1
|
Erman B, Aba U, Ipsir C, Pehlivan D, Aytekin C, Cildir G, Cicek B, Bozkurt C, Tekeoglu S, Kaya M, Aydogmus C, Cipe F, Sucak G, Eltan SB, Ozen A, Barıs S, Karakoc-Aydiner E, Kıykım A, Karaatmaca B, Kose H, Uygun DFK, Celmeli F, Arikoglu T, Ozcan D, Keskin O, Arık E, Aytekin ES, Cesur M, Kucukosmanoglu E, Kılıc M, Yuksek M, Bıcakcı Z, Esenboga S, Ayvaz DÇ, Sefer AP, Guner SN, Keles S, Reisli I, Musabak U, Demirbas ND, Haskologlu S, Kilic SS, Metin A, Dogu F, Ikinciogulları A, Tezcan I. Genetic Evaluation of the Patients with Clinically Diagnosed Inborn Errors of Immunity by Whole Exome Sequencing: Results from a Specialized Research Center for Immunodeficiency in Türkiye. J Clin Immunol 2024; 44:157. [PMID: 38954121 PMCID: PMC11219406 DOI: 10.1007/s10875-024-01759-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 06/22/2024] [Indexed: 07/04/2024]
Abstract
Molecular diagnosis of inborn errors of immunity (IEI) plays a critical role in determining patients' long-term prognosis, treatment options, and genetic counseling. Over the past decade, the broader utilization of next-generation sequencing (NGS) techniques in both research and clinical settings has facilitated the evaluation of a significant proportion of patients for gene variants associated with IEI. In addition to its role in diagnosing known gene defects, the application of high-throughput techniques such as targeted, exome, and genome sequencing has led to the identification of novel disease-causing genes. However, the results obtained from these different methods can vary depending on disease phenotypes or patient characteristics. In this study, we conducted whole-exome sequencing (WES) in a sizable cohort of IEI patients, consisting of 303 individuals from 21 different clinical immunology centers in Türkiye. Our analysis resulted in likely genetic diagnoses for 41.1% of the patients (122 out of 297), revealing 52 novel variants and uncovering potential new IEI genes in six patients. The significance of understanding outcomes across various IEI cohorts cannot be overstated, and we believe that our findings will make a valuable contribution to the existing literature and foster collaborative research between clinicians and basic science researchers.
Collapse
Affiliation(s)
- Baran Erman
- Institute of Child Health, Hacettepe University, Ankara, Turkey.
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey.
| | - Umran Aba
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
- Department of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Canberk Ipsir
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
- Department of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Damla Pehlivan
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
| | - Caner Aytekin
- Pediatric Immunology, SBU Ankara Dr Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Gökhan Cildir
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5000, Australia
| | - Begum Cicek
- Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Ceren Bozkurt
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
| | - Sidem Tekeoglu
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
| | - Melisa Kaya
- Can Sucak Research Laboratory for Translational Immunology, Hacettepe University, Ankara, Turkey
| | - Cigdem Aydogmus
- Department of Pediatric Allergy and Clinical Immunology, University of Health Sciences, Istanbul Basaksehir Cam and Sakura City Hospital, Istanbul, Turkey
| | - Funda Cipe
- Department of Pediatric Allergy and Clinical Immunology, Altinbas University School of Medicine, Istanbul, Turkey
| | - Gulsan Sucak
- Medical Park Bahçeşehir Hospital, Clinic of Hematology and Transplantation, İstanbul, Turkey
| | - Sevgi Bilgic Eltan
- Marmara University, Faculty of Medicine, Department of Pediatric Allergy and Immunology, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ahmet Ozen
- Marmara University, Faculty of Medicine, Department of Pediatric Allergy and Immunology, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Safa Barıs
- Marmara University, Faculty of Medicine, Department of Pediatric Allergy and Immunology, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Marmara University, Faculty of Medicine, Department of Pediatric Allergy and Immunology, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkey
| | - Ayca Kıykım
- Pediatric Allergy and Immunology, Cerrahpasa School of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| | - Betul Karaatmaca
- Department of Pediatric Allergy and Immunology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Hulya Kose
- Department of Pediatric Immunology, Diyarbakir Children Hospital, Diyarbakır, Turkey
| | - Dilara Fatma Kocacık Uygun
- Division of Allergy Immunology, Department of Pediatrics, Akdeniz University Faculty of Medicine, Antalya, Turkey
| | - Fatih Celmeli
- Republic of Turkey Ministry of Health Antalya Training and Research Hospital Pediatric Immunology and Allergy Diseases, Antalya, Turkey
| | - Tugba Arikoglu
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Mersin University, Mersin, Turkey
| | - Dilek Ozcan
- Division of Pediatric Allergy and Immunology, Faculty of Medicine, Balcali Hospital, Cukurova University, Adana, Turkey
| | - Ozlem Keskin
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Elif Arık
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Elif Soyak Aytekin
- Department of Pediatric Allergy and Immunology, Etlik City Hospital, Ankara, Turkey
| | - Mahmut Cesur
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Ercan Kucukosmanoglu
- Department of Pediatric Allergy and Immunology, Faculty of Medicine, Gaziantep University, Gaziantep, Turkey
| | - Mehmet Kılıc
- Division of Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, University of Firat, Elazığ, Turkey
| | - Mutlu Yuksek
- Department of Pediatric Immunology and Allergy, Faculty of Medicine, Zonguldak Bulent Ecevit University, Zonguldak, Turkey
| | - Zafer Bıcakcı
- Department of Pediatric Hematology, Faculty of Medicine, Ataturk University, Erzurum, Turkey
| | - Saliha Esenboga
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| | - Deniz Çagdaş Ayvaz
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University School of Medicine, Ankara, Turkey
- Section of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Asena Pınar Sefer
- Department of Pediatric Allergy and Immunology, Şanlıurfa Training and Research Hospital, Şanlıurfa, Turkey
| | - Sukrü Nail Guner
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Sevgi Keles
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Ismail Reisli
- Department of Pediatric Immunology and Allergy, Medicine Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Ugur Musabak
- Department of Immunology and Allergy, Baskent University School of Medicine, Ankara, Turkey
| | - Nazlı Deveci Demirbas
- Department of Pediatric Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sule Haskologlu
- Department of Pediatric Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sara Sebnem Kilic
- Division of Pediatric Immunology-Rheumatology, Bursa Uludag University Faculty of Medicine, Bursa, Turkey
- Translational Medicine, Bursa Uludag University, Bursa, Turkey
| | - Ayse Metin
- Department of Pediatric Allergy and Immunology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Figen Dogu
- Department of Pediatric Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Aydan Ikinciogulları
- Department of Pediatric Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Ilhan Tezcan
- Department of Pediatrics, Division of Pediatric Immunology, Hacettepe University School of Medicine, Ankara, Turkey
| |
Collapse
|
2
|
Fawzy MM, Nazmy MH, El-Sheikh AAK, Fathy M. Evolutionary preservation of CpG dinucleotides in RAG1 may elucidate the relatively high rate of methylation-mediated mutagenesis of RAG1 transposase. Immunol Res 2024; 72:438-449. [PMID: 38240953 PMCID: PMC11217092 DOI: 10.1007/s12026-023-09451-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 12/24/2023] [Indexed: 07/03/2024]
Abstract
Recombination-activating gene 1 (RAG1) is a vital player in V(D)J recombination, a fundamental process in primary B cell and T cell receptor diversification of the adaptive immune system. Current vertebrate RAG evolved from RAG transposon; however, it has been modified to play a crucial role in the adaptive system instead of being irreversibly silenced by CpG methylation. By interrogating a range of publicly available datasets, the current study investigated whether RAG1 has retained a disproportionate level of its original CpG dinucleotides compared to other genes, thereby rendering it more exposed to methylation-mediated mutation. Here, we show that 57.57% of RAG1 pathogenic mutations and 51.6% of RAG1 disease-causing mutations were associated with CpG methylation, a percentage that was significantly higher than that of its RAG2 cofactor alongside the whole genome. The CpG scores and densities for all RAG ancestors suggested that RAG transposon was CpG denser. The percentage of the ancestral CpG of RAG1 and RAG2 were 6% and 4.2%, respectively, with no preference towards CG containing codons. Furthermore, CpG loci of RAG1 in sperms were significantly higher methylated than that of RAG2. In conclusion, RAG1 has been exposed to CpG mediated methylation mutagenesis more than RAG2 and the whole genome, presumably due to its late entry to the genome later with an initially higher CpG content.
Collapse
Affiliation(s)
- Mariam M Fawzy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Maiiada H Nazmy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt
| | - Azza A K El-Sheikh
- Basic Health Sciences Department, College of Medicine, Princess Nourah bint Abdulrahman University, 11671, Riyadh, Saudi Arabia
| | - Moustafa Fathy
- Department of Biochemistry, Faculty of Pharmacy, Minia University, Minia, 61519, Egypt.
| |
Collapse
|
3
|
Ver Heul AM, Mack M, Zamidar L, Tamari M, Yang TL, Trier AM, Kim DH, Janzen-Meza H, Van Dyken SJ, Hsieh CS, Karo JM, Sun JC, Kim BS. RAG suppresses group 2 innate lymphoid cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.23.590767. [PMID: 38712036 PMCID: PMC11071423 DOI: 10.1101/2024.04.23.590767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2024]
Abstract
Antigen specificity is the central trait distinguishing adaptive from innate immune function. Assembly of antigen-specific T cell and B cell receptors occurs through V(D)J recombination mediated by the Recombinase Activating Gene endonucleases RAG1 and RAG2 (collectively called RAG). In the absence of RAG, mature T and B cells do not develop and thus RAG is critically associated with adaptive immune function. In addition to adaptive T helper 2 (Th2) cells, group 2 innate lymphoid cells (ILC2s) contribute to type 2 immune responses by producing cytokines like Interleukin-5 (IL-5) and IL-13. Although it has been reported that RAG expression modulates the function of innate natural killer (NK) cells, whether other innate immune cells such as ILC2s are affected by RAG remains unclear. We find that in RAG-deficient mice, ILC2 populations expand and produce increased IL-5 and IL-13 at steady state and contribute to increased inflammation in atopic dermatitis (AD)-like disease. Further, we show that RAG modulates ILC2 function in a cell-intrinsic manner independent of the absence or presence of adaptive T and B lymphocytes. Lastly, employing multiomic single cell analyses of RAG1 lineage-traced cells, we identify key transcriptional and epigenomic ILC2 functional programs that are suppressed by a history of RAG expression. Collectively, our data reveal a novel role for RAG in modulating innate type 2 immunity through suppression of ILC2s.
Collapse
Affiliation(s)
- Aaron M. Ver Heul
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Madison Mack
- Immunology & Inflammation Research Therapeutic Area, Sanofi, Cambridge, MA 02141, USA
| | - Lydia Zamidar
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Masato Tamari
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Ting-Lin Yang
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Anna M. Trier
- Division of Dermatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Do-Hyun Kim
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
- Department of Life Science, College of Natural Sciences, Hanyang University, Seoul 04763, Republic of Korea
| | - Hannah Janzen-Meza
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Steven J. Van Dyken
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63130, USA
| | - Chyi-Song Hsieh
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Jenny M. Karo
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
| | - Joseph C. Sun
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; Immunology and Microbial Pathogenesis Program, Graduate School of Medical Sciences, Weill Cornell Medical College, New York, NY 10065, USA
| | - Brian S. Kim
- Kimberly and Eric J. Waldman Department of Dermatology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Mark Lebwohl Center for Neuroinflammation and Sensation, Icahn School of Medicine at Mount Sinai, New York, NY 10019, USA
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
- Allen Discovery Center for Neuroimmune Interactions, Icahn School of Medicine at Mount Sinai 10019
| |
Collapse
|
4
|
Fayyaz H, Zaman A, Shabbir S, Khan ZK, Haider N, Saleem AF, Ahamad W, Ullah I. Mutational analysis in different genes underlying severe combined immunodeficiency in seven consanguineous Pakistani families. Mol Biol Rep 2024; 51:302. [PMID: 38355773 DOI: 10.1007/s11033-024-09222-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 01/06/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND Severe Combined Immunodeficiency (SCID) is an autosomal recessive inborn error of immunity (IEI) characterized by recurrent chest and gastrointestinal (GI) infections and in some cases associated with life-threatening disorders. METHODOLOGY AND RESULTS This current study aims to unwind the molecular etiology of SCID and also extended the patients' phenotype associated with identified particular variants. Herein, we present 06 disease-causing variants identified in 07 SCID-patients in three different SCID related genes. Whole Exome Sequencing (WES) followed by Sanger Sequencing was employed to explore genetic variations. The results included identification of two previously reported heterozygous variants in homozygous form for the first time in RAG1gene [(p.Arg410Gln);(p.Arg737His)], followed by a recurrent variant (p.Trp959*) in RAG1, a novel variant in IL2RG (p.Asp48Lfs*24), a recurrent variant in IL2RG (p.Gly271Glu) and a recurrent variant in DCLRE1C (p.Arg191*) gene. CONCLUSION To conclude, the immune-profiling and WES revealed two novel, two as homozygous state for the first time, and two recurrent disease causing variants contributing valuably to our existing knowledge of SCID.
Collapse
Affiliation(s)
- Hajra Fayyaz
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Atteaya Zaman
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
- Department of Biochemistry, Federal Medical & Dental College, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Sheeba Shabbir
- Forensic Medicine & Toxicology, School of Health Sciences, National University of Sciences and Technology, Islamabad, Pakistan
| | - Zara Khalid Khan
- Department of Biochemistry, HBS Medical & Dental College, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Nighat Haider
- Department of Pediatrics, Pakistan Institute of Medical Sciences Islamabad, Shaheed Zulfiqar Ali Bhutto Medical University, Islamabad, Pakistan
| | - Ali Faisal Saleem
- Department of Paediatrics & Paediatrics Infections Disease, Agha Khan University Hospital, Karachi, Pakistan
| | - Wasim Ahamad
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan
| | - Imran Ullah
- Department of Biochemistry, Faculty of Biological Sciences, Quaid-i-Azam University, Islamabad, 45320, Pakistan.
| |
Collapse
|
5
|
Karaatmaca B, Cagdas D, Esenboga S, Erman B, Tan C, Turul Ozgur T, Boztug K, van der Burg M, Sanal O, Tezcan I. Heterogeneity in RAG1 and RAG2 deficiency: 35 cases from a single-centre. Clin Exp Immunol 2024; 215:160-176. [PMID: 37724703 PMCID: PMC10847812 DOI: 10.1093/cei/uxad110] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 09/03/2023] [Accepted: 09/17/2023] [Indexed: 09/21/2023] Open
Abstract
Recombination activating genes (RAG)1 and RAG2 deficiency leads to combined T/B-cell deficiency with varying clinical presentations. This study aimed to define the clinical/laboratory spectrum of RAG1 and RAG2 deficiency. We retrospectively reviewed the clinical/laboratory data of 35 patients, grouped them as severe combined immunodeficiency (SCID), Omenn syndrome (OS), and delayed-onset combined immunodeficiency (CID) and reported nine novel mutations. The male/female ratio was 23/12. Median age of clinical manifestations was 1 months (mo) (0.5-2), 2 mo (1.25-5), and 14 mo (3.63-27), age at diagnosis was 4 mo (3-6), 4.5 mo (2.5-9.75), and 27 mo (14.5-70) in SCID (n = 25; 71.4%), OS (n = 5; 14.3%), and CID (n = 5; 14.3%) patients, respectively. Common clinical manifestations were recurrent sinopulmonary infections 82.9%, oral moniliasis 62.9%, diarrhea 51.4%, and eczema/dermatitis 42.9%. Autoimmune features were present in 31.4% of the patients; 80% were in CID patients. Lymphopenia was present in 92% of SCID, 80% of OS, and 80% of CID patients. All SCID and CID patients had low T (CD3, CD4, and CD8), low B, and increased NK cell numbers. Twenty-eight patients underwent hematopoietic stem cell transplantation (HSCT), whereas seven patients died before HSCT. Median age at HSCT was 7 mo (4-13.5). Survival differed in groups; maximum in SCID patients who had an HLA-matched family donor, minimum in OS. Totally 19 (54.3%) patients survived. Early molecular genetic studies will give both individualized therapy options, and a survival advantage because of timely diagnosis and treatment. Further improvement in therapeutic outcomes will be possible if clinicians gain time for HSCT.
Collapse
Affiliation(s)
- Betul Karaatmaca
- Hacettepe University School of Medicine, Department of Pediatrics, Division of Pediatric Immunology, Ankara, Turkey
- Department of Pediatric Allergy and Immunology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Deniz Cagdas
- Hacettepe University School of Medicine, Department of Pediatrics, Division of Pediatric Immunology, Ankara, Turkey
- Section of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Saliha Esenboga
- Hacettepe University School of Medicine, Department of Pediatrics, Division of Pediatric Immunology, Ankara, Turkey
| | - Baran Erman
- Section of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Cagman Tan
- Section of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| | - Tuba Turul Ozgur
- Hacettepe University School of Medicine, Department of Pediatrics, Division of Pediatric Immunology, Ankara, Turkey
| | - Kaan Boztug
- St. Anna Children's Cancer Research Institute (CCRI), Vienna, Austria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
- Medical University of Vienna, Department of Pediatrics and Adolescent Medicine, Vienna, Austria
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria
- St. Anna Children's Hospital, Vienna, Austria
| | - Mirjam van der Burg
- Department of Pediatrics, Laboratory for Pediatric Immunology, Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, The Netherlands
| | - Ozden Sanal
- Hacettepe University School of Medicine, Department of Pediatrics, Division of Pediatric Immunology, Ankara, Turkey
| | - Ilhan Tezcan
- Hacettepe University School of Medicine, Department of Pediatrics, Division of Pediatric Immunology, Ankara, Turkey
- Section of Pediatric Immunology, Institute of Child Health, Hacettepe University, Ankara, Turkey
| |
Collapse
|
6
|
Peng Y, Chen Y, Wang Y, Wang W, Qiao S, Lan J, Wang M. Dysbiosis and primary B-cell immunodeficiencies: current knowledge and future perspective. Immunol Res 2023; 71:528-536. [PMID: 36933165 DOI: 10.1007/s12026-023-09365-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 01/27/2023] [Indexed: 03/19/2023]
Abstract
According to Elie Metchnikoff, an originator of modern immunology, several pivotal functions for disease and health are provided by indigenous microbiota. Nonetheless, important mechanistic insights have been elucidated more recently, owing to the growing availability of DNA sequencing technology. There are 10 to 100 trillion symbiotic microbes (such as viruses, bacteria, and yeast) in each human gut microbiota. Both locally and systemically, the gut microbiota has been demonstrated to impact immune homeostasis. Primary B-cell immunodeficiencies (PBIDs) are a group of primary immunodeficiency diseases (PIDs) referring to the dysregulated antibody production due to either intrinsic genetic defects or failures in functions of B cells. Recent studies have found that PBIDs cause disruptions in the gut's typical homeostatic systems, resulting in inadequate immune surveillance in the gastrointestinal (GI) tract, which is linked to increased dysbiosis, which is characterized by a disruption in the microbial homeostasis. This study aimed to review the published articles in this field to provide a comprehensive view of the existing knowledge about the crosstalk between the gut microbiome and PBID, the factors shaping the gut microbiota in PBID, as well as the potential clinical approaches for restoring a normal microbial community.
Collapse
Affiliation(s)
- Ye Peng
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Zhejiang, 310014, Hangzhou, China
| | - Yirui Chen
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Zhejiang, 310014, Hangzhou, China
| | - Yanzhong Wang
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Zhejiang, Hangzhou, China
| | - Wensong Wang
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Zhejiang, 310014, Hangzhou, China
| | - Sai Qiao
- Department of Clinical Laboratory, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 3 East Qingchun Road, Zhejiang, Hangzhou, China
| | - Jianping Lan
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Zhejiang, 310014, Hangzhou, China.
| | - Manling Wang
- Cancer Center, Department of Hematology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 58 Shangtang Road, Zhejiang, 310014, Hangzhou, China.
| |
Collapse
|
7
|
Braams M, Pike-Overzet K, Staal FJT. The recombinase activating genes: architects of immune diversity during lymphocyte development. Front Immunol 2023; 14:1210818. [PMID: 37497222 PMCID: PMC10367010 DOI: 10.3389/fimmu.2023.1210818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2023] [Accepted: 06/19/2023] [Indexed: 07/28/2023] Open
Abstract
The mature lymphocyte population of a healthy individual has the remarkable ability to recognise an immense variety of antigens. Instead of encoding a unique gene for each potential antigen receptor, evolution has used gene rearrangements, also known as variable, diversity, and joining gene segment (V(D)J) recombination. This process is critical for lymphocyte development and relies on recombination-activating genes-1 (RAG1) and RAG2, here collectively referred to as RAG. RAG serves as powerful genome editing tools for lymphocytes and is strictly regulated to prevent dysregulation. However, in the case of dysregulation, RAG has been implicated in cases of cancer, autoimmunity and severe combined immunodeficiency (SCID). This review examines functional protein domains and motifs of RAG, describes advances in our understanding of the function and (dys)regulation of RAG, discuss new therapeutic options, such as gene therapy, for RAG deficiencies, and explore in vitro and in vivo methods for determining RAG activity and target specificity.
Collapse
Affiliation(s)
- Merijn Braams
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Karin Pike-Overzet
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
| | - Frank J. T. Staal
- Department of Immunology, Leiden University Medical Centre, Leiden, Netherlands
- Novo Nordisk Foundation Centre for Stem Cell Medicine (reNEW), Leiden University Medical Centre, Leiden, Netherlands
- Department of Paediatrics, Leiden University Medical Centre, Leiden, Netherlands
| |
Collapse
|
8
|
Min Q, Csomos K, Li Y, Dong L, Hu Z, Meng X, Yu M, Walter JE, Wang JY. B cell abnormalities and autoantibody production in patients with partial RAG deficiency. Front Immunol 2023; 14:1155380. [PMID: 37475856 PMCID: PMC10354446 DOI: 10.3389/fimmu.2023.1155380] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 06/15/2023] [Indexed: 07/22/2023] Open
Abstract
Mutations in the recombination activating gene 1 (RAG1) and RAG2 in humans are associated with a broad spectrum of clinical phenotypes, from severe combined immunodeficiency to immune dysregulation. Partial (hypomorphic) RAG deficiency (pRD) in particular, frequently leads to hyperinflammation and autoimmunity, with several underlying intrinsic and extrinsic mechanisms causing a break in tolerance centrally and peripherally during T and B cell development. However, the relative contributions of these processes to immune dysregulation remain unclear. In this review, we specifically focus on the recently described tolerance break and B cell abnormalities, as well as consequent molecular and cellular mechanisms of autoantibody production in patients with pRD.
Collapse
Affiliation(s)
- Qing Min
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Krisztian Csomos
- Division of Pediatric Allergy/Immunology, University of South Florida at Johns Hopkins All Children's Hospital, St. Petersburg, FL, United States
| | - Yaxuan Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Lulu Dong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ziying Hu
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Meiping Yu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - 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/Immunology, Massachusetts General Hospital for Children, Boston, MA, United States
| | - Ji-Yang Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
- Shanghai Huashen Institute of Microbes and Infections, Shanghai, China
| |
Collapse
|
9
|
Mou W, Yang Z, Wang X, Hei M, Wang Y, Gui J. Immunological assessment of a patient with Omenn syndrome resulting from compound heterozygous mutations in the RAG1 gene. Immunogenetics 2023:10.1007/s00251-023-01309-5. [PMID: 37269334 DOI: 10.1007/s00251-023-01309-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 05/17/2023] [Indexed: 06/05/2023]
Abstract
The recombination activating gene 1 (RAG1) is essential for V(D)J recombination during T- and B-cell development. In this study, we presented a case study of a 41-day-old female infant who exhibited symptoms of generalized erythroderma, lymphadenopathy, hepatosplenomegaly, and recurrent infections including suppurative meningitis and septicemia. The patient showed a T+B-NK+ immunophenotype. We observed an impaired thymic output, as indicated by reduced levels of naive T cells and sjTRECs, coupled with a restricted TCR repertoire. Additionally, T-cell CFSE proliferation was impaired, indicating a suboptimal T-cell response. Notably, our data further revealed that T cells were in an activated state. Genetic analysis revealed a previously reported compound heterozygous mutation (c. 1186C > T, p. R396C; c. 1210C > T, p. R404W) in the RAG1 gene. Structural analysis of RAG1 suggested that the R396C mutation might lead to the loss of hydrogen bonds with neighboring amino acids. These findings contribute to our understanding of RAG1 deficiency and may have implications for the development of novel therapies for patients with this condition.
Collapse
Affiliation(s)
- Wenjun Mou
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Zixin Yang
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Xiaojiao Wang
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China
| | - Mingyan Hei
- Department of Neonatology, Neonatal Center, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| | - Yajuan Wang
- Department of Neonatology, Children's Hospital, Capital Institute of Pediatrics, Beijing, 100020, China.
| | - Jingang Gui
- Laboratory of Tumor Immunology, Beijing Pediatric Research Institute, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, 100045, China.
| |
Collapse
|
10
|
Yonkof JR, Basu A, Redmond MT, Dobbs AK, Perelygina L, Notarangelo LD, Abraham RS, Rangarajan HG. Refractory, fatal autoimmune hemolytic anemia due to ineffective thymic-derived T-cell reconstitution following allogeneic hematopoietic cell transplantation for hypomorphic RAG1 deficiency. Pediatr Blood Cancer 2023; 70:e30183. [PMID: 36583469 PMCID: PMC10038854 DOI: 10.1002/pbc.30183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Accepted: 11/23/2022] [Indexed: 12/31/2022]
Affiliation(s)
- Jennifer R Yonkof
- Department of Pediatrics, Division of Allergy & Immunology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Amrita Basu
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Margaret T Redmond
- Department of Pediatrics, Division of Allergy & Immunology, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Adam Kerry Dobbs
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, Bethesda, Maryland, USA
| | - Ludmila Perelygina
- Division of Viral Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Health, Bethesda, Maryland, USA
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio, USA
| | - Hemalatha G Rangarajan
- Department of Pediatrics, Division of Hematology, Oncology, Blood and Marrow Transplant, Nationwide Children's Hospital, Columbus, Ohio, USA
| |
Collapse
|
11
|
Castiello MC, Brandas C, Capo V, Villa A. HyperIgE in hypomorphic recombination-activating gene defects. Curr Opin Immunol 2023; 80:102279. [PMID: 36529093 DOI: 10.1016/j.coi.2022.102279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 11/28/2022] [Indexed: 12/23/2022]
Abstract
Increased immunogloblulin-E (IgE) levels associated with eosinophilia represent a common finding observed in Omenn syndrome, a severe immunodeficiency caused by decreased V(D)J recombination, leading to restricted T- and B-cell receptor repertoire. V(D)J recombination is initiated by the lymphoid-restricted recombination-activating gene (RAG) recombinases. The lack of RAG proteins causes a block in lymphocyte differentiation, resulting in T-B- severe combined immunodeficiency. Conversely, hypomorphic mutations allow the generation of few T and B cells, leading to a spectrum of immunological phenotypes, in which immunodeficiency associates to inflammation, immune dysregulation, and autoimmunity. Elevated IgE levels are frequently observed in hypomorphic RAG patients. Here, we describe the role of RAG genes in lymphocyte differentiation and maintenance of immune tolerance.
Collapse
Affiliation(s)
- Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan, Italy
| | - Chiara Brandas
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Translational and Molecular Medicine (DIMET), University of Milano-Bicocca, Monza, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy; Institute of Genetic and Biomedical Research, Milan Unit, National Research Council, Milan, Italy.
| |
Collapse
|
12
|
Jardine L, Schim van der Loeff I, Haq IJ, Sproat TDR. Gestational Development of the Human Immune System. Immunol Allergy Clin North Am 2023; 43:1-15. [PMID: 36410996 DOI: 10.1016/j.iac.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Building an immune system is a monumental task critical to the survival of the fetus and newborn. A functional fetal immune system must complement the maternal immune system in handling in utero infection; abstain from damaging non-self-reactions that would compromise the materno-fetal interface; mobilize in response to infection and equip mucosal tissues for pathogen exposure at birth. There is growing appreciation that immune cells also have noncanonical roles in development and specifically may contribute to tissue morphogenesis. In this review we detail how hematopoietic and lymphoid organs jointly establish cellular constituents of the immune system; how these constituents are organized in 2 mucosal sites-gut and lung-where early life immune function has long-term consequences for health; and how exemplar diseases of prematurity and inborn errors of immunity reveal dominant pathways in prenatal immunity.
Collapse
Affiliation(s)
- Laura Jardine
- Biosciences Institute, Newcastle University, Faculty of Medical Sciences, Newcastle Upon Tyne NE2 4HH, United Kingdom; Haematology Department, Freeman Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom.
| | - Ina Schim van der Loeff
- Translational and Clinical Research Institute, Newcastle University, Faculty of Medical Sciences, Newcastle Upon Tyne NE2 4HH, United Kingdom
| | - Iram J Haq
- Translational and Clinical Research Institute, Newcastle University, Faculty of Medical Sciences, Newcastle Upon Tyne NE2 4HH, United Kingdom; Department of Paediatric Respiratory Medicine, Great North Children's Hospital, Newcastle Hospitals NHS Foundation Trust, Newcastle Upon Tyne, United Kingdom
| | - Thomas D R Sproat
- Neonatal Unit, Royal Victoria Infirmary, Newcastle Hospitals NHS Foundation Trust, Richardson Road, Newcastle Upon Tyne NE1 4LP, United Kingdom
| |
Collapse
|
13
|
Sjøgren T, Bratland E, Røyrvik EC, Grytaas MA, Benneche A, Knappskog PM, Kämpe O, Oftedal BE, Husebye ES, Wolff ASB. Screening patients with autoimmune endocrine disorders for cytokine autoantibodies reveals monogenic immune deficiencies. J Autoimmun 2022; 133:102917. [PMID: 36191466 DOI: 10.1016/j.jaut.2022.102917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/09/2022] [Accepted: 09/12/2022] [Indexed: 12/13/2022]
Abstract
BACKGROUND Autoantibodies against type I interferons (IFN) alpha (α) and omega (ω), and interleukins (IL) 17 and 22 are a hallmark of autoimmune polyendocrine syndrome type 1 (APS-1), caused by mutations in the autoimmune regulator (AIRE) gene. Such antibodies are also seen in a number of monogenic immunodeficiencies. OBJECTIVES To determine whether screening for cytokine autoantibodies (anti-IFN-ω and anti-IL22) can be used to identify patients with monogenic immune disorders. METHODS A novel ELISA assay was employed to measure IL22 autoantibodies in 675 patients with autoimmune primary adrenal insufficiency (PAI) and a radio immune assay (RIA) was used to measure autoantibodies against IFN-ω in 1778 patients with a variety of endocrine diseases, mostly of autoimmune aetiology. Positive cases were sequenced for all coding exons of the AIRE gene. If no AIRE mutations were found, we applied next generation sequencing (NGS) to search for mutations in immune related genes. RESULTS We identified 29 patients with autoantibodies against IFN-ω and/or IL22. Of these, four new APS-1 cases with disease-causing variants in AIRE were found. In addition, we identified two patients with pathogenic heterozygous variants in CTLA4 and NFKB2, respectively. Nine rare variants in other immune genes were identified in six patients, although further studies are needed to determine their disease-causing potential. CONCLUSION Screening of cytokine autoantibodies can efficiently identify patients with previously unknown monogenic and possible oligogenic causes of autoimmune and immune deficiency diseases. This information is crucial for providing personalised treatment and follow-up of patients and their relatives.
Collapse
Affiliation(s)
- Thea Sjøgren
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Eirik Bratland
- Department of Clinical Science, University of Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Ellen C Røyrvik
- Department of Clinical Science, University of Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Marianne Aa Grytaas
- Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Andreas Benneche
- Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Per M Knappskog
- Department of Clinical Science, University of Bergen, Norway; Department of Medical Genetics, Haukeland University Hospital, Bergen, Norway
| | - Olle Kämpe
- KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway; Department of Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Bergithe E Oftedal
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway
| | - Eystein S Husebye
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway.
| | - Anette S B Wolff
- Department of Clinical Science, University of Bergen, Norway; Department of Medicine, Haukeland University Hospital, Bergen, Norway; KG Jebsen Center for Autoimmune Diseases, University of Bergen, Norway.
| |
Collapse
|
14
|
Pan C, Zhao A, Li M. Atopic Dermatitis-like Genodermatosis: Disease Diagnosis and Management. Diagnostics (Basel) 2022; 12:diagnostics12092177. [PMID: 36140582 PMCID: PMC9498295 DOI: 10.3390/diagnostics12092177] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/23/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Eczema is a classical characteristic not only in atopic dermatitis but also in various genodermatosis. Patients suffering from primary immunodeficiency diseases such as hyper-immunoglobulin E syndromes, Wiskott-Aldrich syndrome, immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, STAT5B deficiency, Omenn syndrome, atypical complete DiGeorge syndrome; metabolic disorders such as acrodermatitis enteropathy, multiple carboxylase deficiency, prolidase deficiency; and other rare syndromes like severe dermatitis, multiple allergies and metabolic wasting syndrome, Netherton syndrome, and peeling skin syndrome frequently perform with eczema-like lesions. These genodermatosis may be misguided in the context of eczematous phenotype. Misdiagnosis of severe disorders unavoidably affects appropriate treatment and leads to irreversible outcomes for patients, which underlines the importance of molecular diagnosis and genetic analysis. Here we conclude clinical manifestations, molecular mechanism, diagnosis and management of several eczema-related genodermatosis and provide accessible advice to physicians.
Collapse
Affiliation(s)
- Chaolan Pan
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Anqi Zhao
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Ming Li
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Department of Dermatology, The Children’s Hospital of Fudan University, Shanghai 200092, China
- Correspondence: ; Tel.: +86-2125078571
| |
Collapse
|
15
|
Wang Y, Abolhassani H, Hammarström L, Pan-Hammarström Q. SARS-CoV-2 infection in patients with inborn errors of immunity due to DNA repair defects. Acta Biochim Biophys Sin (Shanghai) 2022; 54:836-846. [PMID: 35713311 PMCID: PMC9827799 DOI: 10.3724/abbs.2022071] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Clinical information on severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in patients with inborn errors of immunity (IEI) during the current Coronavirus disease 2019 (COVID-19) pandemic is still limited. Proper DNA repair machinery is required for the development of the adaptive immune system, which provides specific and long-term protection against SARS-CoV-2. This review highlights the impact of SARS-CoV-2 infections on IEI patients with DNA repair disorders and summarizes susceptibility risk factors, pathogenic mechanisms, clinical manifestations and management strategies of COVID-19 in this special patient population.
Collapse
|
16
|
Padron GT, Hernandez-Trujillo VP. Autoimmunity in Primary Immunodeficiencies (PID). Clin Rev Allergy Immunol 2022:10.1007/s12016-022-08942-0. [PMID: 35648371 DOI: 10.1007/s12016-022-08942-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/04/2022] [Indexed: 11/25/2022]
Abstract
Primary immunodeficiency (PID) may impact any component of the immune system. The number of PID and immune dysregulation disorders is growing steadily with advancing genetic detection methods. These expansive recognition methods have changed the way we characterize PID. While PID were once characterized by their susceptibility to infection, the increase in genetic analysis has elucidated the intertwined relationship between PID and non-infectious manifestations including autoimmunity. The defects permitting opportunistic infections to take hold may also lead the way to the development of autoimmune disease. In some cases, it is the non-infectious complications that may be the presenting sign of PID autoimmune diseases, such as autoimmune cytopenia, enteropathy, endocrinopathies, and arthritis among others, have been reported in PID. While autoimmunity may occur with any PID, this review will look at certain immunodeficiencies most often associated with autoimmunity, as well as their diagnosis and management strategies.
Collapse
Affiliation(s)
- Grace T Padron
- Nicklaus Children's Hospital, Miami, FL, USA.
- Allergy and Immunology Care Center of South Florida, Miami Lakes, FL, USA.
| | - Vivian P Hernandez-Trujillo
- Nicklaus Children's Hospital, Miami, FL, USA
- Allergy and Immunology Care Center of South Florida, Miami Lakes, FL, USA
| |
Collapse
|
17
|
Nelson RW, Geha RS, McDonald DR. Inborn Errors of the Immune System Associated With Atopy. Front Immunol 2022; 13:860821. [PMID: 35572516 PMCID: PMC9094424 DOI: 10.3389/fimmu.2022.860821] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Atopic disorders, including atopic dermatitis, food and environmental allergies, and asthma, are increasingly prevalent diseases. Atopic disorders are often associated with eosinophilia, driven by T helper type 2 (Th2) immune responses, and triggered by disrupted barrier function leading to abnormal immune priming in a susceptible host. Immune deficiencies, in contrast, occur with a significantly lower incidence, but are associated with greater morbidity and mortality. A subset of atopic disorders with eosinophilia and elevated IgE are associated with monogenic inborn errors of immunity (IEI). In this review, we discuss current knowledge of IEI that are associated with atopy and the lessons these immunologic disorders provide regarding the fundamental mechanisms that regulate type 2 immunity in humans. We also discuss further mechanistic insights provided by animal models.
Collapse
Affiliation(s)
- Ryan W Nelson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Douglas R McDonald
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
18
|
Barreiros LA, Sousa JL, Geier C, Leiss-Piller A, Kanegae MPP, França TT, Boisson B, Lima AM, Costa-Carvalho BT, Aranda CS, de Moraes-Pinto MI, Segundo GRS, Ferreira JFS, Tavares FS, Guimarães FATDM, Toledo EC, da Matta Ain AC, Moreira IF, Soldatelli G, Grumach AS, de Barros Dorna M, Weber CW, Di Gesu RSW, Dantas VM, Fernandes FR, Torgerson TR, Ochs HD, Bustamante J, Walter JE, Condino-Neto A. SCID and Other Inborn Errors of Immunity with Low TRECs - the Brazilian Experience. J Clin Immunol 2022; 42:1171-1192. [PMID: 35503492 DOI: 10.1007/s10875-022-01275-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 04/17/2022] [Indexed: 11/26/2022]
Abstract
Severe combined immunodeficiency, SCID, is a pediatric emergency that represents the most critical group of inborn errors of immunity (IEI). Affected infants present with early onset life-threatening infections due to absent or non-functional T cells. Without early diagnosis and curative treatment, most die in early infancy. As most affected infants appear healthy at birth, newborn screening (NBS) is essential to identify and treat patients before the onset of symptoms. Here, we report 47 Brazilian patients investigated between 2009 and 2020 for SCID due to either a positive family history and/or clinical impression and low TRECs. Based on clinical presentation, laboratory finding, and genetic information, 24 patients were diagnosed as typical SCID, 14 as leaky SCID, and 6 as Omenn syndrome; 2 patients had non-SCID IEI, and 1 remained undefined. Disease onset median age was 2 months, but at the time of diagnosis and treatment, median ages were 6.5 and 11.5 months, respectively, revealing considerable delay which affected negatively treatment success. While overall survival was 51.1%, only 66.7% (30/45) lived long enough to undergo hematopoietic stem-cell transplantation, which was successful in 70% of cases. Forty-three of 47 (91.5%) patients underwent genetic testing, with a 65.1% success rate. Even though our patients did not come from the NBS programs, the diagnosis of SCID improved in Brazil during the pilot programs, likely due to improved medical education. However, we estimate that at least 80% of SCID cases are still missed. NBS-SCID started to be universally implemented in the city of São Paulo in May 2021, and it is our hope that other cities will follow, leading to early diagnosis and higher survival of SCID patients in Brazil.
Collapse
Affiliation(s)
- Lucila Akune Barreiros
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Jusley Lira Sousa
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | | | | | - Marilia Pylles Patto Kanegae
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Tábata Takahashi França
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
| | | | | | | | | | | | | | | | | | | | - Ana Carolina da Matta Ain
- Departamento de Pediatria E Imunologia, Hospital Universitário de Taubaté, Universidade de Taubaté, Taubate, SP, Brazil
| | | | - Gustavo Soldatelli
- Hospital das Clínicas, Universidade Federal de Santa Caratina, Florianopolis, SC, Brazil
| | | | - Mayra de Barros Dorna
- Instituto da Criança, Hospital das Clínicas, Faculdade de Medicina da Universidade de São Paulo, Sao Paulo, SP, Brazil
| | | | | | - Vera Maria Dantas
- Departamento de Pediatria, Universidade Federal Do Rio Grande Do Norte, Natal, RN, Brazil
| | | | | | - Hans Dietrich Ochs
- Department of Pediatrics, University of Washington School of Medicine and Seattle Children's Research Institute, Seattle, USA
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, University of Paris, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Jolan Eszter Walter
- University of South Florida at Johns Hopkins All Children's Hospital, Saint Petersburg, FL, USA
- Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Antonio Condino-Neto
- Laboratory of Human Immunology, Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, 1730, Av. Professor Lineu Prestes, Sao Paulo, SP, 05508-000, Brazil.
- Immunogenic Laboratories Inc, Sao Paulo, SP, Brazil.
| |
Collapse
|
19
|
Cifaldi C, Rivalta B, Amodio D, Mattia A, Pacillo L, Di Cesare S, Chiriaco M, Ursu GM, Cotugno N, Giancotta C, Manno EC, Santilli V, Zangari P, Federica G, Palumbo G, Merli P, Palma P, Rossi P, Di Matteo G, Locatelli F, Finocchi A, Cancrini C. Clinical, Immunological, and Molecular Variability of RAG Deficiency: A Retrospective Analysis of 22 RAG Patients. J Clin Immunol 2022; 42:130-145. [PMID: 34664192 PMCID: PMC8821501 DOI: 10.1007/s10875-021-01130-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 08/29/2021] [Indexed: 11/05/2022]
Abstract
PURPOSE We described clinical, immunological, and molecular characterization within a cohort of 22 RAG patients focused on the possible correlation between clinical and genetic data. METHODS Immunological and genetic features were investigated by multiparametric flow cytometry and by Sanger or next generation sequencing (NGS) as appropriate. RESULTS Patients represented a broad spectrum of RAG deficiencies: SCID, OS, LS/AS, and CID. Three novel mutations in RAG1 gene and one in RAG2 were reported. The primary symptom at presentation was infections (81.8%). Infections and autoimmunity occurred together in the majority of cases (63.6%). Fifteen out of 22 (68.2%) patients presented autoimmune or inflammatory manifestations. Five patients experienced severe autoimmune cytopenia refractory to different lines of therapy. Total lymphocytes count was reduced or almost lacking in SCID group and higher in OS patients. B lymphocytes were variably detected in LS/AS and CID groups. Eighteen patients underwent HSCT permitting definitive control of autoimmune/hyperinflammatory manifestations in twelve of them (80%). CONCLUSION We reinforce the notion that different clinical phenotype can be found in patients with identical mutations even within the same family. Infections may influence genotype-phenotype correlation and function as trigger for immune dysregulation or autoimmune manifestations. Severe and early autoimmune refractory cytopenia is frequent and could be the first symptom of onset. Prompt recognition of RAG deficiency in patients with early onset of autoimmune/hyperinflammatory manifestations could contribute to the choice of a timely and specific treatment preventing the onset of other complications.
Collapse
Affiliation(s)
- Cristina Cifaldi
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy.
| | - Beatrice Rivalta
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Donato Amodio
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Algeri Mattia
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Scientific Institute for Research and Healthcare, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Lucia Pacillo
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Silvia Di Cesare
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Maria Chiriaco
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Giorgiana Madalina Ursu
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Nicola Cotugno
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Carmela Giancotta
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Emma C Manno
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Veronica Santilli
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Paola Zangari
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Galaverna Federica
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Scientific Institute for Research and Healthcare, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Giuseppe Palumbo
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Scientific Institute for Research and Healthcare, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Pietro Merli
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Scientific Institute for Research and Healthcare, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Paolo Palma
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Paolo Rossi
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
- Academic Department of Pediatrics (DPUO), Research Unit of Clinical Immunology and Vaccinology, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
| | - Gigliola Di Matteo
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hemato-Oncology and Cell and Gene Therapy, Scientific Institute for Research and Healthcare, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
| | - Andrea Finocchi
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy
| | - Caterina Cancrini
- Academic Department of Pediatrics (DPUO), Immune and Infectious Diseases Division, Research Unit of Primary Immunodeficiencies, Bambino Gesù Children's Hospital, IRCCS, 00165, Rome, Italy.
- Chair of Pediatrics, Department of Systems Medicine, University of Rome "Tor Vergata", via Montpellier, 1, 00133, Rome, Italy.
| |
Collapse
|
20
|
Min Q, Meng X, Zhou Q, Wang Y, Li Y, Lai N, Xiong E, Wang W, Yasuda S, Yu M, Zhang H, Sun J, Wang X, Wang JY. RAG1 splicing mutation causes enhanced B cell differentiation and autoantibody production. JCI Insight 2021; 6:148887. [PMID: 34622798 PMCID: PMC8525647 DOI: 10.1172/jci.insight.148887] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Accepted: 08/26/2021] [Indexed: 11/30/2022] Open
Abstract
Hypomorphic RAG1 or RAG2 mutations cause primary immunodeficiencies and can lead to autoimmunity, but the underlying mechanisms are elusive. We report here a patient carrying a c.116+2T>G homozygous splice site mutation in the first intron of RAG1, which led to aberrant splicing and greatly reduced RAG1 protein expression. B cell development was blocked at both the pro-B to pre-B transition and the pre-B to immature B cell differentiation step. The patient B cells had reduced B cell receptor repertoire diversity and decreased complementarity determining region 3 lengths. Despite B cell lymphopenia, the patient had abundant plasma cells in the BM and produced large quantities of IgM and IgG Abs, including autoantibodies. The proportion of naive B cells was reduced while the frequency of IgD–CD27– double-negative (DN) B cells, which quickly differentiated into Ab-secreting plasma cells upon stimulation, was greatly increased. Immune phenotype analysis of 52 patients with primary immunodeficiency revealed a strong association of the increased proportion of DN B and memory B cells with decreased number and proportion of naive B cells. These results suggest that the lymphopenic environment triggered naive B cell differentiation into DN B and memory B cells, leading to increased Ab production.
Collapse
Affiliation(s)
- Qing Min
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Xin Meng
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Qinhua Zhou
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Ying Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Yaxuan Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Nannan Lai
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Ermeng Xiong
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China
| | - Wenjie Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Shoya Yasuda
- School of Computing, Tokyo Institute of Technology, Yokohama, Japan
| | - Meiping Yu
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Hai Zhang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Jinqiao Sun
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Xiaochuan Wang
- Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China
| | - Ji-Yang Wang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, China.,Department of Clinical Immunology, Children's Hospital of Fudan University, National Children's Medical Center, Shanghai, China.,Department of Microbiology and Immunology, College of Basic Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
21
|
Wu X, Sun M, Yang Z, Lu C, Wang Q, Wang H, Deng C, Liu Y, Yang Y. The Roles of CCR9/CCL25 in Inflammation and Inflammation-Associated Diseases. Front Cell Dev Biol 2021; 9:686548. [PMID: 34490243 PMCID: PMC8416662 DOI: 10.3389/fcell.2021.686548] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 07/23/2021] [Indexed: 12/15/2022] Open
Abstract
Chemokine is a structure-related protein with a relatively small molecular weight, which can target cells to chemotaxis and promote inflammatory response. Inflammation plays an important role in aging. C-C chemokine receptor 9 (CCR9) and its ligand C-C chemokine ligand 25 (CCL25) are involved in the regulating the occurrence and development of various diseases, which has become a research hotspot. Early research analysis of CCR9-deficient mouse models also confirmed various physiological functions of this chemokine in inflammatory responses. Moreover, CCR9/CCL25 has been shown to play an important role in a variety of inflammation-related diseases, such as cardiovascular disease (CVD), rheumatoid arthritis, hepatitis, inflammatory bowel disease, asthma, etc. Therefore, the purpose of this review gives an overview of the recent advances in understanding the roles of CCR9/CCL25 in inflammation and inflammation-associated diseases, which will contribute to the design of future experimental studies on the potential of CCR9/CCL25 and advance the research of CCR9/CCL25 as pharmacological inflammatory targets.
Collapse
Affiliation(s)
- Xue Wu
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Meng Sun
- Department of Cardiology, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Zhi Yang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Chenxi Lu
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| | - Qiang Wang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
| | - Haiying Wang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
| | - Chao Deng
- Department of Cardiovascular Surgery, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yonglin Liu
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
| | - Yang Yang
- Department of Paediatrics, Shenmu Hospital, School of Life Sciences and Medicine, Northwest University, Shenmu, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Faculty of Life Sciences, Northwest University, Xi’an, China
| |
Collapse
|
22
|
Bosticardo M, Pala F, Notarangelo LD. RAG deficiencies: Recent advances in disease pathogenesis and novel therapeutic approaches. Eur J Immunol 2021; 51:1028-1038. [PMID: 33682138 PMCID: PMC8325549 DOI: 10.1002/eji.202048880] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/13/2021] [Accepted: 03/03/2021] [Indexed: 12/26/2022]
Abstract
The RAG1 and RAG2 proteins initiate the process of V(D)J recombination and therefore play an essential role in adaptive immunity. While null mutations in the RAG genes cause severe combined immune deficiency with lack of T and B cells (T- B- SCID) and susceptibility to life-threatening, early-onset infections, studies in humans and mice have demonstrated that hypomorphic RAG mutations are associated with defects of central and peripheral tolerance resulting in immune dysregulation. In this review, we provide an overview of the extended spectrum of RAG deficiencies and their associated clinical and immunological phenotypes in humans. We discuss recent advances in the mechanisms that control RAG expression and function, the effects of perturbed RAG activity on lymphoid development and immune homeostasis, and propose novel approaches to correct this group of disorders.
Collapse
Affiliation(s)
- Marita Bosticardo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Francesca Pala
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
23
|
Benhsaien I, Essadssi S, Elkhattabi L, Bakhchane A, Abdelghaffar H, Bousfiha AA, Badou A, Barakat A. Omenn syndrome caused by a novel homozygous mutation in recombination activating gene 1. Immunobiology 2021; 226:152090. [PMID: 33964732 DOI: 10.1016/j.imbio.2021.152090] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 02/12/2021] [Accepted: 03/29/2021] [Indexed: 11/20/2022]
Abstract
Omenn syndrome (OS) is a type of severe combined immunodeficiency (SCID) that is distinguished by, lymphadenopathy, hepatosplenomegaly, erythroderma, alopecia with normal to elevated T-cell counts, eosinophilia, and elevated serum IgE levels. Recombination activation gene (RAG) 1 or RAG2 mutations that result in partial V(D)J recombination activity are known to be the main cause of OS. Other genes (DCLRE1C, LIG4, IL7RA, common gamma chain, ADA, RMRP, and CHD7) have also been linked to OS, although with low frequency. Here, we report a two-month-old Moroccan girl from consanguineous marriage with chronic diarrhea, recurrent and opportunistic infections, failure to thrive, desquamative erythroderma, hepatosplenomegaly, and axillary lymphadenitis. The immunological assessment showed normal lymphocyte and NK cell counts but an absence of B cells, agammaglobulinemia contrasting with a high level of IgE. On the other hand, Sanger sequencing of RAG1 and RAG2 exon 2 regions revealed a new homozygous deleterious mutation in the RAG1 gene. This c.1184C > T mutation caused a change from Proline to Leucine at position 395 of the protein, leading to a partial loss of function. Early and rapid diagnosis of the disease may facilitate urgent life-saving treatment.
Collapse
Affiliation(s)
- Ibtihal Benhsaien
- Clinical Immunology Unit, Infectious Disease Department, Children Hospital, Ibn Rochd University Hospital, Casablanca, Morocco; Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco; Clinical Immunology, Autoimmunity and Inflammation Laboratory (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Soukaina Essadssi
- Laboratory of Genomics and Human Genetics,Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360 Casablanca, Morocco; Laboratory of Biosciences, Integrated and Molecular Functional Exploration (LBEFIM), Faculty of Science and Technology of Mohammedia, Hassan II University of Casablanca, Casablanca, Morocco
| | - Lamiae Elkhattabi
- Laboratory of Genomics and Human Genetics,Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360 Casablanca, Morocco
| | - Amina Bakhchane
- Laboratory of Genomics and Human Genetics,Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360 Casablanca, Morocco
| | - Houria Abdelghaffar
- Laboratory of Biosciences, Integrated and Molecular Functional Exploration (LBEFIM), Faculty of Science and Technology of Mohammedia, Hassan II University of Casablanca, Casablanca, Morocco
| | - Ahmed Aziz Bousfiha
- Clinical Immunology Unit, Infectious Disease Department, Children Hospital, Ibn Rochd University Hospital, Casablanca, Morocco; Clinical Immunology, Autoimmunity and Inflammation Laboratory (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdallah Badou
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdelhamid Barakat
- Laboratory of Genomics and Human Genetics,Institut Pasteur du Maroc, 1 Place Louis Pasteur, 20360 Casablanca, Morocco.
| |
Collapse
|
24
|
Tahiat A, Yagoubi A, Ladj MS, Belbouab R, Aggoune S, Atek L, Bouziane D, Melzi S, Boubidi C, Drali W, Bendahmane C, Iguerguesdaoune H, Taguemount S, Soufane A, Oukil A, Ketfi A, Messaoudi H, Boukhenfouf N, Ifri MA, Bencharif Madani T, Belhadj H, Benhala KN, Khiari M, Cherif N, Smati L, Arada Z, Zeroual Z, Bouzerar Z, Ibsaine O, Maouche H, Boukari R, Djenouhat K. Diagnostic and Predictive Contribution of Autoantibodies Screening in a Large Series of Patients With Primary Immunodeficiencies. Front Immunol 2021; 12:665322. [PMID: 33868317 PMCID: PMC8047634 DOI: 10.3389/fimmu.2021.665322] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 03/22/2021] [Indexed: 11/13/2022] Open
Abstract
Objectives To evaluate the diagnostic and predictive contribution of autoantibodies screening in patients with primary immunodeficiencies (PIDs). Methods In the present study, PID patients and healthy controls have been screened for 54 different autoantibodies. The results of autoantibodies screening in PID patients were correlated to the presence of autoimmune diseases. Results A total of 299 PID patients were included in this study with a predominance of antibody deficiencies (27.8%) followed by immunodeficiencies affecting cellular and humoral immunity (26.1%) and complement deficiencies (22.7%). Autoimmune manifestations were present in 82 (27.4%) patients. Autoimmune cytopenia (10.4%) was the most common autoimmune disease followed by gastrointestinal disorders (10.0%), rheumatologic diseases (3.7%), and endocrine disorders (3.3%). Autoantibodies were found in 32.4% of PID patients and 15.8% of healthy controls (P < 0.0005). Anti-nuclear antibodies (ANA) (10.0%), transglutaminase antibody (TGA) (8.4%), RBC antibodies (6.7%), anti-smooth muscle antibody (ASMA) (5.4%), and ASCA (5.0%) were the most common autoantibodies in our series. Sixty-seven out of the 82 patients with autoimmune manifestations (81.7%) were positive for one or more autoantibodies. Eleven out of the 14 patients (78.6%) with immune thrombocytopenia had positive platelet-bound IgM. The frequencies of ASCA and ANCA among patients with IBD were 47.4% and 21.0% respectively. All patients with celiac disease had TGA-IgA, while six out of the 11 patients with rheumatologic diseases had ANA (54.5%). Almost one third of patients (30/97) with positive autoantibodies had no autoimmune manifestations. ANA, rheumatoid factor, ASMA, anti-phospholipid antibodies and ANCA were often detected while specific AID was absent. Despite the low positive predictive value of TGA-IgA and ASCA for celiac disease and inflammatory bowel disease respectively, screening for these antibodies identified undiagnosed disease in four patients with positive TGA-IgA and two others with positive ASCA. Conclusion The present study provides valuable information about the frequency and the diagnostic/predictive value of a large panel of autoantibodies in PIDs. Given the frequent association of some AIDs with certain PIDs, screening for corresponding autoantibodies would be recommended. However, positivity for autoantibodies should be interpreted with caution in patients with PIDs due to their low positive predictive value.
Collapse
Affiliation(s)
- Azzeddine Tahiat
- Department of Medical Biology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Abdelghani Yagoubi
- Pediatric Gastroenterology, Centre Algérois de Pédiatrie, Algiers, Algeria
| | - Mohamed Samir Ladj
- Department of Pediatrics, Mustapha University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Reda Belbouab
- Department of Pediatrics, Mustapha University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Samira Aggoune
- Department of Pediatrics, El-Harrach Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Laziz Atek
- Department of Pediatrics, El-Harrach Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Djamila Bouziane
- Department of Pediatrics, Ain Taya Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Souhila Melzi
- Department of Pediatrics, Bab El-Oued University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Chahinez Boubidi
- Department of Pediatrics A, Hussein Dey University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Warda Drali
- Department of Pediatrics B, Hussein Dey University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | | | - Hamza Iguerguesdaoune
- Department of Medical Biology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Sihem Taguemount
- Department of Medical Biology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Asma Soufane
- Department of Medical Biology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Asma Oukil
- Department of Medical Biology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Abdalbasset Ketfi
- Department of Pneumology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Hassen Messaoudi
- Department of Internal Medicine, Mustapha University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | | | | | | | - Hayet Belhadj
- Department of Pediatrics, Central Hospital of the Army, Algiers, Algeria
| | - Keltoum Nafissa Benhala
- Department of Pediatrics A, Beni Messous University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Mokhtar Khiari
- Department of Pediatrics A, Beni Messous University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Nacera Cherif
- Department of Pediatrics B, Beni Messous University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Leila Smati
- Department of Pediatrics, Bologhine Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Zakia Arada
- Department of Pediatrics B, Hussein Dey University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Zoulikha Zeroual
- Department of Pediatrics A, Hussein Dey University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Zair Bouzerar
- Department of Pediatrics, Bab El-Oued University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Ouardia Ibsaine
- Department of Pediatrics, Ain Taya Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Hachemi Maouche
- Department of Pediatrics, El-Harrach Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Rachida Boukari
- Department of Pediatrics, Mustapha University Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| | - Kamel Djenouhat
- Department of Medical Biology, Rouiba Hospital, Algiers Faculty of Medicine, University of Algiers 1, Algiers, Algeria
| |
Collapse
|
25
|
Inborn errors of immunity with atopic phenotypes: A practical guide for allergists. World Allergy Organ J 2021; 14:100513. [PMID: 33717395 PMCID: PMC7907539 DOI: 10.1016/j.waojou.2021.100513] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Inborn errors of immunity (IEI) are a heterogeneous group of disorders, mainly resulting from mutations in genes associated with immunoregulation and immune host defense. These disorders are characterized by different combinations of recurrent infections, autoimmunity, inflammatory manifestations, lymphoproliferation, and malignancy. Interestingly, it has been increasingly observed that common allergic symptoms also can represent the expression of an underlying immunodeficiency and/or immune dysregulation. Very high IgE levels, peripheral or organ-specific hypereosinophilia, usually combined with a variety of atopic symptoms, may sometimes be the epiphenomenon of a monogenic disease. Therefore, allergists should be aware that severe and/or therapy-resistant atopic disorders might be the main clinical phenotype of some IEI. This could pave the way to target therapies, leading to better quality of life and improved survival in affected patients.
Collapse
|
26
|
Vignesh P, Rawat A, Kumrah R, Singh A, Gummadi A, Sharma M, Kaur A, Nameirakpam J, Jindal A, Suri D, Gupta A, Khadwal A, Saikia B, Minz RW, Sharma K, Desai M, Taur P, Gowri V, Pandrowala A, Dalvi A, Jodhawat N, Kambli P, Madkaikar MR, Bhattad S, Ramprakash S, Cp R, Jayaram A, Sivasankaran M, Munirathnam D, Balaji S, Rajendran A, Aggarwal A, Singh K, Na F, George B, Mehta A, Lashkari HP, Uppuluri R, Raj R, Bartakke S, Gupta K, Sreedharanunni S, Ogura Y, Kato T, Imai K, Chan KW, Leung D, Ohara O, Nonoyama S, Hershfield M, Lau YL, Singh S. Clinical, Immunological, and Molecular Features of Severe Combined Immune Deficiency: A Multi-Institutional Experience From India. Front Immunol 2021; 11:619146. [PMID: 33628209 PMCID: PMC7897653 DOI: 10.3389/fimmu.2020.619146] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/17/2020] [Indexed: 01/04/2023] Open
Abstract
Background Severe Combined Immune Deficiency (SCID) is an inherited defect in lymphocyte development and function that results in life-threatening opportunistic infections in early infancy. Data on SCID from developing countries are scarce. Objective To describe clinical and laboratory features of SCID diagnosed at immunology centers across India. Methods A detailed case proforma in an Excel format was prepared by one of the authors (PV) and was sent to centers in India that care for patients with primary immunodeficiency diseases. We collated clinical, laboratory, and molecular details of patients with clinical profile suggestive of SCID and their outcomes. Twelve (12) centers provided necessary details which were then compiled and analyzed. Diagnosis of SCID/combined immune deficiency (CID) was based on 2018 European Society for Immunodeficiencies working definition for SCID. Results We obtained data on 277 children; 254 were categorized as SCID and 23 as CID. Male-female ratio was 196:81. Median (inter-quartile range) age of onset of clinical symptoms and diagnosis was 2.5 months (1, 5) and 5 months (3.5, 8), respectively. Molecular diagnosis was obtained in 162 patients - IL2RG (36), RAG1 (26), ADA (19), RAG2 (17), JAK3 (15), DCLRE1C (13), IL7RA (9), PNP (3), RFXAP (3), CIITA (2), RFXANK (2), NHEJ1 (2), CD3E (2), CD3D (2), RFX5 (2), ZAP70 (2), STK4 (1), CORO1A (1), STIM1 (1), PRKDC (1), AK2 (1), DOCK2 (1), and SP100 (1). Only 23 children (8.3%) received hematopoietic stem cell transplantation (HSCT). Of these, 11 are doing well post-HSCT. Mortality was recorded in 210 children (75.8%). Conclusion We document an exponential rise in number of cases diagnosed to have SCID over the last 10 years, probably as a result of increasing awareness and improvement in diagnostic facilities at various centers in India. We suspect that these numbers are just the tip of the iceberg. Majority of patients with SCID in India are probably not being recognized and diagnosed at present. Newborn screening for SCID is the need of the hour. Easy access to pediatric HSCT services would ensure that these patients are offered HSCT at an early age.
Collapse
Affiliation(s)
- Pandiarajan Vignesh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Rajni Kumrah
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankita Singh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anjani Gummadi
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Madhubala Sharma
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anit Kaur
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Johnson Nameirakpam
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur Jindal
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepti Suri
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Anju Gupta
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Alka Khadwal
- Bone Marrow Transplantation Unit, Department of Internal Medicine, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Biman Saikia
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ranjana Walker Minz
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Kaushal Sharma
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Mukesh Desai
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Prasad Taur
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Vijaya Gowri
- Department of Immunology, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Ambreen Pandrowala
- Bone Marrow Transplantation Unit, Bai Jerbai Wadia Hospital for Children, Mumbai, India
| | - Aparna Dalvi
- ICMR-National Institute of Immunohematology, Mumbai, India
| | - Neha Jodhawat
- ICMR-National Institute of Immunohematology, Mumbai, India
| | | | | | - Sagar Bhattad
- Pediatric Immunology and Rheumatology, Aster CMI hospital, Bengaluru, India
| | - Stalin Ramprakash
- Pediatric Hemat-oncology and BMT Unit, Aster CMI Hospital, Bengaluru, India
| | - Raghuram Cp
- Pediatric Hemat-oncology and BMT Unit, Aster CMI Hospital, Bengaluru, India
| | | | | | | | - Sarath Balaji
- Institute of Child Health, Madras Medical College, Chennai, India
| | - Aruna Rajendran
- Institute of Child Health, Madras Medical College, Chennai, India
| | - Amita Aggarwal
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Komal Singh
- Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow, India
| | - Fouzia Na
- Christian Medical College, Vellore, India
| | | | | | | | | | | | | | - Kirti Gupta
- Department of Histopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Sreejesh Sreedharanunni
- Department of Hematology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Yumi Ogura
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Tamaki Kato
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | - Kohsuke Imai
- Department of Pediatrics, National Defense Medical College, Saitama, Japan.,Department of Community Pediatrics, Perinatal and Maternal Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Koon Wing Chan
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Daniel Leung
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | | | - Shigeaki Nonoyama
- Department of Pediatrics, National Defense Medical College, Saitama, Japan
| | | | - Yu-Lung Lau
- Department of Paediatrics and Adolescent Medicine, The University of Hong Kong, Hong Kong, Hong Kong
| | - Surjit Singh
- Allergy Immunology Unit, Department of Pediatrics, Advanced Pediatrics Centre, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
27
|
Buchbinder D, Walter JE, Butte MJ, Chan WY, Chitty Lopez M, Dimitriades VR, Dorsey MJ, Nugent DJ, Puck JM, Singh J, Collins CA. When Screening for Severe Combined Immunodeficiency (SCID) with T Cell Receptor Excision Circles Is Not SCID: a Case-Based Review. J Clin Immunol 2021; 41:294-302. [PMID: 33411155 PMCID: PMC8179373 DOI: 10.1007/s10875-020-00931-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/18/2020] [Indexed: 12/19/2022]
Abstract
Newborn screening efforts focusing on the quantification of T cell receptor excision circles (TRECs), as a biomarker for abnormal thymic production of T cells, have allowed for the identification and definitive treatment of severe combined immunodeficiency (SCID) in asymptomatic neonates. With the adoption of TREC quantification in Guthrie cards across the USA and abroad, typical, and atypical SCID constitutes only ~ 10% of cases identified with abnormal TRECs associated with T cell lymphopenia. Several other non-SCID-related conditions may be identified by newborn screening in a term infant. Thus, it is important for physicians to recognize that other factors, such as prematurity, are often associated with low TRECs initially, but often improve with age. This paper focuses on a challenge that immunologists face: the diagnostic evaluation and management of cases in which abnormal TRECs are associated with variants of T cell lymphopenia in the absence of a genetically defined form of typical or atypical SCID. Various syndromes associated with T cell impairment, secondary forms of T cell lymphopenia, and idiopathic T cell lymphopenia are identified using this screening approach. Yet there is no consensus or guidelines to assist in the evaluation and management of these newborns, despite representing 90% of the patients identified, resulting in significant work for the clinical teams until a diagnosis is made. Using a case-based approach, we review pearls relevant to the evaluation of these newborns, as well as the management dilemmas for the families and team related to the resolution of genetic ambiguities.
Collapse
Affiliation(s)
- David Buchbinder
- Department of Hematology, Children's Hospital of Orange County, Orange, CA, USA.
- Department of Pediatrics, University of California at Irvine, Orange, CA, USA.
| | - Jolan E Walter
- Division of Pediatric, University of South Florida at Johns Hopkins All Children's Hospital, Allergy/ Immunology, St. Petersburg, FL, USA
- Division of Pediatric Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA, USA
| | - Manish J Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics, University of California Los Angeles, Los Angeles, CA, USA
| | - Wan-Yin Chan
- Department of Allergy & Immunology, Children's Hospital of Orange County, Orange, CA, USA
| | - Maria Chitty Lopez
- Division of Pediatric, University of South Florida at Johns Hopkins All Children's Hospital, Allergy/ Immunology, St. Petersburg, FL, USA
| | - Victoria R Dimitriades
- Division of Allergy, Immunology & Rheumatology, Department of Pediatrics, Sacramento, CA, USA
| | - Morna J Dorsey
- Department of Allergy & Immunology, University of California, San Francisco, CA, USA
| | - Diane J Nugent
- Department of Hematology, Children's Hospital of Orange County, Orange, CA, USA
- Department of Pediatrics, University of California at Irvine, Orange, CA, USA
| | - Jennifer M Puck
- Department of Allergy & Immunology, University of California, San Francisco, CA, USA
| | - Jasjit Singh
- Department of Infectious Disease, Children's Hospital of Orange County, Orange, CA, USA
| | - Cathleen A Collins
- Department of Pediatrics, Division of Allergy Immunology, University of California at San Diego, La Jolla, CA, USA
- Department of Pediatrics, Division of Allergy Immunology, Rady Children's Hospital, San Diego, CA, USA
| |
Collapse
|
28
|
Severe Combined Immunodeficiency Disorder due to a Novel Mutation in Recombination Activation Gene 2: About 2 Cases. Case Reports Immunol 2021; 2021:8819368. [PMID: 33505738 PMCID: PMC7808801 DOI: 10.1155/2021/8819368] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 12/07/2020] [Accepted: 12/26/2020] [Indexed: 11/26/2022] Open
Abstract
Severe combined immunodeficiency (SCID) comprises a heterogeneous group of inherited immunologic disorders with profound defects in cellular and humoral immunity. SCID is the most severe PID and constitutes a pediatric emergency. Affected children are highly susceptible to bacterial, viral, fungal, and opportunistic infections with life-threatening in the absence of hematopoietic stem cell transplantation. We report here two cases of SCID. The first case is a girl diagnosed with SCID at birth based on her family history and lymphocyte subpopulation typing. The second case is a 4-month-old boy with a history of recurrent opportunistic infections, BCGitis, and failure to thrive, and the immunology workup confirms a SCID phenotype. The genetic study in the two cases revealed a novel mutation in the RAG2 gene, c.826G > A (p.Gly276Ser), in a homozygous state. The novel mutation in the RAG2 gene identified in our study may help the early diagnosis of SCID.
Collapse
|
29
|
Houghton BC, Booth C. Gene Therapy for Primary Immunodeficiency. Hemasphere 2021; 5:e509. [PMID: 33403354 PMCID: PMC7773329 DOI: 10.1097/hs9.0000000000000509] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 10/21/2020] [Indexed: 12/27/2022] Open
Abstract
Over the past 3 decades, there has been significant progress in refining gene therapy technologies and procedures. Transduction of hematopoietic stem cells ex vivo using lentiviral vectors can now create a highly effective therapeutic product, capable of reconstituting many different immune system dysfunctions when reinfused into patients. Here, we review the key developments in the gene therapy landscape for primary immune deficiency, from an experimental therapy where clinical efficacy was marred by adverse events, to a commercialized product with enhanced safety and efficacy. We also discuss progress being made in preclinical studies for challenging disease targets and emerging gene editing technologies that are showing promising results, particularly for conditions where gene regulation is important for efficacy.
Collapse
Affiliation(s)
- Benjamin C. Houghton
- Molecular and Cellular Immunology, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Claire Booth
- Molecular and Cellular Immunology, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Paediatric Immunology, Great Ormond Street NHS Foundation Trust, London, United Kingdom
| |
Collapse
|
30
|
Villa A, Capo V, Castiello MC. Innovative Cell-Based Therapies and Conditioning to Cure RAG Deficiency. Front Immunol 2020; 11:607926. [PMID: 33329604 PMCID: PMC7711106 DOI: 10.3389/fimmu.2020.607926] [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: 09/18/2020] [Accepted: 10/26/2020] [Indexed: 12/12/2022] Open
Abstract
Genetic defects in recombination activating genes (RAG) 1 and 2 cause a broad spectrum of severe immune defects ranging from early severe and repeated infections to inflammation and autoimmune manifestations. A correlation between in vitro recombination activity and immune phenotype has been described. Hematopoietic cell transplantation is the treatment of care; however, the availability of next generation sequencing and whole genome sequencing has allowed the identification of novel genetic RAG variants in immunodeficient patients at various ages, raising therapeutic questions. This review addresses the recent advances of novel therapeutic approaches for RAG deficiency. As conventional myeloablative conditioning regimens are associated with acute toxicities and transplanted-related mortality, innovative minimal conditioning regimens based on the use of monoclonal antibodies are now emerging and show promising results. To overcome shortage of compatible donors, gene therapy has been developed in various RAG preclinical models. Overall, the transplantation of autologous gene corrected hematopoietic precursors and the use of non-genotoxic conditioning will open a new era, offering a cure to an increasing number of RAG patients regardless of donor availability and severity of clinical conditions.
Collapse
Affiliation(s)
- Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Valentina Capo
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| | - Maria Carmina Castiello
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy.,Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche (IRGB-CNR), Milan, Italy
| |
Collapse
|
31
|
Geier CB, Farmer JR, Foldvari Z, Ujhazi B, Steininger J, Sleasman JW, Parikh S, Dilley MA, Pai SY, Henderson L, Hazen M, Neven B, Moshous D, Sharapova SO, Mihailova S, Yankova P, Naumova E, Özen S, Byram K, Fernandez J, Wolf HM, Eibl MM, Notarangelo LD, Calabrese LH, Walter JE. Vasculitis as a Major Morbidity Factor in Patients With Partial RAG Deficiency. Front Immunol 2020; 11:574738. [PMID: 33193364 PMCID: PMC7609967 DOI: 10.3389/fimmu.2020.574738] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Accepted: 08/31/2020] [Indexed: 12/30/2022] Open
Abstract
Vasculitis can be a life-threatening complication associated with high mortality and morbidity among patients with primary immunodeficiencies (PIDs), including variants of severe and combined immunodeficiencies ((S)CID). Our understanding of vasculitis in partial defects in recombination activating gene (RAG) deficiency, a prototype of (S)CIDs, is limited with no published systematic evaluation of diagnostic and therapeutic modalities. In this report, we sought to establish the clinical, laboratory features, and treatment outcome of patients with vasculitis due to partial RAG deficiency. Vasculitis was a major complication in eight (13%) of 62 patients in our cohort with partial RAG deficiency with features of infections and immune dysregulation. Vasculitis occurred early in life, often as first sign of disease (50%) and was complicated by significant end organ damage. Viral infections often preceded the onset of predominately non-granulomatous-small vessel vasculitis. Autoantibodies against cytokines (IFN-α, -ω, and IL-12) were detected in a large fraction of the cases tested (80%), whereas the majority of patients were anti-neutrophil cytoplasmic antibodies (ANCA) negative (>80%). Genetic diagnosis of RAG deficiency was delayed up to 2 years from the onset of vasculitis. Clinical cases with sole skin manifestation responded well to first-line steroid treatment, whereas systemic vasculitis with severe end-organ complications required second-line immunosuppression and/or hematopoietic stem cell transplantation (HSCT) for definitive management. In conclusion, our data suggest that vasculitis in partial RAG deficiency is prevalent among patients with partial RAG deficiency and is associated with high morbidity. Therefore, partial RAG deficiency should be included in the differential diagnosis of patients with early-onset systemic vasculitis. Diagnostic serology may be misleading with ANCA negative findings, and search for conventional autoantibodies should be extended to include those targeting cytokines.
Collapse
Affiliation(s)
| | - Jocelyn R Farmer
- Harvard Medical School, Massachusetts General Hospital, Boston, MA, United States
| | - Zsofia Foldvari
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Boglarka Ujhazi
- University of South Florida and Johns Hopkins All Children's Hospital, Saint Petersburg, FL, United States
| | | | - John W Sleasman
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC, United States
| | - Suhag Parikh
- Emory University School of Medicine, Atlanta, GA, United States
| | - Meredith A Dilley
- Department of Immunology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Harvard Medical School, Boston Children's Hospital, Boston, MA, United States.,Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA, United States.,Harvard Medical School, Boston, MA, United States
| | - Lauren Henderson
- Division of Immunology, Department of Rheumatology, Boston Children's Hospital, Boston, MA, United States
| | - Melissa Hazen
- Division of Immunology, Department of Rheumatology, Boston Children's Hospital, Boston, MA, United States
| | - Benedicte Neven
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France.,Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Laboratory "Immunogenetics of Pediatric autoimmune diseases", INSERM UMR1163, Institut Imagine, Université Paris Descartes Sorbonne Paris Cité, Paris, France
| | - Despina Moshous
- Imagine Institute, Paris Descartes-Sorbonne Paris Cité University, Paris, France.,Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France.,Laboratory of Genome Dynamics in The Immune System, Paris, France
| | - Svetlana O Sharapova
- Research Department, Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus
| | - Snezhina Mihailova
- Department of Clinical Immunology Medical University of Sofia, Sofia, Bulgaria
| | - Petya Yankova
- Department of Clinical Immunology Medical University of Sofia, Sofia, Bulgaria
| | - Elisaveta Naumova
- Department of Clinical Immunology Medical University of Sofia, Sofia, Bulgaria
| | - Seza Özen
- Division of Rheumatology, Department of Pediatrics, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Kevin Byram
- Cleveland Clinic Center for Vasculitis Care and Research, Cleveland, OH, United States
| | - James Fernandez
- Cleveland Clinic Center for Vasculitis Care and Research, Cleveland, OH, United States
| | - Hermann M Wolf
- Immunology Outpatient Clinic, Vienna, Austria.,Sigmund Freud Private University- Medical School, Vienna, Austria
| | - Martha M Eibl
- Immunology Outpatient Clinic, Vienna, Austria.,Biomedizinische Forschungs GmbH, Vienna, Austria
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, NIAID, National Institutes of Health, Bethesda, MD, United States
| | - Leonard H Calabrese
- Cleveland Clinic Center for Vasculitis Care and Research, Cleveland, OH, United States
| | - Jolan E Walter
- University of South Florida at Johns Hopkins All Children's Hospital, Saint Petersburg, FL, United States.,Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, MA, United States
| |
Collapse
|
32
|
Richards S, Gennery AR, Davies EG, Wong M, Shaw PJ, Peake J, Fraser C, Gray P, Brothers S, Sinclair J, Prestidge T, Preece K, Quinn P, Ramachandran S, Loh R, McLean-Tooke A, Mitchell R, Cole T. Diagnosis and management of severe combined immunodeficiency in Australia and New Zealand. J Paediatr Child Health 2020; 56:1508-1513. [PMID: 33099818 DOI: 10.1111/jpc.15158] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 08/06/2020] [Indexed: 01/06/2023]
Abstract
This consensus document outlines the recommendations from the Australasian Society of Clinical Immunology and Allergy Transplantation and Primary Immunodeficiency group for the diagnosis and management of patients with severe combined immunodeficiency. It also provides a proposed framework for the early investigation, management and supportive care prior to haematopoietic stem cell transplantation.
Collapse
Affiliation(s)
- Stephanie Richards
- Department of Allergy and Immunology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | - Andrew R Gennery
- Paediatric Immunology and Haematopoietic Stem Cell Transplantation, Great North Children's Hospital, Newcastle upon Tyne, United Kingdom.,Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - E Graham Davies
- Department of Immunology, Great Ormond Street Hospital, London, United Kingdom.,UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Melanie Wong
- Department of Allergy and Immunology, Children's Hospital Westmead, Sydney, New South Wales, Australia
| | - Peter J Shaw
- Bone Marrow Transplant Unit, Children's Hospital Westmead, Sydney, New South Wales, Australia.,Discipline of Child and Adolescent Health, University of Sydney, Sydney, New South Wales, Australia
| | - Jane Peake
- Department of Allergy and Immunology, Queensland Children's Hospital, Brisbane, Queensland, Australia.,Discipline of Paediatrics and Child Health, School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Chris Fraser
- Oncology Service, Queensland Children's Hospital, Brisbane, Queensland, Australia
| | - Paul Gray
- Department of Immunology, Sydney Children's Hospital, Sydney, New South Wales, Australia.,School of Women and Children's Health, University of New South Wales, Sydney, New South Wales, Australia
| | - Shannon Brothers
- Department of Immunology and Allergy, Starship Children's Hospital, Auckland, New Zealand.,Newborn Metabolic Screening, Specialist Chemical Pathology Department, LabPlus, Auckland City Hospital, Auckland, New Zealand
| | - Jan Sinclair
- Department of Immunology and Allergy, Starship Children's Hospital, Auckland, New Zealand
| | - Tim Prestidge
- Blood and Cancer Centre, Starship Children's Hospital, Auckland, New Zealand
| | - Kahn Preece
- Allergy and Immunology Department, John Hunter Children's Hospital, Newcastle, New South Wales, Australia
| | - Patrick Quinn
- Department of Allergy and Clinical Immunology, Women and Children's Hospital, Adelaide, South Australia, Australia.,Discipline of Paediatrics, School of Medicine, University of Adelaide, Adelaide, South Australia, Australia
| | - Shanti Ramachandran
- Department of Paediatric and Adolescent Oncology and Haematology, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Richard Loh
- Immunology Department, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Andrew McLean-Tooke
- Immunology Department, Perth Children's Hospital, Perth, Western Australia, Australia
| | - Richard Mitchell
- School of Women and Children's Health, University of New South Wales, Sydney, New South Wales, Australia.,Kids Cancer Centre, Sydney Children's Hospital, Sydney, New South Wales, Australia
| | - Theresa Cole
- Department of Allergy and Immunology, The Royal Children's Hospital, Melbourne, Victoria, Australia
| | | |
Collapse
|
33
|
Znc2 module of RAG1 contributes towards structure-specific nuclease activity of RAGs. Biochem J 2020; 477:3567-3582. [PMID: 32886094 DOI: 10.1042/bcj20200361] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 09/01/2020] [Accepted: 09/04/2020] [Indexed: 02/06/2023]
Abstract
Recombination activating genes (RAGs), consisting of RAG1 and RAG2 have ability to perform spatially and temporally regulated DNA recombination in a sequence specific manner. Besides, RAGs also cleave at non-B DNA structures and are thought to contribute towards genomic rearrangements and cancer. The nonamer binding domain of RAG1 binds to the nonamer sequence of the signal sequence during V(D)J recombination. However, deletion of NBD did not affect RAG cleavage on non-B DNA structures. In the present study, we investigated the involvement of other RAG domains when RAGs act as a structure-specific nuclease. Studies using purified central domain (CD) and C-terminal domain (CTD) of the RAG1 showed that CD of RAG1 exhibited high affinity and specific binding to heteroduplex DNA, which was irrespective of the sequence of single-stranded DNA, unlike CTD which showed minimal binding. Furthermore, we show that ZnC2 of RAG1 is crucial for its binding to DNA structures as deletion and point mutations abrogated the binding of CD to heteroduplex DNA. Our results also provide evidence that unlike RAG cleavage on RSS, central domain of RAG1 is sufficient to cleave heteroduplex DNA harbouring pyrimidines, but not purines. Finally, we show that a point mutation in the DDE catalytic motif is sufficient to block the cleavage of CD on heteroduplex DNA. Therefore, in the present study we demonstrate that the while ZnC2 module in central domain of RAG1 is required for binding to non-B DNA structures, active site amino acids are important for RAGs to function as a structure-specific nuclease.
Collapse
|
34
|
Immune dysregulation in patients with RAG deficiency and other forms of combined immune deficiency. Blood 2020; 135:610-619. [PMID: 31942628 DOI: 10.1182/blood.2019000923] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2019] [Accepted: 09/25/2019] [Indexed: 12/12/2022] Open
Abstract
Traditionally, primary immune deficiencies have been defined based on increased susceptibility to recurrent and/or severe infections. However, immune dysregulation, manifesting with autoimmunity or hyperinflammatory disease, has emerged as a common feature. This is especially true in patients affected by combined immune deficiency (CID), a group of disorders caused by genetic defects that impair, but do not completely abolish, T-cell function. Hypomorphic mutations in the recombination activating genes RAG1 and RAG2 represent the prototype of the broad spectrum of clinical and immunological phenotypes associated with CID. The study of patients with RAG deficiency and with other forms of CID has revealed distinct abnormalities in central and peripheral T- and B-cell tolerance as the key mechanisms involved in immune dysregulation. Understanding the pathophysiology of autoimmunity and hyperinflammation in these disorders may also permit more targeted therapeutic interventions.
Collapse
|
35
|
Watkins TS, Miles JJ. The human T-cell receptor repertoire in health and disease and potential for omics integration. Immunol Cell Biol 2020; 99:135-145. [PMID: 32677130 DOI: 10.1111/imcb.12377] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2020] [Revised: 07/07/2020] [Accepted: 07/12/2020] [Indexed: 12/11/2022]
Abstract
The adaptive immune system arose 600 million years ago in a cold-blooded fish. Over countless generations, our antecedents tuned the function of the T-cell receptor (TCR). The TCR system is arguably the most complex known to science. The TCR evolved hypervariability to fight the hypervariability of pathogens and cancers that look to consume our resources. This review describes the genetics and architecture of the human TCR and highlights surprising new discoveries over the past years that have disproved very old dogmas. The standardization of TCR sequencing data is discussed in preparation for big data bioinformatics and predictive analysis. We next catalogue new signatures and phenomenon discovered by TCR next generation sequencing (NGS) in health and disease and work that remain to be done in this space. Finally, we discuss how TCR NGS can add to immunodiagnostics and integrate with other omics platforms for both a deeper understanding of TCR biology and its use in the clinical setting.
Collapse
Affiliation(s)
- Thomas S Watkins
- The Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia.,Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| | - John J Miles
- The Australian Institute of Tropical Health and Medicine (AITHM), James Cook University, Cairns, QLD, Australia.,Centre for Molecular Therapeutics, James Cook University, Cairns, QLD, Australia.,Centre for Tropical Bioinformatics and Molecular Biology, James Cook University, Cairns, QLD, Australia
| |
Collapse
|
36
|
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
|
37
|
Tyagi RK, Li J, Jacobse J, Snapper SB, Shouval DS, Goettel JA. Humanized mouse models of genetic immune disorders and hematological malignancies. Biochem Pharmacol 2020; 174:113671. [PMID: 31634456 PMCID: PMC7050416 DOI: 10.1016/j.bcp.2019.113671] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 10/15/2019] [Indexed: 02/07/2023]
Abstract
The immune system is quite remarkable having both the ability to tolerate innocuous and self-antigens while possessing a robust capacity to recognize and eradicate infectious pathogens and foreign entities. The genetics that encode this delicate balancing act include multiple genes and specialized cell types. Over the past several years, whole exome and whole genome sequencing has uncovered the genetics driving many human immune-mediated diseases including monogenic disorders and hematological malignancies. With the advent of genome editing technologies, the ability to correct genetic immune defects in autologous cells holds great promise for a number of conditions. Since assessment of novel therapeutic strategies have been difficult in mice, in recent years, immunodeficient mice capable of engrafting human cells and tissue have been developed and utilized for a variety of research applications. In this review, we discuss immune-humanized mice as a research tool to study human immunobiology and genetic immune disorders in vivo and the promise of future applications.
Collapse
Affiliation(s)
- Rajeev K Tyagi
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Jing Li
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Justin Jacobse
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Willem-Alexander Children's Hospital, Leiden University Medical Center, Leiden, the Netherlands
| | - Scott B Snapper
- Division of Gastroenterology, Hepatology and Nutrition, Boston Children's Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Division of Gastroenterology, Brigham and Women's Hospital, Boston, MA, USA
| | - Dror S Shouval
- Pediatric Gastroenterology Unit, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Tel Hashomer, Israel; Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jeremy A Goettel
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Program in Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN, USA; Vanderbilt Institute for Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA; Center for Mucosal Inflammation and Cancer, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
38
|
Bulkhi AA, Dasso JF, Schuetz C, Walter JE. Approaches to patients with variants in RAG genes: from diagnosis to timely treatment. Expert Rev Clin Immunol 2019; 15:1033-1046. [PMID: 31535575 DOI: 10.1080/1744666x.2020.1670060] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Introduction: Patients with primary immunodeficiency secondary to abnormal recombinase activating genes (RAG) can present with broad clinical phenotypes ranging from early severe infections to autoimmune complications and inflammation. Immunological phenotype may also vary from T-B- severe combined immunodeficiency to combined immunodeficiency or antibody deficiencies with near-normal T and B cell counts and even preserved specific antibody response to pathogens. It is not uncommon that RAG variants of uncertain significance are identified by serendipity during a broad genetic screening process and pathogenic RAG variants are increasingly recognized among all age groups, including adults. Establishing the pathogenicity and clinical relevance of novel RAG variants can be challenging since RAG genes are highly polymorphic. This review paper aims to summarize clinical phenotypes of RAG deficiencies and provide practical guidance for confirming the direct link between specific RAG variants and clinical disease. Lastly, we will review the current understanding of treatment option for patients with varying severity of RAG deficiencies. Area covered: This review discusses the different phenotypes and immunological aspects of RAG deficiencies, the diagnosis dilemma facing clinicians, and an overview of current and advancement in treatments. Expert opinion: A careful analysis of immunological and clinical data and their correlation with genetic findings helps to determine the significance of the genetic polymorphism. Advances in functional assays, as well as anti-cytokine antibodies, make it easier to resolve the diagnostic dilemma.
Collapse
Affiliation(s)
- Adeeb A Bulkhi
- Department of Internal Medicine, College of Medicine, Umm Al-Qura University , Makkah , Saudi Arabia.,Division of Allergy and Immunology, Department of Internal Medicine, Morsani College of Medicine, University of South Florida , Tampa , FL , USA
| | - Joseph F Dasso
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Catharina Schuetz
- Department of Pediatrics, Medical Faculty Carl Gustav Carus, Technical University Dresden , Dresden , Germany
| | - Jolan E Walter
- Division of Allergy and Immunology, Department of Pediatrics, Morsani College of Medicine, University of South Florida , Tampa , FL , USA.,Division of Allergy and Immunology, Department of Medicine, Johns Hopkins All Children's Hospital , St. Petersburg , FL , USA.,Division of Allergy and Immunology, Massachusetts General Hospital for Children , Boston , MA , USA
| |
Collapse
|
39
|
Ibusuki A, Nishikawa T, Hiraki T, Okano T, Imai K, Kanegane H, Ohnishi H, Kato Z, Fujii K, Tanimoto A, Kawano Y, Kanekura T. Prominent dermal Langerhans cells in an Omenn syndrome patient with a novel mutation in the IL2RG gene. J Dermatol 2019; 46:1019-1023. [PMID: 31456262 DOI: 10.1111/1346-8138.15054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2018] [Accepted: 07/25/2019] [Indexed: 11/28/2022]
Abstract
Prominent dermal infiltration by Langerhans cells (LC) is a rare finding in patients with Omenn syndrome (OS). Here, we report the case study of a 7-month-old boy with OS and with prominent dermal infiltration by LC, which is a rare histological manifestation of the skin. Striking erythroderma appeared in the patient 2 weeks after birth. We also noted alopecia, lymphadenopathy, hepatosplenomegaly, eosinophilia and an elevated serum immunoglobulin E level with hypogammaglobulinemia. Peripheral blood flow cytometry showed the Tlow NK+ B+ immunophenotype and genetic analysis, a novel mutation in the IL2RG gene (c.337_339delTCT, p.Ser113del). The final diagnosis was that of OS. He responded well to an allograft umbilical cord blood transplantation that was performed when the patient was 8 months of age. We speculate that the LC accumulated in the dermis will eventually migrate to the regional lymph node, then stimulate autoreactive T cells by overpresenting antigens, thus causing OS-specific skin symptoms.
Collapse
Affiliation(s)
- Atsuko Ibusuki
- Departments of, Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takuro Nishikawa
- Department of, Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tsubasa Hiraki
- Department of, Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tsubasa Okano
- Departments of, Pediatrics and Developmental Biology, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohsuke Imai
- Community Pediatrics, Perinatal and Maternal Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hirokazu Kanegane
- Department of, Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hidenori Ohnishi
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Zenichiro Kato
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Kazuyasu Fujii
- Departments of, Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Akihide Tanimoto
- Department of, Pathology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Yoshifumi Kawano
- Department of, Pediatrics, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Takuro Kanekura
- Departments of, Dermatology, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
40
|
Abstract
While widespread genome sequencing ushers in a new era of preventive medicine, the tools for predictive genomics are still lacking. Time and resource limitations mean that human diseases remain uncharacterized because of an inability to predict clinically relevant genetic variants. A strategy of targeting highly conserved protein regions is used commonly in functional studies. However, this benefit is lost for rare diseases where the attributable genes are mostly conserved. An immunological disorder exemplifying this challenge occurs through damaging mutations in RAG1 and RAG2 which presents at an early age with a distinct phenotype of life-threatening immunodeficiency or autoimmunity. Many tools exist for variant pathogenicity prediction, but these cannot account for the probability of variant occurrence. Here, we present a method that predicts the likelihood of mutation for every amino acid residue in the RAG1 and RAG2 proteins. Population genetics data from approximately 146,000 individuals was used for rare variant analysis. Forty-four known pathogenic variants reported in patients and recombination activity measurements from 110 RAG1/2 mutants were used to validate calculated scores. Probabilities were compared with 98 currently known human cases of disease. A genome sequence dataset of 558 patients who have primary immunodeficiency but that are negative for RAG deficiency were also used as validation controls. We compared the difference between mutation likelihood and pathogenicity prediction. Our method builds a map of most probable mutations allowing pre-emptive functional analysis. This method may be applied to other diseases with hopes of improving preparedness for clinical diagnosis.
Collapse
|
41
|
Farmer JR, Foldvari Z, Ujhazi B, De Ravin SS, Chen K, Bleesing JJH, Schuetz C, Al-Herz W, Abraham RS, Joshi AY, Costa-Carvalho BT, Buchbinder D, Booth C, Reiff A, Ferguson PJ, Aghamohammadi A, Abolhassani H, Puck JM, Adeli M, Cancrini C, Palma P, Bertaina A, Locatelli F, Di Matteo G, Geha RS, Kanariou MG, Lycopoulou L, Tzanoudaki M, Sleasman JW, Parikh S, Pinero G, Fischer BM, Dbaibo G, Unal E, Patiroglu T, Karakukcu M, Al-Saad KK, Dilley MA, Pai SY, Dutmer CM, Gelfand EW, Geier CB, Eibl MM, Wolf HM, Henderson LA, Hazen MM, Bonfim C, Wolska-Kuśnierz B, Butte MJ, Hernandez JD, Nicholas SK, Stepensky P, Chandrakasan S, Miano M, Westermann-Clark E, Goda V, Kriván G, Holland SM, Fadugba O, Henrickson SE, Ozen A, Karakoc-Aydiner E, Baris S, Kiykim A, Bredius R, Hoeger B, Boztug K, Pashchenko O, Neven B, Moshous D, Villartay JPD, Bousfiha AA, Hill HR, Notarangelo LD, Walter JE. Outcomes and Treatment Strategies for Autoimmunity and Hyperinflammation in Patients with RAG Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2019; 7:1970-1985.e4. [PMID: 30877075 PMCID: PMC6612449 DOI: 10.1016/j.jaip.2019.02.038] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2018] [Revised: 02/15/2019] [Accepted: 02/19/2019] [Indexed: 11/19/2022]
Abstract
BACKGROUND Although autoimmunity and hyperinflammation secondary to recombination activating gene (RAG) deficiency have been associated with delayed diagnosis and even death, our current understanding is limited primarily to small case series. OBJECTIVE Understand the frequency, severity, and treatment responsiveness of autoimmunity and hyperinflammation in RAG deficiency. METHODS In reviewing the literature and our own database, we identified 85 patients with RAG deficiency, reported between 2001 and 2016, and compiled the largest case series to date of 63 patients with prominent autoimmune and/or hyperinflammatory pathology. RESULTS Diagnosis of RAG deficiency was delayed a median of 5 years from the first clinical signs of immune dysregulation. Most patients (55.6%) presented with more than 1 autoimmune or hyperinflammatory complication, with the most common etiologies being cytopenias (84.1%), granulomas (23.8%), and inflammatory skin disorders (19.0%). Infections, including live viral vaccinations, closely preceded the onset of autoimmunity in 28.6% of cases. Autoimmune cytopenias had early onset (median, 1.9, 2.1, and 2.6 years for autoimmune hemolytic anemia, immune thrombocytopenia, and autoimmune neutropenia, respectively) and were refractory to intravenous immunoglobulin, steroids, and rituximab in most cases (64.7%, 73.7%, and 71.4% for autoimmune hemolytic anemia, immune thrombocytopenia, and autoimmune neutropenia, respectively). Evans syndrome specifically was associated with lack of response to first-line therapy. Treatment-refractory autoimmunity/hyperinflammation prompted hematopoietic stem cell transplantation in 20 patients. CONCLUSIONS Autoimmunity/hyperinflammation can be a presenting sign of RAG deficiency and should prompt further evaluation. Multilineage cytopenias are often refractory to immunosuppressive treatment and may require hematopoietic cell transplantation for definitive management.
Collapse
Affiliation(s)
- Jocelyn R Farmer
- Massachusetts General Hospital, Harvard Medical School, Boston, Mass
| | - Zsofia Foldvari
- Department of Cancer Immunology, Institute for Cancer Research, Oslo University Hospital, Radiumhospitalet, Oslo, Norway
| | - Boglarka Ujhazi
- University of South Florida and Johns Hopkins All Children's Hospital, Saint Petersburg, Fla
| | - Suk See De Ravin
- Laboratory of Host Defenses, National Institutes of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Karin Chen
- Division of Allergy and Immunology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
| | - Jack J H Bleesing
- Division of Bone Marrow Transplantation and Immune Deficiency, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio
| | - Catharina Schuetz
- Department of Pediatrics, Medizinische Fakultät Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Waleed Al-Herz
- Faculty of Medicine, Pediatrics Department, Kuwait University, Kuwait City, Kuwait; Allergy and Clinical Immunology Unit, Pediatrics Department, Alsabah Hospital, Kuwait City, Kuwait
| | - Roshini S Abraham
- Department of Pathology and Laboratory Medicine, Nationwide Children's Hospital, Columbus, Ohio; Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - Avni Y Joshi
- Division of Pediatric Allergy/Immunology, Mayo Clinic Children's Center Rochester, Rochester, Minn
| | | | - David Buchbinder
- Pediatrics/Hematology, CHOC Children's Hospital - UC Irvine, Irvine, Calif
| | - Claire Booth
- Department of Paediatric Immunology, Great Ormond Street Hospital, London, United Kingdom
| | - Andreas Reiff
- Division of Rheumatology, Children's Hospital Los Angeles, Keck School of Medicine, USC, Los Angeles, Calif
| | - Polly J Ferguson
- Department of Pediatrics, University of Iowa Carver College of Medicine, Iowa City, Iowa
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Jennifer M Puck
- Department of Pediatrics, University of California San Francisco and UCSF Benioff Children's Hospital, San Francisco, Calif
| | - Mehdi Adeli
- Sidra Medicine, Weill Cornell Medicine, and Hamad Medical Corporation, Doha, Qatar
| | - Caterina Cancrini
- Academic Department of Pediatrics (DPUO), Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paolo Palma
- Academic Department of Pediatrics (DPUO), Research Unit in Congenital and Perinatal Infection, Children's Hospital Bambino Gesù, Rome, Italy
| | - Alice Bertaina
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Bambino Gesù, Rome, Italy
| | - Franco Locatelli
- Department of Pediatric Hematology/Oncology and Cell and Gene Therapy, IRCCS, Ospedale Bambino Gesù, Rome, Italy; Department of Pediatrics, Sapienza, University of Rome, Rome, Italy
| | - Gigliola Di Matteo
- Academic Department of Pediatrics (DPUO), Unit of Immune and Infectious Diseases, Childrens' Hospital Bambino Gesù, Rome, Italy; Department of Systems Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Raif S Geha
- Immunology Division, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Maria G Kanariou
- Department of Immunology - Histocompatibility, Specialized Center & Referral Center for Primary Immunodeficiencies - Paediatric Immunology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - Lilia Lycopoulou
- 1st Department of Pediatrics, University of Athens, Aghia Sofia Children's Hospital, Athens, Greece
| | - Marianna Tzanoudaki
- Department of Immunology - Histocompatibility, Specialized Center & Referral Center for Primary Immunodeficiencies - Paediatric Immunology, "Aghia Sophia" Children's Hospital, Athens, Greece
| | - John W Sleasman
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC
| | - Suhag Parikh
- Division of Pediatric Blood and Marrow Transplantation, Duke University School of Medicine, Durham, NC
| | - Gloria Pinero
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC
| | - Bernard M Fischer
- Division of Allergy, Immunology and Pulmonary Medicine, Duke University School of Medicine, Durham, NC
| | - Ghassan Dbaibo
- Department of Pediatrics and Adolescent Medicine, Center for Infectious Diseases Research, American University of Beirut, Beirut, Lebanon
| | - Ekrem Unal
- Division of Pediatric Hematology and Oncology & HCST Unit, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Turkan Patiroglu
- Division of Pediatric Hematology and Oncology & HCST Unit, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey; Division of Pediatric Immunology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Musa Karakukcu
- Division of Pediatric Hematology and Oncology & HCST Unit, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, Turkey
| | - Khulood Khalifa Al-Saad
- Salmanyia Medical Complex, Department of Pediatrics, Division of Pediatric Hematology and Oncology, Manama, Bahrain
| | - Meredith A Dilley
- Department of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Sung-Yun Pai
- Division of Hematology-Oncology, Boston Children's Hospital, Boston, Mass; Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, Mass; Harvard Medical School, Boston, Mass
| | - Cullen M Dutmer
- Division of Allergy & Immunology, Children's Hospital Colorado, University of Colorado School of Medicine, Aurora, Colo
| | - Erwin W Gelfand
- Department of Pediatrics, National Jewish Health, Denver, Colo
| | | | - Martha M Eibl
- Immunology Outpatient Clinic, Vienna, Austria; Biomedizinische Forschungs GmbH, Vienna, Austria
| | - Hermann M Wolf
- Immunology Outpatient Clinic, Vienna, Austria; Sigmund Freud Private University-Medical School, Vienna, Austria
| | - Lauren A Henderson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Melissa M Hazen
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, Mass
| | - Carmem Bonfim
- Hospital Infantil Pequeno Principe, Curitiba, Brazil
| | | | - Manish J Butte
- Division of Immunology, Allergy, and Rheumatology, Department of Pediatrics and Jeffrey Modell Diagnostic and Research Center, University of California, Los Angeles, Los Angeles, Calif
| | - Joseph D Hernandez
- Department of Pediatrics, Division of Allergy, Immunology and Rheumatology, Stanford University, Stanford, Calif
| | - Sarah K Nicholas
- Section of Immunology, Allergy, and Rheumatology, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Polina Stepensky
- Department of Bone Marrow Transplantation, Hadassah Hebrew University Hospital, Jerusalem, Israel
| | - Shanmuganathan Chandrakasan
- Division of Bone Marrow Transplant, Aflac Cancer and Blood Disorders Center, Children's Healthcare of Atlanta, Emory University School of Medicine, Atlanta, Ga
| | - Maurizio Miano
- Haematology Unit, Department of Pediatric Haematology-Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Emma Westermann-Clark
- Department of Internal Medicine, Division of Allergy/Immunology, University of South Florida Morsani College of Medicine, Tampa, Fla
| | - Vera Goda
- Department for Pediatric Hematology and Hemopoietic Stem Cell Transplantation, Central Hospital of Southern Pest- National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Gergely Kriván
- Department for Pediatric Hematology and Hemopoietic Stem Cell Transplantation, Central Hospital of Southern Pest- National Institute of Hematology and Infectious Diseases, Budapest, Hungary
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Olajumoke Fadugba
- Division of Pulmonary, Allergy and Critical Care, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pa
| | - Sarah E Henrickson
- Allergy Immunology Division, Department of Pediatrics, The Children's Hospital of Philadelphia, Philadelphia, Pa; Institute for Immunology, the University of Pennsylvania, Philadelphia, Pa
| | - Ahmet Ozen
- Marmara University School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Elif Karakoc-Aydiner
- Marmara University School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Safa Baris
- Marmara University School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkey
| | - Ayca Kiykim
- Ministry of Health, Marmara University Pendik Training and Research Hospital, Istanbul, Turkey
| | - Robbert Bredius
- Department of Pediatrics, Section Pediatric Immunology, Infections and Stem Cell Transplantation, Leiden University Medical Center, Leiden, the Netherlands
| | - Birgit Hoeger
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria; CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, Vienna, Austria; St Anna Kinderspital and Children's Cancer Research Institute, Department of Pediatrics, Medical University of Vienna, Vienna, Austria; Department of Pediatrics and Adolescent Medicine, Medical University of Vienna, Vienna, Austria
| | - Olga Pashchenko
- Department of Immunology, Pirogov Russian National Research Medical University, Russian Clinical Children's Hospital, Moscow, Russia
| | - Benedicte Neven
- Paris Descartes Sorbonne Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory "Immunogenetics of Pediatric Autoimmune Diseases", INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Despina Moshous
- Paris Descartes Sorbonne Paris Cité University, Imagine Institute, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker-Enfants Malades University Hospital, Assistance Publique-Hôpitaux de Paris, Paris, France; Laboratory "Genome Dynamics in The Immune System", INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Jean-Pierre de Villartay
- Laboratory "Genome Dynamics in The Immune System", INSERM UMR1163, Université Paris Descartes Sorbonne Paris Cité, Institut Imagine, Paris, France
| | - Ahmed Aziz Bousfiha
- Laboratoire d'Immunologie Clinique, d'Inflammation et d'Allergie LICIA, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco; Clinical Immunology Unit, Casablanca Children's Hospital, Ibn Rochd Medical School, Hassan II University, Casablanca, Morocco
| | - Harry R Hill
- Division of Clinical Immunology, Departments of Pathology, Pediatrics and Medicine, University of Utah, Salt Lake City, Utah
| | - Luigi D Notarangelo
- Haematology Unit, Department of Pediatric Haematology-Oncology, IRCCS Istituto Giannina Gaslini, Genova, Italy
| | - Jolan E Walter
- University of South Florida and Johns Hopkins All Children's Hospital, Saint Petersburg, Fla; Division of Allergy and Immunology, Massachusetts General Hospital for Children, Boston, Mass.
| |
Collapse
|
42
|
Abstract
Recombination-activating genes (
RAG)
1 and
RAG2 initiate the molecular processes that lead to lymphocyte receptor formation through VDJ recombination. Nonsense mutations in
RAG1/
RAG2 cause the most profound immunodeficiency syndrome, severe combined immunodeficiency (SCID). Other severe and less-severe clinical phenotypes due to mutations in
RAG genes are now recognized. The degree of residual protein function may permit some lymphocyte receptor formation, which confers a less-severe clinical phenotype. Many of the non-SCID phenotypes are associated with autoimmunity. New findings into the effect of mutations in
RAG1/2 on the developing T- and B-lymphocyte receptor give insight into the development of autoimmunity. This article summarizes recent findings and places the genetic and molecular findings in a clinical context.
Collapse
Affiliation(s)
- Andrew Gennery
- Paediatric Immunology and Haematopoietic Stem Cell Transplantation, Great North Childrens' Hospital, Newcastle upon Tyne, UK.,Primary Immunodeficiency Group, Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
43
|
Meshaal SS, El Hawary RE, Abd Elaziz DS, Eldash A, Alkady R, Lotfy S, Mauracher AA, Opitz L, Pachlopnik Schmid J, van der Burg M, Chou J, Galal NM, Boutros JA, Geha R, Elmarsafy AM. Phenotypical heterogeneity in RAG-deficient patients from a highly consanguineous population. Clin Exp Immunol 2019; 195:202-212. [PMID: 30307608 PMCID: PMC6330646 DOI: 10.1111/cei.13222] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2018] [Indexed: 12/16/2022] Open
Abstract
Mutations affecting recombination activation genes RAG1 and RAG2 are associated with variable phenotypes, depending on the residual recombinase activity. The aim of this study is to describe a variety of clinical phenotypes in RAG-deficient patients from the highly consanguineous Egyptian population. Thirty-one patients with RAG mutations (from 28 families) were included from 2013 to 2017. On the basis of clinical, immunological and genetic data, patients were subdivided into three groups; classical T- B- severe combined immunodeficiency (SCID), Omenn syndrome (OS) and atypical SCID. Nineteen patients presented with typical T- B- SCID; among these, five patients carried a homozygous RAG2 mutation G35V and five others carried two homozygous RAG2 mutations (T215I and R229Q) that were detected together. Four novel mutations were reported in the T- B- SCID group; three in RAG1 (A565P, N591Pfs*14 and K621E) and one in RAG2 (F29S). Seven patients presented with OS and a novel RAG2 mutation (C419W) was documented in one patient. The atypical SCID group comprised five patients. Two had normal B cell counts; one had a previously undescribed RAG2 mutation (V327D). The other three patients presented with autoimmune cytopaenias and features of combined immunodeficiency and were diagnosed at a relatively late age and with a substantial diagnostic delay; one patient had a novel RAG1 mutation (C335R). PID disorders are frequent among Egyptian children because of the high consanguinity. RAG mutations stand behind several variable phenotypes, including classical SCID, OS, atypical SCID with autoimmunity and T- B+ CID.
Collapse
Affiliation(s)
- S. S. Meshaal
- Clinical Pathology Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - R. E. El Hawary
- Clinical Pathology Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - D. S. Abd Elaziz
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - A. Eldash
- Clinical Pathology Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - R. Alkady
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - S. Lotfy
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - A. A. Mauracher
- Division of ImmunologyUniversity Children’s Hospital ZurichZurichSwitzerland
| | - L. Opitz
- Functional Genomics Center ZürichUniversity of Zurich, ETH ZurichZurichSwitzerland
| | | | - M. van der Burg
- Department of ImmunologyErasmus MC, University Medical Center RotterdamRotterdamNetherlands
| | - J. Chou
- Division of ImmunologyBoston Children’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - N. M. Galal
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - J. A. Boutros
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| | - R. Geha
- Division of ImmunologyBoston Children’s Hospital, Harvard Medical SchoolBostonMAUSA
| | - A. M. Elmarsafy
- Pediatrics Department, Faculty of MedicineCairo UniversityCairoEgypt
| |
Collapse
|
44
|
The microbiome and immunodeficiencies: Lessons from rare diseases. J Autoimmun 2019; 98:132-148. [PMID: 30704941 DOI: 10.1016/j.jaut.2019.01.008] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Revised: 01/21/2019] [Accepted: 01/23/2019] [Indexed: 12/20/2022]
Abstract
Primary immunodeficiencies (PIDs) are inherited disorders of the immune system, associated with a considerable increase in susceptibility to infections. PIDs can also predispose to malignancy, inflammation and autoimmunity. There is increasing awareness that some aspects of the immune dysregulation in PIDs may be linked to intestinal microbiota. Indeed, the gut microbiota and its metabolites have been shown to influence immune functions and immune homeostasis both locally and systemically. Recent studies have indicated that genetic defects causing PIDs lead to perturbations in the conventional mechanisms underlying homeostasis in the gut, resulting in poor immune surveillance at the intestinal barrier, which associates with altered intestinal permeability and bacterial translocation. Consistently, a substantial proportion of PID patients presents with clinically challenging IBD-like pathology. Here, we describe the current body of literature reporting on dysbiosis of the gut microbiota in different PIDs and how this can be either the result or cause of immune dysregulation. Further, we report how infections in PIDs enhance pathobionts colonization and speculate how, in turn, pathobionts may be responsible for increased disease susceptibility and secondary infections in these patients. The potential relationship between the microbial composition in the intestine and other sites, such as the oral cavity and skin, is also highlighted. Finally, we provide evidence, in preclinical models of PIDs, for the efficacy of microbiota manipulation to ameliorate disease complications, and suggest that the potential use of dietary intervention to correct dysbiotic flora in PID patients may hold promise.
Collapse
|
45
|
Abstract
PURPOSE OF REVIEW Natural killer cells are innate lymphoid cells (ILCs) that play critical roles in human host defense and are especially useful in combating viral pathogens and malignancy. RECENT FINDINGS The NK cell deficiency (NKD) is particularly underscored in patients with a congenital immunodeficiency in which NK cell development or function is affected. The classical NK cell deficiency (cNKD) is a result of absent or a profound decrease in the number of circulating NK cells. In contrast, functional NKD (fNKD) is characterized by abnormal NK cell function but with normal number of NK cells. The combined immune deficiencies with significant impact on NK cells are not considered classical or functional NK cell deficiencies. In these disorders, the impairment of NK cells represents an important aspect of the overall immunodeficiency. In turn, this leads to improved insights on the NK cell development and function. Here, we detail the NK cell biology based upon recent natural killer cell defects described in combined immune deficiencies.
Collapse
|
46
|
Pazmandi J, Kalinichenko A, Ardy RC, Boztug K. Early-onset inflammatory bowel disease as a model disease to identify key regulators of immune homeostasis mechanisms. Immunol Rev 2019; 287:162-185. [PMID: 30565237 PMCID: PMC7379380 DOI: 10.1111/imr.12726] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 09/23/2018] [Indexed: 12/11/2022]
Abstract
Rare, monogenetic diseases present unique models to dissect gene functions and biological pathways, concomitantly enhancing our understanding of the etiology of complex (and often more common) traits. Although inflammatory bowel disease (IBD) is a generally prototypic complex disease, it can also manifest in an early-onset, monogenic fashion, often following Mendelian modes of inheritance. Recent advances in genomic technologies have spurred the identification of genetic defects underlying rare, very early-onset IBD (VEO-IBD) as a disease subgroup driven by strong genetic influence, pinpointing key players in the delicate homeostasis of the immune system in the gut and illustrating the intimate relationships between bowel inflammation, systemic immune dysregulation, and primary immunodeficiency with increased susceptibility to infections. As for other human diseases, it is likely that adult-onset diseases may represent complex diseases integrating the effects of host genetic susceptibility and environmental triggers. Comparison of adult-onset IBD and VEO-IBD thus provides beautiful models to investigate the relationship between monogenic and multifactorial/polygenic diseases. This review discusses the present and novel findings regarding monogenic IBD as well as key questions and future directions of IBD research.
Collapse
Affiliation(s)
- Julia Pazmandi
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Artem Kalinichenko
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Rico Chandra Ardy
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
| | - Kaan Boztug
- Ludwig Boltzmann Institute for Rare and Undiagnosed DiseasesViennaAustria
- CeMM Research Center for Molecular Medicine of the Austrian Academy of SciencesViennaAustria
- Department of Pediatrics and Adolescent MedicineMedical University of ViennaViennaAustria
- Department of PediatricsSt. Anna Kinderspital and Children's Cancer Research InstituteMedical University of ViennaViennaAustria
| |
Collapse
|
47
|
Villa A, Notarangelo LD. RAG gene defects at the verge of immunodeficiency and immune dysregulation. Immunol Rev 2019; 287:73-90. [PMID: 30565244 PMCID: PMC6309314 DOI: 10.1111/imr.12713] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 08/21/2018] [Indexed: 12/18/2022]
Abstract
Mutations of the recombinase activating genes (RAG) in humans underlie a broad spectrum of clinical and immunological phenotypes that reflect different degrees of impairment of T- and B-cell development and alterations of mechanisms of central and peripheral tolerance. Recent studies have shown that this phenotypic heterogeneity correlates, albeit imperfectly, with different levels of recombination activity of the mutant RAG proteins. Furthermore, studies in patients and in newly developed animal models carrying hypomorphic RAG mutations have disclosed various mechanisms underlying immune dysregulation in this condition. Careful annotation of clinical outcome and immune reconstitution in RAG-deficient patients who have received hematopoietic stem cell transplantation has shown that progress has been made in the treatment of this disease, but new approaches remain to be tested to improve stem cell engraftment and durable immune reconstitution. Finally, initial attempts have been made to treat RAG deficiency with gene therapy.
Collapse
Affiliation(s)
- Anna Villa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cell and Gene Therapy, San Raffaele Scientific Institute, Milan, Italy
- Milan Unit, Istituto di Ricerca Genetica e Biomedica, Consiglio Nazionale delle Ricerche, Milan, Italy
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
48
|
Yazdani R, Fekrvand S, Shahkarami S, Azizi G, Moazzami B, Abolhassani H, Aghamohammadi A. The hyper IgM syndromes: Epidemiology, pathogenesis, clinical manifestations, diagnosis and management. Clin Immunol 2018; 198:19-30. [PMID: 30439505 DOI: 10.1016/j.clim.2018.11.007] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2018] [Accepted: 11/11/2018] [Indexed: 12/17/2022]
Abstract
Hyper Immunoglobulin M syndrome (HIGM) is a rare primary immunodeficiency disorder characterized by low or absent levels of serum IgG, IgA, IgE and normal or increased levels of serum IgM. Various X-linked and autosomal recessive/dominant mutations have been reported as the underlying cause of the disease. Based on the underlying genetic defect, the affected patients present a variety of clinical manifestations including pulmonary and gastrointestinal complications, autoimmune disorders, hematologic abnormalities, lymphoproliferation and malignancies which could be controlled by multiple relevant therapeutic approaches. Herein, the epidemiology, pathogenesis, clinical manifestations, diagnosis, management, prognosis and treatment in patients with HIGM syndrome have been reviewed.
Collapse
Affiliation(s)
- Reza Yazdani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Saba Fekrvand
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Sepideh Shahkarami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Gholamreza Azizi
- Non-Communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| | - Bobak Moazzami
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran
| | - Hassan Abolhassani
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran; Division of Clinical Immunology, Department of Laboratory Medicine, Karolinska Institute at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Asghar Aghamohammadi
- Research Center for Immunodeficiencies, Pediatrics Center of Excellence, Children's Medical Center, Tehran University of Medical Science, Tehran, Iran.
| |
Collapse
|
49
|
Hematopoietic stem cell gene therapy for the cure of blood diseases: primary immunodeficiencies. RENDICONTI LINCEI-SCIENZE FISICHE E NATURALI 2018. [DOI: 10.1007/s12210-018-0742-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
50
|
Chi ZH, Wei W, Bu DF, Li HH, Ding F, Zhu P. Targeted high-throughput sequencing technique for the molecular diagnosis of primary immunodeficiency disorders. Medicine (Baltimore) 2018; 97:e12695. [PMID: 30290665 PMCID: PMC6200533 DOI: 10.1097/md.0000000000012695] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
The aim of this study was to investigate the usefulness of targeted high-throughput sequencing (HTS) for the molecular diagnosis of primary immunodeficiency diseases (PID).A total of 56 clinically diagnosed or suspected PID patients were divided into 4 groups according to the International Union of Immunological Societies Expert Committee for Primary Immunodeficiency 2015 and their chief clinical presentations. Patients and their biological family members were examined by targeted HTS, which sequenced the exons and ±10 bp flanking introns of 171 PID-related genes panel. All significant variants were confirmed by PCR-Sanger sequencing. Pathogenicity of the variants was evaluated by using bioinformatics.A total of 117 variants in 73 genes were found in 56 patients. Accurate molecular diagnosis of PID was made in 13 (23.2%) patients, and 12 novel mutations were detected in these patients. Twenty-seven patients carried heterozygous variants that are probably pathogenic in ≥2 genes; 16 patients had only 1 missense variant, or had several variants but not >1 variant was deleterious as evaluated by bioinformatics. The meaning of the targeted HTS results of these patients remains to be studied.Targeted HTS can make a precise molecular diagnosis of PID and detect more novel pathogenic mutations. More and more variations with ambiguous significance are discovered and explanation of these variations is a challenge to the clinicians.
Collapse
Affiliation(s)
- Zuo Hua Chi
- The First Affiliated Hospital/School of Clinical Medicine, Guang Dong Pharmaceutical University, Guangzhou
| | | | - Ding Fang Bu
- Hematology Research Laboratory, Peking University First Hospital, Beijing
| | - Huan Huan Li
- Department of Pediatrics, Zhengzhou University First Hospital, Zhengzhou
| | - Fei Ding
- Hematology & Oncology Institute, Beijing Aerospace General Hospital, Beijing, China
| | - Ping Zhu
- Hematology Research Laboratory, Peking University First Hospital, Beijing
| |
Collapse
|