AP-HP, Hôpital Necker-Enfants Malades, Immunologie et Hématologie Pédiatriques, Paris, France; INSERM UMR1163, Genome Dynamics in the Immune System, Université Paris Descartes - Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Nikolaus Rieber
- Department of Pediatrics I, University of Tübingen, Tübingen, Germany; Department of Pediatrics, StKM GmbH and Technical University Muenchen, Munich, Germany
| | - Ansgar Schulz
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University of Ulm, and the Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service, Baden-Württemberg-Hessen, Ulm, Germany
| | - Markus G Seidel
- Department of Pediatrics and Adolescent Medicine, Division of Pediatric Hematology-Oncology, Medical University Graz, Graz, Austria
| | - Pere Soler-Palacin
- Immunology Division, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institut de Recerca, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Polina Stepensky
- Pediatric Hematology-Oncology and Bone Marrow Transplantation, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Brigitte Strahm
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Thomas Vraetz
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Klaus Warnatz
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Christine Winterhalter
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Clinical Trials Unit, Medical Center - University of Freiburg, Freiburg, Germany
| | - Austen Worth
- Department of Immunology, Great Ormond Street Hospital NHS Foundation Trust, London, United Kingdom
| | - Sebastian Fuchs
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany
| | - Annette Uhlmann
- Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Clinical Trials Unit, Medical Center - University of Freiburg, Freiburg, Germany
| | - Stephan Ehl
- Department of Pediatric Hematology and Oncology, Center for Pediatrics, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany; Center for Chronic Immunodeficiency, Medical Center - University of Freiburg, Faculty of Medicine, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|
44
|
Cuvelier GDE, Rubin TS, Wall DA, Schroeder ML. Long-Term Outcomes of Hematopoietic Stem Cell Transplantation for ZAP70 Deficiency. J Clin Immunol 2016; 36:713-24. [PMID: 27438785 DOI: 10.1007/s10875-016-0316-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2016] [Accepted: 07/07/2016] [Indexed: 12/23/2022]
Abstract
ZAP70 deficiency is a rare T + B + NK+ combined immunodeficiency with limited outcome data to help guide decisions around hematopoietic stem cell transplant (HSCT). We sought to understand the long-term clinical and immunologic outcomes of both conditioned and unconditioned HSCT for ZAP70 deficiency following transplant from a variety of graft sources. We performed a retrospective, single center review of all cases of HSCT for genetically confirmed ZAP70 deficiency since 1992. At a median of 13.5-year post-HSCT, 8/8 (100 %) patients are alive. Three received unconditioned bone marrow transplants from human leukocyte antigen (HLA)-matched siblings and achieved stable mixed donor-recipient T cell chimerism but low B cell (4-9 %) and absent to near-absent myeloid donor engraftment. Despite this, all three have normal immunoglobulin levels, have developed specific protective antibody responses to post-HSCT vaccinations, and have discontinued immunoglobulin replacement. Five patients received myeloablative conditioning (three T cell-depleted haploidentical and two unrelated cord blood) and have full donor chimerism for T and B cells and myeloid lineages. One patient experienced primary graft failure after serotherapy only. CD8 T cell count is normal in 5/8, high in 1/8, and low in 2/8. Infectious complications in 5/5 and autoimmune thrombocytopenia in one patient resolved post-HSCT. Mitogen proliferation to phytohemagglutinin was normal after HSCT in 8/8 patients. In total, seven have discontinued immunoglobulin replacement. In conclusion, HSCT using a variety of graft sources and approaches, including unconditioned matched sibling donor transplant, is a life-saving therapy for ZAP70 deficiency, providing excellent long-term immune function and resolution of clinical problems.
Collapse
Affiliation(s)
- Geoffrey D E Cuvelier
- Manitoba Blood and Marrow Transplant Program, Division of Pediatric Hematology-Oncology, CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada. .,Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada. .,CancerCare Manitoba, ON2011-675 McDermot Avenue, Winnipeg, Manitoba, R3E 0V9, Canada.
| | - Tamar S Rubin
- Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada.,Division of Allergy and Clinical Immunology, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Donna A Wall
- Manitoba Blood and Marrow Transplant Program, Division of Pediatric Hematology-Oncology, CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada.,Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Marlis L Schroeder
- Manitoba Blood and Marrow Transplant Program, Division of Pediatric Hematology-Oncology, CancerCare Manitoba, 675 McDermot Avenue, Winnipeg, Manitoba, Canada.,Department of Pediatrics and Child Health, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
45
|
Abstract
INTRODUCTION Newborn screening (NBS) for Severe combined immunodeficiency (SCID)/severe T cell lymphopenia (sTCL) is being increasingly used worldwide. AREAS COVERED In this manuscript we will discuss the following: 1) The rationale for screening newborns for SCID/sTCL; 2) The scientific basis for the use of the T cell receptor excision circle (TREC) assay in screening newborns for SCID/sTCL; 3) The published outcomes of current NBS programs. Expert commentary: 4) Some of the ethical dilemmas that occur when screening newborns for SCID. Finally, we will discuss the future directions for expanding NBS to include other primary immunodeficiencies.
Collapse
Affiliation(s)
- Becky J Buelow
- a Department of Pediatrics , Medical College of Wisconsin , Milwaukee , WI , USA
| | - James W Verbsky
- a Department of Pediatrics , Medical College of Wisconsin , Milwaukee , WI , USA.,b Department of Microbiology and Molecular Genetics , Medical College of Wisconsin and the Children's Research Institute, Medical College of Wisconsin , Milwaukee , WI , USA
| | - John M Routes
- a Department of Pediatrics , Medical College of Wisconsin , Milwaukee , WI , USA.,b Department of Microbiology and Molecular Genetics , Medical College of Wisconsin and the Children's Research Institute, Medical College of Wisconsin , Milwaukee , WI , USA
| |
Collapse
|
46
|
O'Keefe AW, Halbrich M, Ben-Shoshan M, McCusker C. Primary immunodeficiency for the primary care provider. Paediatr Child Health 2016; 21:e10-4. [PMID: 27095888 DOI: 10.1093/pch/21.2.e10] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Primary immunodeficiencies are a group of heterogeneous disorders resulting from defects affecting the function of ≥1 parts of the immune system. Current estimates of the prevalence of primary immunodeficiency disease are one in 1200 patients. In Ontario, where the average general practitioner follows 1300 to 2000 patients, an estimated two patients will have primary immunodeficiency. With new primary immunodeficiencies being described at an exponential rate, and those previously described becoming better understood, it is challenging for health care providers to stay up to date. Knowledge gaps delay diagnosis and treatment, leading to increased morbidity and mortality. The present review aims to provide the primary care provider with the tools necessary to recognize primary immunodeficiency and assist in establishing diagnoses.
Collapse
Affiliation(s)
- A W O'Keefe
- Division of Pediatrics, Faculty of Medicine, Memorial University, St John's, Newfoundland & Labrador
| | - M Halbrich
- Pediatrics, Faculty of Medicine, University of Manitoba,Winnipeg, Manitoba
| | - M Ben-Shoshan
- Division of Pediatric Allergy and Clinical Immunology, Faculty of Medicine, McGill University Health Centre, Montreal, Quebec
| | - C McCusker
- Division of Pediatric Allergy and Clinical Immunology, Faculty of Medicine, McGill University Health Centre, Montreal, Quebec
| |
Collapse
|
47
|
The effects of STAT1 dysfunction on the gut. LYMPHOSIGN JOURNAL-THE JOURNAL OF INHERITED IMMUNE DISORDERS 2016. [DOI: 10.14785/lpsn-2015-0012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Introduction: Mutations in the signal transducer and activator of transcription1 (STAT1) have been associated with a variety of clinical patterns. Interestingly patients with heterozygous mutations in the DNA binding domain (DBD) of STAT1 suffer acute and chronic colitis. Methods: To further analyze the role of STAT1 deficiency in intestinal inflammation, we employed protein expression analysis of total and activated STAT1 in intestinal biopsy samples from 2 patients with heterozygous mutations in the DBD of the STAT1 gene. Results: Both patients showed clinical and histological features of colitis. Total and activated STAT1 were decreased in duodenal and colonic enterocytes, and total STAT1 was found to be mislocalized in aggregates subapically. In addition, intestinal biopsy samples showed decreased numbers of lymphocytes. Patient-derived lymphoblasts demonstrated lack of viability and high susceptibility for cell death. Conclusion: STAT1 expression and distribution in the gut of patients with mutations in the DBD are abnormal, suggesting a primary role of STAT1 dysfunction in enterocytes in addition to the secondary effect of aberrant inflammation. Statement of novelty: Colitis associated with STAT1 mutations appears to have unique features distinct from typical inflammatory bowel disease.
Collapse
|
48
|
Machnes-Maayan D, Lev A, Katz U, Mishali D, Vardi A, Simon AJ, Somech R. Insight into normal thymic activity by assessment of peripheral blood samples. Immunol Res 2015; 61:198-205. [PMID: 25294167 DOI: 10.1007/s12026-014-8558-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The thymus is a highly specialized organ for T cell receptor (TCR) rearrangement and selection mechanisms that ensure the formation of functional and self-tolerant cells. Little is known about how peripheral blood assessment of thymic function reflects thymus activity during infancy. We compared thymic function-related markers in the thymus with those in peripheral blood in order to check their correlations. We concomitantly blood samples from immunocompetent infants who underwent cardiac surgery that involved thymectomy. The studied thymic markers included TCR excision circles (TRECs), four different TCRD (TCR delta chain) gene rearrangements, the TCR repertoire, regulatory T cells (Tregs, defined as the CD4+CD25+FOXP3+ cell population) and real-time quantitative polymerase chain reaction (RQ-PCR) mRNA expression of forkhead box P3 (FOXP3). Twenty patients were enrolled in this study. Their mean age at the time of the surgery was 3 months/5 days ± 3 months/18 days. There was a significant correlation between thymic and peripheral blood levels of TREC, all four TCRD gene rearrangements and the amount of Tregs. The levels of these parameters were significantly higher in the thymus than those detected in the peripheral blood. The TCR repertoire distribution in both samples was similar. FOXP3 mRNA levels in the thymus and peripheral blood correlated well. Our findings demonstrated a strong and significant correlation between peripheral blood and intra-thymic activity parameters during infancy. Assessment of these parameters in peripheral blood can be used to accurately estimate different intra-thymic capacities for assessing T cell function in health and disease.
Collapse
Affiliation(s)
- Diti Machnes-Maayan
- Pediatric Department B, Pediatric Immunology Service, Jeffrey Modell Foundation (JMF) Center, Edmond and Lily Safra Children's Hospital, Chaim Sheba Medical Center, 52621, Tel Hashomer, Israel
| | | | | | | | | | | | | |
Collapse
|
49
|
Sharfe N, Merico D, Karanxha A, Macdonald C, Dadi H, Ngan B, Herbrick JA, Roifman CM. The effects of RelB deficiency on lymphocyte development and function. J Autoimmun 2015; 65:90-100. [DOI: 10.1016/j.jaut.2015.09.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Revised: 09/03/2015] [Accepted: 09/05/2015] [Indexed: 10/23/2022]
|
50
|
Elgizouli M, Lowe DM, Speckmann C, Schubert D, Hülsdünker J, Eskandarian Z, Dudek A, Schmitt-Graeff A, Wanders J, Jørgensen SF, Fevang B, Salzer U, Nieters A, Burns S, Grimbacher B. Activating PI3Kδ mutations in a cohort of 669 patients with primary immunodeficiency. Clin Exp Immunol 2015; 183:221-9. [PMID: 26437962 DOI: 10.1111/cei.12706] [Citation(s) in RCA: 73] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/17/2015] [Indexed: 12/17/2022] Open
Abstract
The gene PIK3CD codes for the catalytic subunit of phosphoinositide 3-kinase δ (PI3Kδ), and is expressed solely in leucocytes. Activating mutations of PIK3CD have been described to cause an autosomal dominant immunodeficiency that shares clinical features with common variable immunodeficiency (CVID). We screened a cohort of 669 molecularly undefined primary immunodeficiency patients for five reported mutations (four gain-of-function mutations in PIK3CD and a loss of function mutation in PIK3R1) using pyrosequencing. PIK3CD mutations were identified in three siblings diagnosed with CVID and two sporadic cases with a combined immunodeficiency (CID). The PIK3R1 mutation was not identified in the cohort. Our patients with activated PI3Kδ syndrome (APDS) showed a range of clinical and immunological findings, even within a single family, but shared a reduction in naive T cells. PIK3CD gain of function mutations are more likely to occur in patients with defective B and T cell responses and should be screened for in CVID and CID, but are less likely in patients with a pure B cell/hypogammaglobulinaemia phenotype.
Collapse
Affiliation(s)
- M Elgizouli
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany.,Faculty of Biology, Albert Ludwigs University, Freiburg, Germany
| | - D M Lowe
- Institute of Immunity and Transplantation, University College London, London, UK
| | - C Speckmann
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany.,Department of Pediatrics and Adolescent Medicine, University Medical Center, Freiburg, Germany
| | - D Schubert
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University, Freiburg, Germany.,Faculty of Biology, Albert Ludwigs University, Freiburg, Germany
| | - J Hülsdünker
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University, Freiburg, Germany
| | - Z Eskandarian
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - A Dudek
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany.,Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University, Freiburg, Germany
| | - A Schmitt-Graeff
- Department of Pathology, University Medical Center, Freiburg, Germany
| | - J Wanders
- Institute of Immunity and Transplantation, University College London, London, UK
| | - S F Jørgensen
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - B Fevang
- Research Institute of Internal Medicine, Oslo University Hospital and University of Oslo, and Section of Clinical Immunology and Infectious Diseases, Oslo University Hospital Rikshospitalet, Oslo, Norway
| | - U Salzer
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - A Nieters
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - S Burns
- Institute of Immunity and Transplantation, University College London, London, UK
| | - B Grimbacher
- Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany.,Institute of Immunity and Transplantation, University College London, London, UK
| |
Collapse
|