1
|
Di Spirito F, Pisano M, Di Palo MP, De Benedetto G, Rizki I, Franci G, Amato M. Periodontal Status and Herpesiviridae, Bacteria, and Fungi in Gingivitis and Periodontitis of Systemically Compromised Pediatric Subjects: A Systematic Review. CHILDREN (BASEL, SWITZERLAND) 2025; 12:375. [PMID: 40150657 PMCID: PMC11941093 DOI: 10.3390/children12030375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/18/2025] [Revised: 03/10/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Gingivitis and periodontitis are microbially associated diseases, with some features characteristic of pediatric age and others linked to systemic diseases. While the role of periodontal pathogenic bacteria is well recognized, the contribution of fungi and viruses, particularly Herpesviridae, remains controversial. Studies in adults have highlighted the presence of Herpesviridae, but evidence in pediatric subjects, especially systemically compromised, is limited. This systematic review aimed to assess periodontal status (e.g., health, gingivitis, periodontitis, necrotizing gingivitis, and/or periodontitis) and the subgingival and/or salivary microbial (bacterial, viral, and fungal) profile in systemically compromised pediatric (≤18 years) subjects with gingivitis and/or periodontitis compared to clinical periodontal health. METHODS The review protocol was registered on PROSPERO (CRD42024597695) and followed the PRISMA statement. Data from eight studies were descriptively analyzed and qualitatively assessed through ROBINS-I and JBI tools. RESULTS CMV was frequently detected, particularly in necrotizing gingivitis (19.40%). EBV was found in necrotizing gingivitis (20.69%) and periodontitis (10.34%); HSV was mainly associated with gingivitis and necrotizing gingivitis. Bacteria species in periodontitis included Porphyromonas gingivalis, Tannerella forsythia, Fusobacterium, and Campylobacter species. Candida albicans was detected in periodontitis, suggesting a fungal involvement in the disease's pathogenesis. Although the bacterial and fungal profile was not investigated, limited viral presence was noted in subjects with healthy periodontium, indicating a stable microbiome. CONCLUSIONS These findings underscore the dynamics of microbial interactions in the progression of periodontal disease in systemically compromised pediatric subjects.
Collapse
Affiliation(s)
- Federica Di Spirito
- Department of Medicine, Surgery and Dentistry, University of Salerno, Via S. Allende, 84081 Baronissi, Italy; (M.P.); (M.P.D.P.); (G.D.B.); (I.R.); (M.A.)
| | | | | | | | | | | | | |
Collapse
|
2
|
Zhang M, Cron RR, Chu N, Nguyen J, Gordon SM, Eloseily EM, Atkinson TP, Weiser P, Walter MR, Kreiger PA, Canna SW, Behrens EM, Cron RQ. Role of DOCK8 in cytokine storm syndromes. J Allergy Clin Immunol 2025; 155:1015-1026.e5. [PMID: 39423879 PMCID: PMC11875994 DOI: 10.1016/j.jaci.2024.10.004] [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: 04/03/2024] [Revised: 09/18/2024] [Accepted: 10/08/2024] [Indexed: 10/21/2024]
Abstract
BACKGROUND Cytokine storm syndromes (CSSs), including hemophagocytic lymphohistiocytosis (HLH), are increasingly recognized as hyperinflammatory states leading to multiorgan failure and death. Familial HLH in infancy results from homozygous genetic defects in perforin-mediated cytolysis by CD8 T lymphocytes and natural killer (NK) cells. Later-onset CSSs are often associated with heterozygous defects in familial HLH genes, but genetic etiologies for most are unknown. We identified rare dedicator of cytokinesis 8 (DOCK8) variants in patients with CSS. OBJECTIVE We sought to explore the role of CSS patient-derived DOCK8 mutations on cytolytic activity in NK cells and to further study effects of DOCK8 deficiency in murine models of CSSs. METHODS DOCK8 cDNAs from 2 unrelated patients with CSS with different missense mutations were introduced into human NK-92 cells by foamy virus transduction. NK-cell degranulation (CD107a), cytolytic activity against K562 target cells, and IFN-γ production were explored by flow cytometry. A third patient with CSS with DOCK8 mRNA splice acceptor site variant was explored by exon trapping. Dock8-/- mice were assessed for features of CSS (weight loss, splenomegaly, hepatic inflammation, cytopenias, and IFN-γ levels) on challenge with lymphocytic choriomeningitis virus and excess IL-18. RESULTS Both patient DOCK8 missense mutations decreased cytolytic function in NK cells in a partial dominant-negative fashion in vitro. The patient DOCK8 splice variant disrupted mRNA splicing in vitro. Lymphocytic choriomeningitis virus infection promoted CSS in Dock8-/- mice and interacted with excess IL-18, limiting T-cell numbers while promoting CD8 T-cell hyperactivation. CONCLUSIONS Mutations in DOCK8 may contribute to CSS-like hyperinflammatory states by altering cytolytic function in a threshold model of disease.
Collapse
Affiliation(s)
- Mingce Zhang
- University of Alabama at Birmingham, Birmingham, Ala
| | - Remy R Cron
- University of Alabama at Birmingham, Birmingham, Ala
| | | | | | | | - Esraa M Eloseily
- University of Texas Southwestern, Dallas, Tex; Faculty of Medicine, Assiut University, Asyut, Egypt
| | | | - Peter Weiser
- University of Alabama at Birmingham, Birmingham, Ala
| | - Mark R Walter
- University of Alabama at Birmingham, Birmingham, Ala
| | | | | | | | - Randy Q Cron
- University of Alabama at Birmingham, Birmingham, Ala.
| |
Collapse
|
3
|
Trombello S, Jarisch A, Willasch A, Rettinger E, Fekadu-Siebald J, Holzinger D, Adelmann R, Bader P, Bakhtiar S. Case report: Advanced age at transplantation and pre-emptive treatment with dupilumab in DOCK8 deficiency. Front Immunol 2025; 15:1507494. [PMID: 39936153 PMCID: PMC11810938 DOI: 10.3389/fimmu.2024.1507494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 12/02/2024] [Indexed: 02/13/2025] Open
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency is a combined immunodeficiency (CID) due to biallelic mutations in the gene encoding DOCK8. Major clinical phenomena are recurrent severe infections of the lungs and skin, atopic eczema, and predisposition to malignancy leading to a poor prognosis. Typical findings include highly elevated IgE and eosinophilia. Allogeneic hematopoietic stem cell transplantation (alloHSCT) is indicated as the only curative treatment option. We present a patient with advanced disease undergoing alloHSCT at the age of 11 years after individualized pre-treatment using dupilumab and rituximab resulting in a decrease in IgE levels and clinical improvement of the skin condition. Additionally, in a review of the literature, we summarize morbidity and outcome in DOCK8-deficient patients older than 8 years of age receiving alloHSCT. Life-threatening infections, malignancy, and disease-related complications with organ damage pre-transplant are challenging in older DOCK8-deficient patients. The therapeutic role of dupilumab in DOCK8 deficiency should be evaluated in larger studies.
Collapse
Affiliation(s)
- Sophia Trombello
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
- Children’s Hospital, Heidelberg University Hospital, Heidelberg, Germany
| | - Andrea Jarisch
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Andre Willasch
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Eva Rettinger
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Julia Fekadu-Siebald
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Dirk Holzinger
- Department of Pediatric Hematology-Oncology, University of Duisburg-Essen, Essen, Germany
- Department of Applied Health Sciences, University of Applied Sciences Bochum, Bochum, Germany
| | - Roland Adelmann
- Department for Children and Adolescents Medicine, Hospital Oberberg, Gummersbach, Germany
| | - Peter Bader
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| | - Shahrzad Bakhtiar
- Division for Stem Cell Transplantation and Immunology, Department for Pediatrics, Goethe University Hospital Frankfurt, Frankfurt am Main, Germany
| |
Collapse
|
4
|
Freeman AF, Gonzalez CE, Yates B, Cole K, Little L, Flannelly E, Steinberg SM, Mo G, Piette N, Hughes TE, Cuellar-Rodriguez J, Gea-Banacloche J, Heller T, Hammoud DA, Holland SM, Kong HH, Young FD, Jing H, Kayaoglu B, Su HC, Pai SY, Hickstein DD, Shah NN. Hematopoietic cell transplantation for DOCK8 deficiency: Results from a prospective clinical trial. J Allergy Clin Immunol 2025; 155:176-187. [PMID: 39233015 DOI: 10.1016/j.jaci.2024.08.021] [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: 02/22/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND DOCK8 deficiency is a primary immunodeficiency in which allogeneic hematopoietic cell transplantation (HCT) represents the only known cure. We tested the ability of a busulfan-based regimen to achieve reliable engraftment and high levels of donor chimerism with acceptable toxicity in a prospective clinical trial in DOCK8 deficiency. OBJECTIVES To both evaluate the ability of HCT to reverse the clinical phenotype and to correct the immunologic abnormalities by 1 year post HCT. METHODS We conducted a prospective HCT trial for recipients with DOCK8 deficiency. Subjects were recruited from October 5, 2010, to December 30, 2022. Donor sources included fully matched related and unrelated donors and haploidentical donors. The reduced toxicity, myeloablative conditioning regimen contained no serotherapy. Graft-versus-host disease (GVHD) prophylaxis included either a calcineurin inhibitor with methotrexate or post-HCT cyclophosphamide (PT/Cy) followed by tacrolimus and mycophenolate mofetil. The trial was later amended to study PT/Cy in all patients. (Pilot Study of Reduced-Intensity Hematopoietic Stem Cell Transplant of DOCK8 [NCT01176006].) RESULTS: Thirty-six subjects, both children and adults (median age 16.4 years), underwent HCT for DOCK8 deficiency. Most patients, 33 of 36 (92%), achieved full (≥98%) donor chimerism in whole blood as early as day +30. With a median potential follow-up of 7.4 years, 29 (80.6%) were alive with no evidence of new DOCK8 deficiency-related complications. PT/Cy was effective in reducing the risk of acute GVHD in patients who had received matched unrelated donor and haploidentical transplants, but it was associated with transient delays in immune-reconstitution and hemorrhagic cystitis. CONCLUSIONS A busulfan-based HCT regimen using PT/Cy for GVHD prophylaxis and a broad range of donor types and hematopoietic cell sources were well tolerated, leading to the reversal of the clinical immunophenotype.
Collapse
Affiliation(s)
- Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Corina E Gonzalez
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md.
| | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | - Kristen Cole
- Transplant and Cell Therapy Program, Clinical Center, Bethesda, Md
| | - Lauren Little
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | - Erin Flannelly
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Seth M Steinberg
- Biostatistics and Data Management Section, National Cancer Institute, Bethesda, Md
| | - George Mo
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| | | | | | - Jennifer Cuellar-Rodriguez
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Juan Gea-Banacloche
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Theo Heller
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Md
| | - Dima A Hammoud
- Center for Infectious Disease Imaging, Radiology and Imaging Sciences, Clinical Center, Bethesda, Md
| | - Steve M Holland
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Heidi H Kong
- Dermatology Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, Md
| | - Fernanda D Young
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Basak Kayaoglu
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| | - Sung-Yun Pai
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Dennis D Hickstein
- Immune-Deficiency Cellular Therapy Program, National Cancer Institute, Bethesda, Md
| | - Nirali N Shah
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Md
| |
Collapse
|
5
|
Chen JS, Lee D, Gowthaman U. T follicular helper cells in food allergy. Curr Opin Immunol 2024; 91:102461. [PMID: 39276414 DOI: 10.1016/j.coi.2024.102461] [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: 06/16/2024] [Revised: 08/21/2024] [Accepted: 08/24/2024] [Indexed: 09/17/2024]
Abstract
T follicular helper (Tfh) cells help direct the production of antibodies by B cells. In addition to promoting antibody responses to vaccination and infection, Tfh cells have been found to mediate antibody production to food antigens. Work over the past decade has delineated the specific phenotypes of Tfh cells that induce antibodies to food while also clarifying the divergent Tfh cell requirement for different food-specific antibody isotypes. Furthermore, Tfh and antibody responses to food can occur at multiple barrier sites - namely, skin, airway, and gut. Depending on the context of food antigen exposure, the immune response to food at these sites can be protective, as in the case of tolerance or immunotherapy, or pathogenic, as in the case of allergy. This review will highlight recent advances in our understanding of how Tfh cells promote antibodies to food as well as future avenues for continued discovery.
Collapse
Affiliation(s)
- Jennifer S Chen
- Department of Internal Medicine, Lankenau Medical Center, Wynnewood, PA, USA
| | - Donguk Lee
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA
| | - Uthaman Gowthaman
- Department of Pathology, University of Massachusetts Chan Medical School, Worcester, MA, USA.
| |
Collapse
|
6
|
Oktelik FB, Wang M, Keles S, Eke Gungor H, Cansever M, Can S, Karakoc-Aydiner E, Baris S, Schmitz-Abe K, Benamar M, Chatila TA. DOCK8 deficiency due to a deep intronic variant in two kindreds with hyper-IgE syndrome. Clin Immunol 2024; 268:110384. [PMID: 39437980 PMCID: PMC11531991 DOI: 10.1016/j.clim.2024.110384] [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: 10/10/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency underlies the majority of cases of patients with autosomal recessive form of the hyper-immunoglobulin E syndrome (HIES). Most DOCK8 mutations involve deletions and splice junction mutations that abrogate protein expression. However, a few patients whose presentation is reminiscent of DOCK8 deficiency have no identifiable mutations. Using Whole Exome Sequencing (WES), we identified a deep intronic homozygous DOCK8 variant located in intron 36 (c.4626 + 76 A > G) in two unrelated patients with features of HIES that resulted in an in-frame 75 base pair intronic sequence insertion in DOCK8 cDNA, resulting in a premature stop codon (p.S1542ins6Ter). This variant resulted in variable decrease in DOCK8 expression that was associated with impaired T cell receptor-triggered actin polymerization, decreased IL-6-induced STAT3 phosphorylation, reduced expression of the Th17 cell markers CCR6 and IL-17, and higher frequencies of GATA3+ T cells indicative of Th2 skewing. Our approach extends the reach of WES in identifying disease-related intronic variants. It highlights the role of non-coding mutations in immunodeficiency disorders, including DOCK8 deficiency, and emphasizes the need to explore these mutations in unexplained inborn errors of immunity.
Collapse
Affiliation(s)
- Fatma Betul Oktelik
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Immunology, Aziz Sancar Institute of Experimental Medicine (Aziz Sancar DETAE), Istanbul University, Istanbul, Turkiye; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Muyun Wang
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA
| | - Sevgi Keles
- Necmettin Erbakan University, Medical Faculty, Department of Pediatric Allergy and Immunology, Konya, Turkiye
| | - Hatice Eke Gungor
- University of Health Sciences, Kayseri City Hospital, Department of Pediatric Allergy and Immunology, Kayseri, Turkiye
| | - Murat Cansever
- University of Health Sciences, Kayseri City Hospital, Department of Pediatric Allergy and Immunology, Kayseri, Turkiye
| | - Salim Can
- Marmara University, School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkiye; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkiye; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkiye
| | - Elif Karakoc-Aydiner
- Marmara University, School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkiye; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkiye; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkiye
| | - Safa Baris
- Marmara University, School of Medicine, Division of Pediatric Allergy and Immunology, Istanbul, Turkiye; Istanbul Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Istanbul, Turkiye; The Isil Berat Barlan Center for Translational Medicine, Istanbul, Turkiye
| | - Klaus Schmitz-Abe
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA; Division of Neonatology, Department of Pediatrics, University of Miami Miller School of Medicine and Jackson Health System, Miami, FL, USA
| | - Mehdi Benamar
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA
| | - Talal A Chatila
- Division of Immunology, Boston Children's Hospital, Boston, MA, USA; Department of Pediatrics, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
7
|
Jiang P, Zhao S, Li X, Hu S, Chen S, Liang Y, Zhang L, Lu L, Fang G, Yang L, Huang Y, Miller H, Guan F, Lei J, Liu C. Dedicator of cytokinesis 8 (DOCK8) mutation impairs the differentiation of helper T cells by regulating the glycolytic pathway of CD4 + T cells. MedComm (Beijing) 2024; 5:e747. [PMID: 39329018 PMCID: PMC11424684 DOI: 10.1002/mco2.747] [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: 04/29/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/28/2024] Open
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency is a primary immunodeficiency disease caused by mutations in exon 45 of the DOCK8 gene. The clinical signs primarily consist of increased serum IgE levels, eczema, repeated skin infections, allergies, and upper respiratory tract infections. Using CRISPR/Cas9 technology, we generated a DOCK8 exon 45 mutation in mice, mirroring the mutation found in patients. The results indicated that DOCK8 mutation impairs peripheral T cell homeostasis, disrupts regulatory T cells (Tregs) development, increases ICOS expression in Tregs within peripheral lymph nodes (pLn), and promotes Th17 cell differentiation within the spleen and pLn. Upon virus infection, DOCK8 mutation CD4+ T cells have a Th2 effector fate. RNA-bulk sequencing data revealed alternations in the mTOR pathway of DOCK8 mutant CD4+ T cells. We observed that DOCK8 mutation upregulates the glycolysis levels in CD4+ T cells, which is related to the Akt/mTOR/S6/HIF-1α pathway. In summary, our research elucidates that DOCK8 regulates the differentiation of helper T cells by modulating the glycolytic pathway in CD4+ T cells, thereby advancing the comprehension and offering potential treatment of diseases in DOCK8-deficient patients.
Collapse
Affiliation(s)
- Panpan Jiang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Siyu Zhao
- Department Immunology School of Medicine Yangtze University Jingzhou China
| | - Xiaoyu Li
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Shiyan Hu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Shuhan Chen
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Yinming Liang
- Center of Disease Model and Immunology Hunan Academy of Chinese Medicine Changsha China
| | - Lichen Zhang
- Laboratory of Genetic Regulators in the Immune System School of Medical Technology Xinxiang Medical University Xinxiang China
| | - Liaoxun Lu
- Laboratory of Genetic Regulators in the Immune System School of Medical Technology Xinxiang Medical University Xinxiang China
| | - Guofeng Fang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Lu Yang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Yanmei Huang
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Heather Miller
- Cytek Biosciences R&D Clinical Reagents Fremont California USA
| | - Fei Guan
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Jiahui Lei
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| | - Chaohong Liu
- Department of Pathogen Biology School of Basic Medicine Tongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases Huazhong University of Science and Technology Wuhan China
| |
Collapse
|
8
|
Huynh A, Gray PE, Sullivan A, Mackie J, Guerin A, Rao G, Pathmanandavel K, Mina ED, Hollway G, Hobbs M, Enthoven K, O'Young P, McManus S, Wainwright LH, Higgins M, Noon F, Wong M, Bastard P, Zhang Q, Casanova JL, Hsiao KC, Pinzon-Charry A, Ma CS, Tangye SG. A Novel Case of IFNAR1 Deficiency Identified a Common Canonical Splice Site Variant in DOCK8 in Western Polynesia: The Importance of Validating Variants of Unknown Significance in Under-Represented Ancestries. J Clin Immunol 2024; 44:170. [PMID: 39098944 PMCID: PMC11298505 DOI: 10.1007/s10875-024-01774-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 07/23/2024] [Indexed: 08/06/2024]
Abstract
Advanced genomic technologies such as whole exome or whole genome sequencing have improved diagnoses and disease outcomes for individuals with genetic diseases. Yet, variants of unknown significance (VUS) require rigorous validation to establish disease causality or modification, or to exclude them from further analysis. Here, we describe a young individual of Polynesian ancestry who in the first 13 mo of life presented with SARS-CoV-2 pneumonia, severe enterovirus meningitis and adenovirus gastroenteritis, and severe adverse reaction to MMR vaccination. Genomic analysis identified a previously reported pathogenic homozygous variant in IFNAR1 (c.1156G > T, p.Glu386* LOF), which is common in Western Polynesia. Moreover, a new and putatively deleterious canonical splice site variant in DOCK8 was also found in homozygosity (c.3234 + 2T > C). This DOCK8 variant is common in Polynesians and other under-represented ancestries in large genomic databases. Despite in silico bioinformatic predictions, extensive in vitro and ex vivo analysis revealed the DOCK8 variant likely be neutral. Thus, our study reports a novel case of IFNAR1 deficiency, but also highlights the importance of functional validation of VUS, including those predicted to be deleterious, and the pressing need to expand our knowledge of the genomic architecture and landscape of under-represented populations and ancestries.
Collapse
Affiliation(s)
- Aimee Huynh
- Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, QLD, 4101, Australia
| | - Paul E Gray
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- School Medicine, Western Sydney University, Penrith, NSW, Australia
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Randwick, Australia
| | - Anna Sullivan
- Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, QLD, 4101, Australia
- Clinical Immunogenomics Research Consortium, Australasia, Australia
| | - Joseph Mackie
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Antoine Guerin
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Geetha Rao
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Karrnan Pathmanandavel
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Erika Della Mina
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Georgina Hollway
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Matthew Hobbs
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Karen Enthoven
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Patrick O'Young
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
| | - Sam McManus
- Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | | | | | - Fallon Noon
- Genetic Health Queensland, Brisbane, Australia
| | - Melanie Wong
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Department of Allergy and Immunology, The Children's Hospital at Westmead, Sydney, NSW, Australia
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- University of Paris, Imagine Institute, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
| | - Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, USA
- University of Paris, Imagine Institute, Paris, France
- Department of Pediatrics, Necker Hospital for Sick Children, AP-HP, Paris, France
- Howard Hughes Medical Institute, New York, USA
| | - Kuang-Chih Hsiao
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Starship Child Health, Auckland, New Zealand
- Department of Paediatrics, University of Auckland, Auckland, New Zealand
| | - Alberto Pinzon-Charry
- Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, QLD, 4101, Australia
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Griffith University and University of Queensland, Queensland, Australia
| | - Cindy S Ma
- Clinical Immunogenomics Research Consortium, Australasia, Australia
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia
| | - Stuart G Tangye
- Clinical Immunogenomics Research Consortium, Australasia, Australia.
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, NSW, 2010, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| |
Collapse
|
9
|
Ewing A, Madan RP. Viral infections and inborn errors of immunity. Curr Opin Infect Dis 2024; 37:227-231. [PMID: 38747352 DOI: 10.1097/qco.0000000000001021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/28/2024]
Abstract
PURPOSE OF REVIEW The purpose of this focused review is to discuss unusual presentations of viral infections in the context of specific inborn errors of immunity. We will discuss hyper immunoglobulin E (IgE) syndromes, epidermodysplasia verruciformis, and X-linked agammaglobulinemia as examples of inborn errors of immunity associated with specific presentations of viral infection and disease. RECENT FINDINGS Advances in both genetic and viral diagnostics have broadened our understanding of viral pathogenesis in the setting of immune dysfunction and the variable phenotype of inborn errors of immunity. Dedicator of cytokinesis 8 (DOCK8) deficiency is now recognized as an inborn error of immunity within the hyper IgE syndrome phenotype and is associated with unusually aggressive cutaneous disease caused by herpes simplex and other viruses. Studies of patients with epidermodysplasia verruciformis have proven that rarely detected human papillomavirus subtypes may cause malignancy in the absence of adequate host defenses. Finally, patients with X-linked agammaglobulinemia may remain at risk for severe and chronic viral infections, even as immune globulin supplementation reduces the risk of bacterial infection. SUMMARY Susceptibility to viral infections in patients with inborn errors of immunity is conferred by specific, molecular defects. Recurrent, severe, or otherwise unusual presentations of viral disease should prompt investigation for an underlying genetic defect.
Collapse
Affiliation(s)
- Anne Ewing
- Department of Pediatrics, NYU Grossman School of Medicine
| | - Rebecca Pellett Madan
- Department of Pediatrics, NYU Grossman School of Medicine
- NYU Langone Transplant Institute, New York, New York, USA
| |
Collapse
|
10
|
Sun Z, Wei R, Pan C, Ni C, Zhang X, Guan W, Cheng R, Gu Y, Yu H, He K, Zhang Z, Yu X, Yao Z. Successfully treated with siltuximab and prednisone in a 7-year-old girl with DOCK8-deficiency presenting as recurrent wart-like lesions: a case report. Front Immunol 2024; 15:1414573. [PMID: 39044832 PMCID: PMC11263070 DOI: 10.3389/fimmu.2024.1414573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/27/2024] [Indexed: 07/25/2024] Open
Abstract
Dedicator of cytokinesis 8 (DOCK8) deficiency represents a primary immunodeficiency with a wide range of clinical symptoms, including recurrent infections, atopy, and increased malignancy risk. This study presents a case of a 6-year-old girl with DOCK8 deficiency, characterized by severe, treatment-resistant herpetic infections who was successfully treated with siltuximab and glucocorticoids. The successful use of siltuximab in achieving remission highlights the pivotal role of interleukin-6 (IL-6) in DOCK8 deficiency pathogenesis and suggests that IL-6 modulation can be critical in managing DOCK8 deficiency-related viral infections, which may inform future therapeutic strategies for DOCK8 deficiency and similar immunodeficiencies.
Collapse
Affiliation(s)
- Zhe Sun
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruoqu Wei
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Chaolan Pan
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Cheng Ni
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xue Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Wenbin Guan
- Department of Pathology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Ruhong Cheng
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Gu
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Hong Yu
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Kejun He
- Department of Pediatric Hematology and Oncology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhen Zhang
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Xia Yu
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Zhirong Yao
- Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai, China
- Department of Allergy, Dermatology Center, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| |
Collapse
|
11
|
Liquidano-Perez E, Maza-Ramos G, Perez Arias BA, Lugo Reyes SO, Barragan Arevalo T, Solorzano-Morales SA, Venegas Montoya E, Staines-Boone AT, Guzmán Cotaya R, Okada S, Picard C, Patin E, Ramirez-Uribe N, Bustamante-Ogando JC, Scheffler-Mendoza SC, Yamazaki-Nakashimada MA, Saez-de-Ocariz M, Espinosa Padilla SE, Gonzalez-Serrano ME. Clinical, immunological, and genetic description of a Mexican cohort of patients with DOCK8 deficiency. Pediatr Allergy Immunol 2024; 35:e14073. [PMID: 38351896 DOI: 10.1111/pai.14073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 12/16/2023] [Accepted: 01/07/2024] [Indexed: 02/16/2024]
Abstract
PURPOSE We aimed to describe the clinical, immunological, and genetic features of patients with DOCK8 deficiency (DOCK8-Def) in a tertiary care center for children. METHODS Retrospective chart review of patients' clinical, immunological, and genetic characteristics with DOCK8-Def. Genetic analysis was performed with targeted- or whole-exome sequencing; we also assessed DOCK8 protein expression and a lymphoproliferation assay and analyzed survival by the Kaplan-Meier method. RESULTS We described 11 patients from 8 unrelated kindreds. The median age at symptoms' onset was 10 months (range 1-54 months). The median follow-up time was 53.4 months (4.8-118.8). All patients presented eczema and recurrent sinopulmonary and cutaneous infections. Besides those symptoms, the most frequent manifestations were bronchiectases (8/11), food allergies (6/11), and severe infections (6/11). Infrequent characteristics were detection of CMV in bronchial lavage, C. parvum-driven sclerosing cholangitis, Takayasu vasculitis, neurological syndromes, pulmonary tuberculosis, and lymphomatoid granulomatosis. CONCLUSION DOCK8-Def has a broad spectrum of manifestations, including allergy, autoimmunity, inflammation, infection, and cancer. The hallmark of this inborn error of immunity is IEI-associated eczema with eosinophilia and increased IgE. Here, we report six new mutations causing human DOCK8 deficiency and symptoms previously unrecognized to occur in DOCK8-Def. Therefore, an early diagnosis of DOCK8-Def is essential to facilitate an adequate treatment such as HSCT.
Collapse
Affiliation(s)
| | | | | | | | - Tania Barragan Arevalo
- Department of Genetics, Institute of Ophthalmology "Conde de Valenciana", Mexico City, Mexico
| | | | - Edna Venegas Montoya
- Highly Specialized Medical Unit 25, Mexican Social Security Institute, Torreón, Mexico
| | | | | | | | | | | | - Nideshda Ramirez-Uribe
- Hematopoietic Stem Cell Transplantation Unit, National Institute of Pediatrics, Mexico City, Mexico
| | | | | | | | | | | | | |
Collapse
|
12
|
Ruan J, Cui X, Yan H, Jia C, Ou T, Shang Z. Expression profiles of circular RNAs and interaction networks of competing endogenous RNAs in neurogenic bladder of rats following suprasacral spinal cord injury. PeerJ 2023; 11:e16042. [PMID: 37744239 PMCID: PMC10512963 DOI: 10.7717/peerj.16042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Accepted: 08/15/2023] [Indexed: 09/26/2023] Open
Abstract
Background Neurogenic bladder (NB) following suprasacral spinal cord injury (SSCI) is an interstitial disease with the structural remodeling of bladder tissue and matrix over-deposition. Circular RNAs (circRNAs) are involved in fibrotic disease development through their post-transcriptional regulatory functions. This study aimed to use transcriptome high-throughput sequencing to investigate the process of NB and bladder fibrosis after SSCI. Methods Spinal cord transection at the T10-T11 level was used to construct the SSCI model in rats (10-week-old female Wistar rats, weighing 200 ± 20 g). The bladders were collected without (sham group) and with (SSCI 1-3 groups) NB status. Morphological examination was conducted to assess the extent of bladder fibrosis. Additionally, RNA sequencing was utilized to determine mRNAs and circRNAs expression patterns. The dynamic changes of differentially expressed mRNAs (DEMs) and circRNAs (DECs) in different periods of SSCI were further analyzed. Results Bladder weight, smooth muscle cell hypertrophy, and extracellular matrix gradually increased after SSCI. Compared with the sham group, 3,255 DEMs and 1,339 DECs, 3,449 DEMs and 1,324 DECs, 884 DEMs, and 1,151 DECs were detected in the SSCI 1-3 groups, respectively. Specifically, circRNA3621, circRNA0617, circRNA0586, and circRNA4426 were significant DECs common to SSCI 1-3 groups compared with the sham group. Moreover, Gene Ontology (GO) enrichment suggested that inflammatory and chronic inflammatory responses were the key events in NB progression following SSCI. Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways enrichment associated with the "Chemokine signaling pathway", the "IL-17 signaling pathway", and the "TGF-beta signaling pathway" suggests their potential involvement in regulating biological processes. The circRNA-miRNA-mRNA interaction networks of DECs revealed rno-circ-2239 (micu2) as the largest node, indicating that the rno-circ-2239-miRNA-mRNA-mediated network may play a critical role in the pathogenesis of SSCI-induced NB. Conclusions This study offers a comprehensive outlook on the possible roles of DEMs and DECs in bladder fibrosis and NB progression following SSCI. These findings have the potential to serve as novel biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Jimeng Ruan
- Department of Urology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Xin Cui
- Department of Urology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Hao Yan
- Department of Urology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Chunsong Jia
- Department of Urology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Tongwen Ou
- Department of Urology, Xuanwu Hospital Capital Medical University, Beijing, China
| | - Zhenhua Shang
- Department of Urology, Xuanwu Hospital Capital Medical University, Beijing, China
| |
Collapse
|
13
|
Ner-Gaon H, Peleg R, Gazit R, Reiner-Benaim A, Shay T. Mapping the splicing landscape of the human immune system. Front Immunol 2023; 14:1116392. [PMID: 37711610 PMCID: PMC10499523 DOI: 10.3389/fimmu.2023.1116392] [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: 12/05/2022] [Accepted: 08/14/2023] [Indexed: 09/16/2023] Open
Abstract
Most human genes code for more than one transcript. Different ratios of transcripts of the same gene can be found in different cell types or states, indicating differential use of transcription start sites or differential splicing. Such differential transcript use (DTUs) events provide an additional layer of regulation and protein diversity. With the exceptions of PTPRC and CIITA, there are very few reported cases of DTU events in the immune system. To rigorously map DTUs between different human immune cell types, we leveraged four publicly available RNA sequencing datasets. We identified 282 DTU events between five human healthy immune cell types that appear in at least two datasets. The patterns of the DTU events were mostly cell-type-specific or lineage-specific, in the context of the five cell types tested. DTUs correlated with the expression pattern of potential regulators, namely, splicing factors and transcription factors. Of the several immune related conditions studied, only sepsis affected the splicing of more than a few genes and only in innate immune cells. Taken together, we map the DTUs landscape in human peripheral blood immune cell types, and present hundreds of genes whose transcript use changes between cell types or upon activation.
Collapse
Affiliation(s)
- Hadas Ner-Gaon
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Ronnie Peleg
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Roi Gazit
- The Shraga Segal Department of Microbiology, Immunology and Genetics, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Anat Reiner-Benaim
- Department of Epidemiology, Biostatistics and Community Health Sciences, School of Public Health, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tal Shay
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| |
Collapse
|
14
|
Tangye SG, Meyts I. DOCK11 and Immune Disease. N Engl J Med 2023; 389:563-567. [PMID: 37590454 DOI: 10.1056/nejme2305431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/19/2023]
Affiliation(s)
- Stuart G Tangye
- From the Garvan Institute of Medical Research, Darlinghurst, NSW, and the School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Randwick, NSW - both in Australia (S.G.T.); and the Department of Immunology and Microbiology, Laboratory for Inborn Errors of Immunity, and the Department of Pediatrics, University Hospitals, Leuven, and Fonds voor Wetenschappelijk Onderzoek Vlaanderen, Brussels - all in Belgium (I.M.)
| | - Isabelle Meyts
- From the Garvan Institute of Medical Research, Darlinghurst, NSW, and the School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales Sydney, Randwick, NSW - both in Australia (S.G.T.); and the Department of Immunology and Microbiology, Laboratory for Inborn Errors of Immunity, and the Department of Pediatrics, University Hospitals, Leuven, and Fonds voor Wetenschappelijk Onderzoek Vlaanderen, Brussels - all in Belgium (I.M.)
| |
Collapse
|
15
|
Tangye SG, Puel A. The Th17/IL-17 Axis and Host Defense Against Fungal Infections. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1624-1634. [PMID: 37116791 DOI: 10.1016/j.jaip.2023.04.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 04/30/2023]
Abstract
Chronic mucocutaneous candidiasis (CMC) was recognized as a primary immunodeficiency in the early 1970s. However, for almost 40 years, its genetic etiology remained unknown. The progressive molecular and cellular description of inborn errors of immunity (IEI) with syndromic CMC pointed toward a possible role of IL-17-mediated immunity in protecting against fungal infection and CMC. Since 2011, novel IEI affecting either the response to or production of IL-17A and/or IL-17F (IL-17A/F) in patients with isolated or syndromic CMC provided formal proof of the pivotal role of the IL-17 axis in mucocutaneous immunity to Candida spp, and, to a lesser extent, to Staphylococcus aureus in humans. In contrast, IL-17-mediated immunity seems largely redundant against other common microbes in humans. In this review, we outline the current knowledge of IEI associated with impaired IL-17A/F-mediated immunity, highlighting our current understanding of the role of IL-17A/F in human immunity.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, NSW, Australia; School of Clinical Medicine, UNSW Faculty of Medicine & Health, Darlinghurst, NSW, Australia.
| | - Anne Puel
- 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; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, the Rockefeller University, New York, NY, USA
| |
Collapse
|
16
|
Ma CS, Freeman AF, Fleisher TA. Inborn Errors of Immunity: A Role for Functional Testing and Flow Cytometry in Aiding Clinical Diagnosis. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2023; 11:1579-1591. [PMID: 37054882 PMCID: PMC10330903 DOI: 10.1016/j.jaip.2023.03.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 03/13/2023] [Accepted: 03/31/2023] [Indexed: 04/15/2023]
Abstract
With the exponential discovery of new inborn errors of immunity (IEI), it is becoming increasingly difficult to differentiate between a number of the more recently defined disorders. This is compounded by the fact that although IEI primarily present with immunodeficiency, the spectrum of disease is broad and often extends to features typical of autoimmunity, autoinflammation, atopic disease, and/or malignancy. Here we use case studies to discuss the laboratory and genetic tests used that ultimately led to the specific diagnoses.
Collapse
Affiliation(s)
- Cindy S Ma
- Immunology Program, Garvan Institute of Medical Research, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia.
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Md
| | - Thomas A Fleisher
- Department of Laboratory Medicine, Clinical Center, National Institutes of Health, Bethesda, Md
| |
Collapse
|
17
|
Liquidano-Pérez E, Maza-Ramos G, Yamazaki-Nakashimada MA, Barragán-Arévalo T, Lugo-Reyes SO, Scheffler-Mendoza S, Espinosa-Padilla SE, González-Serrano ME. [Combined immunodeficiency due to DOCK8 deficiency. State of the art]. REVISTA ALERGIA MÉXICO 2022; 69:31-47. [PMID: 36927749 DOI: 10.29262/ram.v69i1.1104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Combinedimmunodeficiency (CID) due to DOCK8 deficiency is an inborn error of immunity (IBD) characterized by dysfunctional T and B lymphocytes; The spectrum of manifestations includes allergy, autoimmunity, inflammation, predisposition to cancer, and recurrent infections. DOCK8 deficiency can be distinguished from other CIDs or within the spectrum of hyper-IgE syndromes by exhibiting profound susceptibility to viral skin infections, associated skin cancers, and severe food allergies. The 9p24.3 subtelomeric locus where DOCK8 is located includes numerous repetitive sequence elements that predispose to the generation of large germline deletions and recombination-mediated somatic DNA repair. Residual production DOCK8 protein contributes to the variable phenotype of the disease. Severe viral skin infections and varicella-zoster virus (VZV)-associated vasculopathy, reflect an essential role of the DOCK8 protein, which is required to maintain lymphocyte integrity as cells migrate through the tissues. Loss of DOCK8 causes immune deficiencies through other mechanisms, including a cell survival defect. In addition, there are alterations in the response of dendritic cells, which explains susceptibility to virus infection and regulatory T lymphocytes that could help explain autoimmunity in patients. Hematopoietic stem cell transplantation (HSCT) is the only curative treatment; it improves eczema, allergies, and susceptibility to infections.
Collapse
Affiliation(s)
- Eduardo Liquidano-Pérez
- Instituto Nacional de Pediatría, Unidad de Investigación en Inmunodeficiencias, Ciudad de México, México
| | | | | | - Tania Barragán-Arévalo
- Fundación de Asistencia Privada, Instituto de Oftalmología Conde de Valenciana, Departamento de Genética, Ciudad de México, México
| | - Saúl Oswaldo Lugo-Reyes
- Instituto Nacional de Pediatría, Unidad de Investigación en Inmunodeficiencias, Ciudad de México, México
| | | | - Sara Elva Espinosa-Padilla
- Instituto Nacional de Pediatría, Unidad de Investigación en Inmunodeficiencias, Ciudad de México, México
| | | |
Collapse
|
18
|
Liquidano-Perez E, Alva-Chaire ADC, Yamazaki-Nakashimada MA, Pesantez Abril ÁA, Solorzano Morales SA, Ramírez Ristori AG, Barragán Arévalo T, Gonzalez-Serrano ME, Scheffler-Mendoza SC, Rodríguez-Jurado R. Lymphomatoid granulomatosis in a patient with DOCK8 deficiency. Pediatr Allergy Immunol 2022; 33:e13804. [PMID: 35754125 DOI: 10.1111/pai.13804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2022] [Revised: 05/02/2022] [Accepted: 05/06/2022] [Indexed: 11/28/2022]
|
19
|
Hyper-IgE syndrome caused by DOCK8 mutation with a tumour-like lesion of the lip: a case report. Int J Oral Maxillofac Surg 2022; 51:1545-1548. [PMID: 35393256 DOI: 10.1016/j.ijom.2022.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Revised: 02/14/2022] [Accepted: 03/17/2022] [Indexed: 11/22/2022]
Abstract
Autosomal recessive hyper-IgE syndrome caused by DOCK8 gene mutation is an immunodeficiency. However, the presentation of a tumour-like lesion of the lip in autosomal recessive hyper-IgE syndrome has not yet been reported. This article reports the case of a 20-year-old man with autosomal recessive hyper-IgE syndrome who presented with a tumour-like lesion of the lip, and hyperplasia and erosion of the gingiva. The clinical manifestations included coarse face and neck skin, a diffuse tumour-like lesion on the upper lip showing a reddish erosive nodular surface with yellowish-white exudation, erosive buccal mucosa, and severe periodontitis. The swollen gingival and palatal mucosa indicated nodular hyperplasia and redness with pseudomembrane. The patient had a significantly increased peripheral blood eosinophil count and serum IgE level and an abnormal T lymphocyte count. His oral lesions improved markedly after prednisolone acetate use and local symptomatic treatment for 2 years. However, the patient unfortunately died of a cerebral infection 6 months after the oral lesions had resolved. The novel features of the labial tumour-like lesion described here extend our understanding of the manifestations of autosomal recessive hyper-IgE syndrome.
Collapse
|
20
|
Ravendran S, Hernández SS, König S, Bak RO. CRISPR/Cas-Based Gene Editing Strategies for DOCK8 Immunodeficiency Syndrome. Front Genome Ed 2022; 4:793010. [PMID: 35373187 PMCID: PMC8969908 DOI: 10.3389/fgeed.2022.793010] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 02/14/2022] [Indexed: 12/17/2022] Open
Abstract
Defects in the DOCK8 gene causes combined immunodeficiency termed DOCK8 immunodeficiency syndrome (DIDS). DIDS previously belonged to the disease category of autosomal recessive hyper IgE syndrome (AR-HIES) but is now classified as a combined immunodeficiency (CID). This genetic disorder induces early onset of susceptibility to severe recurrent viral and bacterial infections, atopic diseases and malignancy resulting in high morbidity and mortality. This pathological state arises from impairment of actin polymerization and cytoskeletal rearrangement, which induces improper immune cell migration-, survival-, and effector functions. Owing to the severity of the disease, early allogenic hematopoietic stem cell transplantation is recommended even though it is associated with risk of unintended adverse effects, the need for compatible donors, and high expenses. So far, no alternative therapies have been developed, but the monogenic recessive nature of the disease suggests that gene therapy may be applied. The advent of the CRISPR/Cas gene editing system heralds a new era of possibilities in precision gene therapy, and positive results from clinical trials have already suggested that the tool may provide definitive cures for several genetic disorders. Here, we discuss the potential application of different CRISPR/Cas-mediated genetic therapies to correct the DOCK8 gene. Our findings encourage the pursuit of CRISPR/Cas-based gene editing approaches, which may constitute more precise, affordable, and low-risk definitive treatment options for DOCK8 deficiency.
Collapse
Affiliation(s)
| | | | | | - Rasmus O. Bak
- Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
21
|
Gertie JA, Zhang B, Liu EG, Hoyt LR, Yin X, Xu L, Long LL, Soldatenko A, Gowthaman U, Williams A, Eisenbarth SC. Oral anaphylaxis to peanut in a mouse model is associated with gut permeability but not with Tlr4 or Dock8 mutations. J Allergy Clin Immunol 2022; 149:262-274. [PMID: 34051223 PMCID: PMC8626534 DOI: 10.1016/j.jaci.2021.05.015] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 05/06/2021] [Accepted: 05/10/2021] [Indexed: 01/03/2023]
Abstract
BACKGROUND The etiology of food allergy is poorly understood; mouse models are powerful systems to discover immunologic pathways driving allergic disease. C3H/HeJ mice are a widely used model for the study of peanut allergy because, unlike C57BL/6 or BALB/c mice, they are highly susceptible to oral anaphylaxis. However, the immunologic mechanism of this strain's susceptibility is not known. OBJECTIVE We aimed to determine the mechanism underlying the unique susceptibility to anaphylaxis in C3H/HeJ mice. We tested the role of deleterious Toll-like receptor 4 (Tlr4) or dedicator of cytokinesis 8 (Dock8) mutations in this strain because both genes have been associated with food allergy. METHODS We generated C3H/HeJ mice with corrected Dock8 or Tlr4 alleles and sensitized and challenged them with peanut. We then characterized the antibody response to sensitization, anaphylaxis response to both oral and systemic peanut challenge, gut microbiome, and biomarkers of gut permeability. RESULTS In contrast to C3H/HeJ mice, C57BL/6 mice were resistant to anaphylaxis after oral peanut challenge; however, both strains undergo anaphylaxis with intraperitoneal challenge. Restoring Tlr4 or Dock8 function in C3H/HeJ mice did not protect from anaphylaxis. Instead, we discovered enhanced gut permeability resulting in ingested allergens in the bloodstream in C3H/HeJ mice compared to C57BL/6 mice, which correlated with an increased number of goblet cells in the small intestine. CONCLUSIONS Our work highlights the potential importance of gut permeability in driving anaphylaxis to ingested food allergens; it also indicates that genetic loci outside of Tlr4 and Dock8 are responsible for the oral anaphylactic susceptibility of C3H/HeJ mice.
Collapse
Affiliation(s)
- Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Singapore Immunology Network (SIgN), Singapore
| | - Elise G Liu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, Conn
| | - Laura R Hoyt
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Xiangyun Yin
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Lauren L Long
- The Jackson Laboratory for Genomic Medicine, Farmington, Conn
| | - Arielle Soldatenko
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn
| | - Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Department of Pathology, University of Massachusetts Medical School, Worcester, Mass
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Farmington, Conn; Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, Conn.
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, Conn; Department of Immunobiology, Yale University School of Medicine, New Haven, Conn; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, Conn.
| |
Collapse
|
22
|
Tangye SG, Gray PE, Pillay BA, Yap JY, Figgett WA, Reeves J, Kummerfeld SK, Stoddard J, Uzel G, Jing H, Su HC, Campbell DE, Sullivan A, Burnett L, Peake J, Ma CS. Hyper-IgE Syndrome due to an Elusive Novel Intronic Homozygous Variant in DOCK8. J Clin Immunol 2022; 42:119-129. [PMID: 34657245 PMCID: PMC10461790 DOI: 10.1007/s10875-021-01152-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 09/27/2021] [Indexed: 11/29/2022]
Abstract
Rare, biallelic loss-of-function mutations in DOCK8 result in a combined immune deficiency characterized by severe and recurrent cutaneous infections, eczema, allergies, and susceptibility to malignancy, as well as impaired humoral and cellular immunity and hyper-IgE. The advent of next-generation sequencing technologies has enabled the rapid molecular diagnosis of rare monogenic diseases, including inborn errors of immunity. These advances have resulted in the implementation of gene-guided treatments, such as hematopoietic stem cell transplant for DOCK8 deficiency. However, putative disease-causing variants revealed by next-generation sequencing need rigorous validation to demonstrate pathogenicity. Here, we report the eventual diagnosis of DOCK8 deficiency in a consanguineous family due to a novel homozygous intronic deletion variant that caused aberrant exon splicing and subsequent loss of expression of DOCK8 protein. Remarkably, the causative variant was not initially detected by clinical whole-genome sequencing but was subsequently identified and validated by combining advanced genomic analysis, RNA-seq, and flow cytometry. This case highlights the need to adopt multipronged confirmatory approaches to definitively solve complex genetic cases that result from variants outside protein-coding exons and conventional splice sites.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
| | - Paul E Gray
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Department of Immunology and Infectious Diseases, Sydney Children's Hospital, Sydney, New South Wales, Australia
- School of Women's and Children's Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Bethany A Pillay
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Jin Yan Yap
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
| | - William A Figgett
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
| | - John Reeves
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
| | - Sarah K Kummerfeld
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
| | - Jennifer Stoddard
- Immunology Service, Department of Laboratory Medicine, Clinical Center, NIH, Bethesda, MD, USA
| | - Gulbu Uzel
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Helen C Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dianne E Campbell
- Department of Allergy and Immunology, Children's Hospital at Westmead, Westmead, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Anna Sullivan
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Queensland Children's Hospital and University of Queensland, South Brisbane, Queensland, Australia
| | - Leslie Burnett
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Sydney Medical School, University of Sydney, Sydney, New South Wales, Australia
| | - Jane Peake
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia
- Queensland Children's Hospital and University of Queensland, South Brisbane, Queensland, Australia
| | - Cindy S Ma
- Garvan Institute of Medical Research, 384 Victoria St, Darlinghurst, New South Wales, 2010, Australia.
- St Vincent's Clinical School, Faculty of Medicine and Health, UNSW Sydney, Sydney, New South Wales, Australia.
- Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Sydney, New South Wales, Australia.
| |
Collapse
|
23
|
Holland EM, Gonzalez C, Levy E, Valera VA, Chalfin H, Klicka-Skeels J, Yates B, Kleiner DE, Hadigan C, Dave H, Shalabi H, Hickstein DD, Su HC, Grimley M, Freeman AF, Shah NN. Case Report: Fatal Complications of BK Virus-Hemorrhagic Cystitis and Severe Cytokine Release Syndrome Following BK Virus-Specific T-Cells. Front Immunol 2021; 12:801281. [PMID: 34975916 PMCID: PMC8718506 DOI: 10.3389/fimmu.2021.801281] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 12/01/2021] [Indexed: 11/13/2022] Open
Abstract
BK virus (BKV)-hemorrhagic cystitis (HC) is a well-known and rarely fatal complication of hematopoietic stem cell transplantation (HSCT). Treatment for BKV-HC is limited, but virus-specific T-cells (VST) represent a promising therapeutic option feasible for use posttransplant. We report on the case of a 16-year-old male with dedicator of cytokinesis 8 (DOCK8) deficiency who underwent haploidentical HSCT complicated by severe BKV-HC, catastrophic renal hemorrhage, and VST-associated cytokine release syndrome (CRS). Gross hematuria refractory to multiple interventions began with initiation of posttransplant cyclophosphamide (PT/Cy). Complete left renal arterial embolization (day +43) was ultimately indicated to control intractable renal hemorrhage. Subsequent infusion of anti-BK VSTs was complicated by CRS and progressive multiorgan failure, with postmortem analysis confirming diagnosis of hepatic sinusoidal obstruction syndrome (SOS). This case illustrates opportunities for improvement in the management of severe BKV-HC posttransplant while highlighting rare and potentially life-threatening complications of BKV-HC and VST therapy.
Collapse
Affiliation(s)
- Elizabeth M. Holland
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Corina Gonzalez
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
- Immune Deficiency- Cellular Therapy Program, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Elliot Levy
- Radiology and Imaging Sciences, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Vladimir A. Valera
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Heather Chalfin
- Urologic Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | | | - Bonnie Yates
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - David E. Kleiner
- Laboratory of Pathology, National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Colleen Hadigan
- Pediatric Gastroenterology, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Hema Dave
- Pediatric Oncology, Children’s National Medical Center, Washington, DC, United States
| | - Haneen Shalabi
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Dennis D. Hickstein
- Immune Deficiency- Cellular Therapy Program, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| | - Helen C. Su
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Disease, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Michael Grimley
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital, Cincinnati, OH, United States
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institutes of Allergy and Infectious Disease, NIH Clinical Center (CC), Bethesda, MD, United States
| | - Nirali N. Shah
- Pediatric Oncology Branch, Center for Cancer Research (CCR), National Cancer Institute (NCI), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
24
|
Kamnev A, Lacouture C, Fusaro M, Dupré L. Molecular Tuning of Actin Dynamics in Leukocyte Migration as Revealed by Immune-Related Actinopathies. Front Immunol 2021; 12:750537. [PMID: 34867982 PMCID: PMC8634686 DOI: 10.3389/fimmu.2021.750537] [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: 07/30/2021] [Accepted: 10/12/2021] [Indexed: 01/13/2023] Open
Abstract
Motility is a crucial activity of immune cells allowing them to patrol tissues as they differentiate, sample or exchange information, and execute their effector functions. Although all immune cells are highly migratory, each subset is endowed with very distinct motility patterns in accordance with functional specification. Furthermore individual immune cell subsets adapt their motility behaviour to the surrounding tissue environment. This review focuses on how the generation and adaptation of diversified motility patterns in immune cells is sustained by actin cytoskeleton dynamics. In particular, we review the knowledge gained through the study of inborn errors of immunity (IEI) related to actin defects. Such pathologies are unique models that help us to uncover the contribution of individual actin regulators to the migration of immune cells in the context of their development and function.
Collapse
Affiliation(s)
- Anton Kamnev
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria
| | - Claire Lacouture
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France.,Laboratoire De Physique Théorique, IRSAMC, Université De Toulouse (UPS), CNRS, Toulouse, France
| | - Mathieu Fusaro
- Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| | - Loïc Dupré
- Ludwig Boltzmann Institute for Rare and Undiagnosed Diseases, Vienna, Austria.,Department of Dermatology, Medical University of Vienna, Vienna, Austria.,Toulouse Institute for Infectious and Inflammatory Diseases (INFINITy), INSERM, CNRS, Toulouse III Paul Sabatier University, Toulouse, France
| |
Collapse
|
25
|
Inborn errors of immunity manifesting as atopic disorders. J Allergy Clin Immunol 2021; 148:1130-1139. [PMID: 34428518 DOI: 10.1016/j.jaci.2021.08.008] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/23/2021] [Accepted: 08/02/2021] [Indexed: 01/29/2023]
Abstract
Inborn errors of immunity are traditionally best known for enhancing susceptibility to infections. However, allergic inflammation, among other types of immune dysregulation, occurs frequently in patients with inborn errors of immunity. As such, the term primary atopic disorders (PADs) was recently coined to describe the group of heritable monogenic allergic disorders. It is becoming increasingly important for clinicians to recognize that allergic diseases such as food allergy, atopic dermatitis, and allergic asthma are expressions of misdirected immunity, and in patients who present with severe, early-onset, or coexisting allergic conditions, these can be indications of an underlying PAD. Identifying monogenic allergic disease through next-generation sequencing can dramatically improve outcomes by allowing the use of precision-based therapy targeting the patient's underlying molecular defect. It is therefore imperative that clinicians recognize PADs to be able to provide informed therapeutic options and improve patient outcomes. Here, we summarize the clinical features commonly seen with each of the currently known PADs, identify clinical warning signs that warrant assessment for PADs, and lastly, discuss the benefits of timely diagnosis and management of these conditions.
Collapse
|
26
|
Common Variable Immunodeficiency and Other Immunodeficiency Syndromes in Bronchiectasis. Semin Respir Crit Care Med 2021; 42:525-536. [PMID: 34261177 DOI: 10.1055/s-0041-1730893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Immunodeficiency represents a vast number of diseases and syndromes. Both primary and secondary forms of immunodeficiency are important contributors to the development of bronchiectasis. Primary immune deficiencies, in particular, are increasingly identified and defined as contributors. Specific immune deficiencies that are closely associated with bronchiectasis and as discussed in this article are common variable immunodeficiency, specific antibody deficiency, immunodeficiencies involving immunoglobulin E, DOCK8 immunodeficiency, phosphoglucomutase 3 deficiency, activated phosphoinositide 3-kinase delta syndrome, and X-linked agammaglobulinemia. Each of these primary immune deficiencies has unique nuances. Vigilance for these unique signs and symptoms is likely to improve recognition of specific immunodeficiency in the idiopathic bronchiectasis patient. Secondary forms of immunodeficiency occur as a result of a separate disease process. Graft versus host disease, malignancy, and human immunodeficiency virus are three classic examples discussed in this article. An awareness of the potential for these disease settings to lead to bronchiectasis is necessary to optimize patient care. With understanding and mindfulness toward the intricate relationship between bronchiectasis and immunodeficiency, there is an opportunity to elucidate pathophysiologic underpinnings between these two syndromes.
Collapse
|
27
|
Matsubara K, Kunimura K, Yamane N, Aihara R, Sakurai T, Sakata D, Uruno T, Fukui Y. DOCK8 deficiency causes a skewing to type 2 immunity in the gut with expansion of group 2 innate lymphoid cells. Biochem Biophys Res Commun 2021; 559:135-140. [PMID: 33940384 DOI: 10.1016/j.bbrc.2021.04.094] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Accepted: 04/21/2021] [Indexed: 12/19/2022]
Abstract
Dedicator of cytokinesis 8 (DOCK8) is a guanine nucleotide exchange factor (GEF) for Cdc42. In humans, homozygous or compound heterozygous deletions in DOCK8 cause a combined immunodeficiency characterized by various allergic diseases including food allergies. Although group 2 innate lymphoid cells (ILC2s) contribute to the development of allergic inflammation by producing interleukin (IL)-5 and IL-13, the role of ILC2s in DOCK8 deficiency has not been fully explored. With the use of cytometry by time-of-flight (CyTOF), we performed high-dimensional phenotyping of intestinal immune cells and found that DOCK8-deficient (Dock8-/-) mice exhibited expansion of ILC2s and other leukocytes associated with type 2 immunity in the small intestine. Moreover, IL-5- and IL-13-producing cells markedly increased in Dock8-/- mice, and the majority of them were lineage-negative cells, most likely ILC2s. Intestinal ILC2s expanded when DOCK8 expression was selectively deleted in hematopoietic cells. Importantly, intestinal ILC2 expansion was also observed in Dock8VAGR mice having mutations in the catalytic center of DOCK8, thereby failing to activate Cdc42. Our findings indicate that DOCK8 is a negative regulator of intestinal ILC2s to inhibit their expansion via Cdc42 activation, and that deletion of DOCK8 causes a skewing to type 2 immunity in the gut.
Collapse
Affiliation(s)
- Keisuke Matsubara
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan.
| | - Nana Yamane
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Ryosuke Aihara
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Tetsuya Sakurai
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Daiji Sakata
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka, 812-8582, Japan
| |
Collapse
|
28
|
Ollech A, Mashiah J, Lev A, Simon AJ, Somech R, Adam E, Barzilai A, Hagin D, Greenberger S. Treatment options for DOCK8 deficiency-related severe dermatitis. J Dermatol 2021; 48:1386-1393. [PMID: 34043252 DOI: 10.1111/1346-8138.15955] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 04/14/2021] [Accepted: 04/19/2021] [Indexed: 11/28/2022]
Abstract
BACKGROUND Cutaneous manifestations of dedicator of cytokinesis 8 gene (DOCK8) deficiency, a combined type of T and B cell immunodeficiency, previously designated as autosomal recessive hyper IgE syndrome, includes dermatitis and skin infections. There are limited treatment options for dermatitis related to the syndrome. OBJECTIVE To describe a cohort of patients with DOCK8 deficiency with a focus on the treatment of their cutaneous manifestations. METHODS A retrospective study on all children with the genetic diagnosis of DOCK8 deficiency treated at the Sheba Medical Center between 1/1/2003 and 1/1/2021 was preformed. Collected data included: demographic features, family history, laboratory, genetic testing, skin manifestations, treatment, and disease course. Description of two cases of severe recalcitrant dermatitis treated with dupilumab is detailed. RESULTS Nine children with a genetic diagnosis of DOCK8 deficiency were included, of whom six were girls (66%) with a median age of 8.5 (±2.2 SD) years. The median age at diagnosis was 2.8 (±2.6 SD) years. Six patients were born to consanguineous parents. Five out of six patients who received hematopoietic stem cell transplantation (HSCT) had a complete response, and one was recently transplanted. Of note, two patients, while awaiting HSCT, were treated with dupilumab for their severe dermatitis resulting in a marked improvement of the cutaneous manifestations and pruritus. CONCLUSIONS Hematopoietic stem cell transplantation is the gold standard and most effective therapy for patients with DOCK8 deficiency. Dupilumab, a biological therapy indicated for atopic dermatitis and other Th2 derived dermatoses, is an excellent option for dermatitis in patients with DOCK8 deficiency and can be used as a bridge before HSCT. Larger studies are needed to confirm this observation.
Collapse
Affiliation(s)
- Ayelet Ollech
- Department of Dermatology, Pediatric Dermatology Service, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Jacob Mashiah
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Dermatology and Venereology, Pediatric Dermatology Unit, Dana Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - Atar Lev
- Department of Pediatrics, Immunodeficiency Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Amos J Simon
- Department of Pediatrics, Immunodeficiency Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Raz Somech
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Department of Pediatrics, Immunodeficiency Unit, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel
| | - Etai Adam
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Division of Pediatric Hematology and Oncology, The Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Aviv Barzilai
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Dermatology and Venereology, Pediatric Dermatology Unit, Dana Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel
| | - David Hagin
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Dermatology and Venereology, Pediatric Dermatology Unit, Dana Children's Hospital, Tel-Aviv Sourasky Medical Center, Tel-Aviv, Israel.,Allergy and Clinical Immunology Unit, Department of Medicine, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Shoshana Greenberger
- Department of Dermatology, Pediatric Dermatology Service, Sheba Medical Center, Tel-Hashomer, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
29
|
Yousefnezhad S, Gharesouran J, Ghafouri-Fard S, Hosseinzadeh H, Ahmadian-Heris J, Jafari-Rouhi AH, Taheri M, Rezazadeh M. DOCK8-related Immunodeficiency Syndrome (DIDS): Report of Novel Mutations in Iranian Patients. J Mol Neurosci 2021; 71:2456-2461. [PMID: 33948880 DOI: 10.1007/s12031-021-01843-5] [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: 03/23/2021] [Accepted: 04/12/2021] [Indexed: 10/21/2022]
Abstract
DOCK8 immunodeficiency syndrome (DIDS) is a rare autosomal recessive (AR) disorder characterized by elevated serum IgE levels, eosinophilia, recurrent cutaneous infections, severe eczema, and sinopulmonary and gastrointestinal infections. This syndrome is a multisystem disease that is associated with both immune deficiency and neurological complications. In this study, we describe the clinical characteristics of two Iranian patients with DOCK8 deficiency and propose possible mechanisms for this condition. By using whole exome sequencing (WES), we identified two novel mutations, namely c.3233_3234del AG (p.Q1078fs) in exon 6 and a large deletion with 94 kb (c.405-3231 deletion, p.K135fs), in these two patients. These variations are confirmed with Sanger sequencing and CGH array. Subsequent co-segregation analysis is performed to identify inheritance patterns. Both patients were homozygote and their parents were heterozygote for the variations. For further investigation, prediction tools were applied to identify the pathogenicity of the variations and also for modeling the truncated proteins. The patients did not show neurological abnormalities associated with a deficiency of the N terminal region of DOCK8. The absence of neurological complications in the first patient is justifiable due to the maintenance of the proline-rich region in DOCK8, but for the second patient with expanded deletion which is almost like null DOCK8 protein, it is not presumable, pointing to the fact that the C terminal region of the protein might have functions in the proliferation and migration neurons in the peripheral nervous system. Alternatively, it is possible that neurological abnormalities follow an age-dependent pattern, leading to the appearance of related symptoms later in life. Further multiple functional studies are needed to model different identified variants in animal models to confirm our results and suggest possible mechanisms associated with DOCK8 deficiency in this study.
Collapse
Affiliation(s)
| | - Jalal Gharesouran
- Molecular Genetics Division, GMG center, Tabriz, Iran.,Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Soudeh Ghafouri-Fard
- Department of Medical Genetics, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hassan Hosseinzadeh
- Molecular Genetics Division, GMG center, Tabriz, Iran.,Department of Allergy and Clinical Immunology, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Javad Ahmadian-Heris
- Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mohammad Taheri
- Skull Base Research Center, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Rezazadeh
- Division of Medical Genetics, Tabriz Children's Hospital, Tabriz University of Medical Sciences, Tabriz, Iran. .,Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
30
|
Sakurai T, Kukimoto-Niino M, Kunimura K, Yamane N, Sakata D, Aihara R, Yasuda T, Yokoyama S, Shirouzu M, Fukui Y, Uruno T. A conserved PI(4,5)P2-binding domain is critical for immune regulatory function of DOCK8. Life Sci Alliance 2021; 4:4/4/e202000873. [PMID: 33574036 PMCID: PMC7893821 DOI: 10.26508/lsa.202000873] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 01/20/2021] [Accepted: 01/20/2021] [Indexed: 01/05/2023] Open
Abstract
This study uncovers a critical role of DOCK8 in coupling PI(4,5)P2 signaling with Cdc42 activation through its DHR-1 domain during interstitial leukocyte migration. DOCK8 is a Cdc42-specific guanine-nucleotide exchange factor that is essential for development and functions of various subsets of leukocytes in innate and acquired immune responses. Although DOCK8 plays a critical role in spatial control of Cdc42 activity during interstitial leukocyte migration, the mechanism remains unclear. We show that the DOCK homology region (DHR)-1 domain of DOCK8 binds specifically to phosphatidylinositol 4,5-bisphosphate (PI(4,5)P2) and is required for its recruitment to the plasma membrane. Structural and biochemical analyses reveal that DOCK8 DHR-1 domain consists of a C2 domain-like core with loops creating the upper surface pocket, where three basic residues are located for stereospecific recognition of phosphoinositides. Substitution of the two basic residues, K576 and R581, with alanine abolished PI(4,5)P2 binding in vitro, ablated the ability of DOCK8 to activate Cdc42 and support leukocyte migration in three-dimensional collagen gels. Dendritic cells carrying the mutation exhibited defective interstitial migration in vivo. Thus, our study uncovers a critical role of DOCK8 in coupling PI(4,5)P2 signaling with Cdc42 activation for immune regulation.
Collapse
Affiliation(s)
- Tetsuya Sakurai
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Mutsuko Kukimoto-Niino
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Nana Yamane
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Daiji Sakata
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Ryosuke Aihara
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yasuda
- Division of Immunology and Genome Biology, Department of Molecular and Structural Biology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Shigeyuki Yokoyama
- RIKEN Cluster for Science, Technology and Innovation Hub, Yokohama, Japan
| | - Mikako Shirouzu
- Laboratory for Protein Functional and Structural Biology, RIKEN Center for Biosystems Dynamics Research, Yokohama, Kanagawa, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| |
Collapse
|
31
|
Randall KL, Law HD, Ziolkowski AF, Wirasinha RC, Goodnow CC, Daley SR. DOCK8 deficiency diminishes thymic T-regulatory cell development but not thymic deletion. Clin Transl Immunology 2021; 10:e1236. [PMID: 33437483 PMCID: PMC7790591 DOI: 10.1002/cti2.1236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Revised: 12/16/2020] [Accepted: 12/16/2020] [Indexed: 01/02/2023] Open
Abstract
Objective To define the effect of DOCK8 deficiency on thymic tolerance in mice. Methods Thymocytes from wild‐type (Dock8+/+) and DOCK8‐deficient (Dock8pri/pri) mice were examined by flow cytometry. Some mice had transgenic expression of the BCL2 anti‐apoptotic protein in haemopoietic cells. Some mice expressed the transgenic 3A9 T‐cell receptor (TCR), which triggers thymocyte deletion in mice also expressing hen egg lysozyme under the insulin promoter. Results In Dock8pr/pri mice, the proportion of thymocytes induced to acquire tolerance at the immature CCR7− stage was normal. Deletion of strongly self‐reactive CD4+ thymocytes occurred efficiently in Dock8pri/pri mice in a TCR‐transgenic model that requires self‐antigen transfer from epithelial cells to bone marrow (BM)‐derived antigen‐presenting cells. Thymic Foxp3+ T‐regulatory cells (TREG) and Helios+ Foxp3− TREG precursors were decreased in Dock8pri/pri mice, including when apoptosis was inhibited by BCL2 transgene expression. Dock8pri/pri thymic TREG expressed CD25 and CTLA‐4 at normal levels. The results suggest that DOCK8 deficiency does not affect the function of BM‐derived antigen‐presenting cells in the thymus, the TCR self‐reactivity threshold that activates tolerance mechanisms in thymocytes or the apoptotic deletion of these thymocytes. However, DOCK8 is required to prevent a subset of developing TREG cells from undergoing cell death via a mechanism that is distinct from apoptosis. Conclusion DOCK8 deficiency diminishes TREG development in the thymus without compromising thymocyte deletion.
Collapse
Affiliation(s)
- Katrina L Randall
- Department of Immunology and Infectious Diseases The John Curtin School of Medical Research The Australian National University Canberra ACT Australia.,Australian National University Medical School The Australian National University Canberra ACT Australia
| | - Hsei Di Law
- Department of Immunology and Infectious Diseases The John Curtin School of Medical Research The Australian National University Canberra ACT Australia
| | - Andrew F Ziolkowski
- Department of Immunology and Infectious Diseases The John Curtin School of Medical Research The Australian National University Canberra ACT Australia
| | - Rushika C Wirasinha
- Infection and Immunity Program Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology Monash University Melbourne VIC Australia
| | - Christopher C Goodnow
- Garvan Institute of Medical Research & Cellular Genomics Futures Institute University of New South Wales Sydney NSW Australia
| | - Stephen R Daley
- Infection and Immunity Program Monash Biomedicine Discovery Institute and Department of Biochemistry and Molecular Biology Monash University Melbourne VIC Australia.,Present address: Centre for Immunology and Infection Control School of Biomedical Sciences Faculty of Health Queensland University of Technology Brisbane QLD Australia
| |
Collapse
|
32
|
Zhang Y, Li R, Wang X. Monogenetic causes of fungal disease: recent developments. Curr Opin Microbiol 2020; 58:75-86. [DOI: 10.1016/j.mib.2020.09.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 08/21/2020] [Accepted: 09/08/2020] [Indexed: 01/12/2023]
|
33
|
Schneider C, Shen C, Gopal AA, Douglas T, Forestell B, Kauffman KD, Rogers D, Artusa P, Zhang Q, Jing H, Freeman AF, Barber DL, King IL, Saleh M, Wiseman PW, Su HC, Mandl JN. Migration-induced cell shattering due to DOCK8 deficiency causes a type 2-biased helper T cell response. Nat Immunol 2020; 21:1528-1539. [PMID: 33020661 PMCID: PMC10478007 DOI: 10.1038/s41590-020-0795-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2019] [Accepted: 08/25/2020] [Indexed: 12/30/2022]
Abstract
Mutations that impact immune cell migration and result in immune deficiency illustrate the importance of cell movement in host defense. In humans, loss-of-function mutations in DOCK8, a guanine exchange factor involved in hematopoietic cell migration, lead to immunodeficiency and, paradoxically, allergic disease. Here, we demonstrate that, like humans, Dock8-/- mice have a profound type 2 CD4+ helper T (TH2) cell bias upon pulmonary infection with Cryptococcus neoformans and other non-TH2 stimuli. We found that recruited Dock8-/-CX3CR1+ mononuclear phagocytes are exquisitely sensitive to migration-induced cell shattering, releasing interleukin (IL)-1β that drives granulocyte-macrophage colony-stimulating factor (GM-CSF) production by CD4+ T cells. Blocking IL-1β, GM-CSF or caspase activation eliminated the type-2 skew in mice lacking Dock8. Notably, treatment of infected wild-type mice with apoptotic cells significantly increased GM-CSF production and TH2 cell differentiation. This reveals an important role for cell death in driving type 2 signals during infection, which may have implications for understanding the etiology of type 2 CD4+ T cell responses in allergic disease.
Collapse
Affiliation(s)
- Caitlin Schneider
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Connie Shen
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Angelica A Gopal
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
- Massachusetts General Hospital, Boston, MA, USA
| | - Todd Douglas
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Benjamin Forestell
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
| | - Keith D Kauffman
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Dakota Rogers
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Patricio Artusa
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada
- Department of Physiology, McGill University, Montreal, Quebec, Canada
| | - Qian Zhang
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller University, New York, NY, USA
| | - Huie Jing
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Alexandra F Freeman
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Daniel L Barber
- T Lymphocyte Biology Section, Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Irah L King
- Meakins-Christie Laboratories, Department of Microbiology and Immunology, McGill University Health Centre Research Institute, Montreal, Quebec, Canada
| | - Maya Saleh
- Department of Medicine, McGill University, Montreal, Quebec, Canada
- University of Bordeaux, Bordeaux, France
| | - Paul W Wiseman
- Department of Chemistry and Department of Physics, McGill University, Montreal, Quebec, Canada
| | - Helen C Su
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Judith N Mandl
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada.
- McGill Research Centre for Complex Traits, McGill University, Montreal, Quebec, Canada.
- Department of Physiology, McGill University, Montreal, Quebec, Canada.
| |
Collapse
|
34
|
Abstract
PURPOSE OF REVIEW Fungal infections have steadily increased in incidence, emerging as a significant cause of morbidity and mortality in patients with iatrogenic immunosuppression. Simultaneously, we have witnessed a growing population of newly described inherited immune disorders that have enhanced our understanding of the human immune response against fungi. In the present review, we provide an overview and diagnostic roadmap to inherited disorders which confer susceptibility to superficial and invasive fungal infections. RECENT FINDINGS Inborn errors of fungal immunity encompass a heterogeneous group of disorders, some of which confer fungal infection-specific susceptibility, whereas others also feature broader infection vulnerability and/or noninfectious manifestations. Infections by Candida, Aspergillus, endemic dimorphic fungi, Pneumocystis, and dermatophytes along with their organ-specific presentations provide clinicians with important clues in the assessment of patients with suspected immune defects. SUMMARY The absence of iatrogenic risk factors should raise suspicion for inborn errors of immunity in children and young adults with recurrent or severe fungal diseases. Expeditious diagnosis and prompt initiation of antifungal therapy and management of complications are paramount to achieve remission of fungal disease in the setting of primary immunodeficiency disorders.
Collapse
|
35
|
Freeman AF, Milner JD. The Child with Elevated IgE and Infection Susceptibility. Curr Allergy Asthma Rep 2020; 20:65. [PMID: 32830295 DOI: 10.1007/s11882-020-00964-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
PURPOSE OF REVIEW Over the last 13 years, the genetic etiologies have been determined for multiple conditions causing elevated serum IgE, infection susceptibilities, and variable other features. In this review, we discuss the clinical presentation, laboratory features, and genetics of these diseases caused by mutations in STAT3, DOCK8, PGM3, IL6ST, ZNF341, IL6R, IL6ST, CARD11, and CARD14, with particular focus given to STAT3LOF and DOCK8 deficiency. RECENT FINDINGS Defining the phenotype of each of these syndromes with high IgE and infection susceptibility shows that some have a pronounced connective tissue phenotype such as STAT3LOF and IL6ST deficiency, some have worse viral susceptibility such as DOCK8 deficiency and heterozygous LOF CARD11, and some have more severe allergy and eczema such as LOF CARD14. Studying these distinct but overlapping monogenic diseases will allow a better understanding of more common disease processes such as allergy, eczema, infection susceptibility, scoliosis, and aneurysm.
Collapse
Affiliation(s)
- Alexandra F Freeman
- National Institute of Allergy and Infectious Diseases, National Institutes of Health, NIH Building 10 Room 12C103, 9000 Rockville Pike, Bethesda, MD, 20892, USA.
| | - Joshua D Milner
- Division of Allergy, Immunology and Rheumatology, Columbia University Medical Center, New York-Presbyterian Morgan Stanley Children's Hospital, New York, NY, USA
| |
Collapse
|
36
|
Haskologlu S, Kostel Bal S, Islamoglu C, Aytekin C, Guner S, Sevinc S, Keles S, Kendirli T, Ceylaner S, Dogu F, Ikinciogullari A. Clinical, immunological features and follow up of 20 patients with dedicator of cytokinesis 8 (DOCK8) deficiency. Pediatr Allergy Immunol 2020; 31:515-527. [PMID: 32108967 PMCID: PMC7228270 DOI: 10.1111/pai.13236] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Revised: 01/27/2020] [Accepted: 02/19/2020] [Indexed: 12/19/2022]
Abstract
Biallelic mutations in the dedicator of cytokinesis 8 gene (DOCK8) cause a progressive combined immunodeficiency (CID) characterized by susceptibility to severe viral skin infections, atopic diseases, recurrent respiratory infections, and malignancy. Hematopoietic stem cell transplantation (HSCT) is only curative treatment for the disease. However, there is limited information about long-term outcome of HSCT and its effect to protect against cancer development in DOCK8-deficient patients. In this study, we retrospectively evaluated clinical and immunologic characteristics of 20 DOCK8-deficient patients and outcome of 11 patients who underwent HSCT. We aimed to report the experience of our center and the result of the largest transplantation series of DOCK8 deficiency in our country. Median follow-up time is 71 months (min-max: 16-172) in all patients and 48 months (min-max: 5-84) in transplanted patients. Atopic dermatitis (18/20), recurrent respiratory tract infections (17/20), and food allergy (14/20) were the most frequent clinical manifestations. Failure to thrive (13/20), liver problems (12/20), bronchiectasis (11/20), chronic diarrhea (10/21), and autism spectrum disorders (3/20) were remarkable findings in our series. Elevated IgE level (20/20) and eosinophilia (17/20), low IgM level (15/20), and decreased CD3+ T (10/20) and CD4+ T (11/20) cell count were prominent laboratory findings. HSCT was performed in 11 patients. All patients achieved adequate engraftment and showed improvement in their clinical and immunologic findings. Atopic dermatitis and food allergies improved in all patients, and their dietary restriction was stopped except one patient who was transplanted recently. The frequency of infections was decreased. The overall survival is 91% in HSCT-received patients and 80% in all. HSCT at the earliest possible period with most suitable donor- and patient-specific appropriate conditioning regimen and GvHD prophylaxis is lifesaving for DOCK8 deficiency cases.
Collapse
Affiliation(s)
- Sule Haskologlu
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sevgi Kostel Bal
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Candan Islamoglu
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Caner Aytekin
- Department of Pediatric Immunology, Dr.Sami Ulus Maternity and Children's Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Sukru Guner
- Department of Pediatrics, Division of Immunology and Allergy, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Selin Sevinc
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Sevgi Keles
- Department of Pediatrics, Division of Immunology and Allergy, Necmettin Erbakan University Faculty of Medicine, Konya, Turkey
| | - Tanil Kendirli
- Pediatric Intensive Care Unit, Ankara University School of Medicine, Ankara, Turkey
| | | | - Figen Dogu
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| | - Aydan Ikinciogullari
- Department of Pediatrics, Division of Immunology and Allergy, Ankara University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
37
|
Kunimura K, Uruno T, Fukui Y. DOCK family proteins: key players in immune surveillance mechanisms. Int Immunol 2020; 32:5-15. [PMID: 31630188 PMCID: PMC6949370 DOI: 10.1093/intimm/dxz067] [Citation(s) in RCA: 60] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 10/16/2019] [Indexed: 12/11/2022] Open
Abstract
Dedicator of cytokinesis (DOCK) proteins constitute a family of evolutionarily conserved guanine nucleotide exchange factors (GEFs) for the Rho family of GTPases. Although DOCK family proteins do not contain the Dbl homology domain typically found in other GEFs, they mediate the GTP–GDP exchange reaction through the DOCK homology region-2 (DHR-2) domain. In mammals, this family consists of 11 members, each of which has unique functions depending on the expression pattern and the substrate specificity. For example, DOCK2 is a Rac activator critical for migration and activation of leukocytes, whereas DOCK8 is a Cdc42-specific GEF that regulates interstitial migration of dendritic cells. Identification of DOCK2 and DOCK8 as causative genes for severe combined immunodeficiency syndromes in humans has highlighted their roles in immune surveillance. In addition, the recent discovery of a naturally occurring DOCK2-inhibitory metabolite has uncovered an unexpected mechanism of tissue-specific immune evasion. On the other hand, GEF-independent functions have been shown for DOCK8 in antigen-induced IL-31 production in helper T cells. This review summarizes multifaced functions of DOCK family proteins in the immune system.
Collapse
Affiliation(s)
- Kazufumi Kunimura
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Takehito Uruno
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan.,Research Center for Advanced Immunology, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| | - Yoshinori Fukui
- Division of Immunogenetics, Department of Immunobiology and Neuroscience, Medical Institute of Bioregulation, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan.,Research Center for Advanced Immunology, Kyushu University, Maidashi, Higashi-ku, Fukuoka, Japan
| |
Collapse
|
38
|
Martin MJ, Estravís M, García-Sánchez A, Dávila I, Isidoro-García M, Sanz C. Genetics and Epigenetics of Atopic Dermatitis: An Updated Systematic Review. Genes (Basel) 2020; 11:E442. [PMID: 32325630 PMCID: PMC7231115 DOI: 10.3390/genes11040442] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2020] [Revised: 04/10/2020] [Accepted: 04/15/2020] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND Atopic dermatitis is a common inflammatory skin disorder that affects up to 15-20% of the population and is characterized by recurrent eczematous lesions with intense itching. As a heterogeneous disease, multiple factors have been suggested to explain the nature of atopic dermatitis (AD), and its high prevalence makes it necessary to periodically compile and update the new information available. In this systematic review, the focus is set at the genetic and epigenetic studies carried out in the last years. METHODS A systematic literature review was conducted in three scientific publication databases (PubMed, Cochrane Library, and Scopus). The search was restricted to publications indexed from July 2016 to December 2019, and keywords related to atopic dermatitis genetics and epigenetics were used. RESULTS A total of 73 original papers met the inclusion criteria established, including 9 epigenetic studies. A total of 62 genes and 5 intergenic regions were described as associated with AD. CONCLUSION Filaggrin (FLG) polymorphisms are confirmed as key genetic determinants for AD development, but also epigenetic regulation and other genes with functions mainly related to the immune system and extracellular matrix, reinforcing the notion of skin homeostasis breakage in AD.
Collapse
Affiliation(s)
- Maria J Martin
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
| | - Miguel Estravís
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Asunción García-Sánchez
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Biomedical and Diagnostics Sciences, University of Salamanca, 37007 Salamanca, Spain
| | - Ignacio Dávila
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Immunoallergy, Salamanca University Hospital, 37007 Salamanca, Spain
| | - María Isidoro-García
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Clinical Biochemistry, University Hospital of Salamanca, 37007 Salamanca, Spain
- Department of Medicine, University of Salamanca, 37007 Salamanca, Spain
| | - Catalina Sanz
- Institute for Biomedical Research of Salamanca (IBSAL), 37007 Salamanca, Spain; (M.J.M.); (M.E.); (I.D.); (C.S.)
- Network for Cooperative Research in Health–RETICS ARADyAL, 37007 Salamanca, Spain
- Department of Microbiology and Genetics, University of Salamanca, 37007 Salamanca, Spain
| |
Collapse
|
39
|
Abstract
The hereditary nature of some forms of cancer was recognized long ago. Over time, recognition of associated findings led to the delineation of numerous hereditary cancer syndromes. Many of these syndromes also have cutaneous manifestations, the recognition of which can lead to their early identification. Recognition of these syndromes allows vigilant surveillance and preemptive treatment, which can dramatically impact the risks of morbidity and mortality for affected patients. The rise of rapid and accurate genetic testing now allows the early identification of asymptomatic at risk family members so that monitoring can be initiated as early as possible. The dermatologist plays a critical role in early identification of these syndromes and, in many cases, their treatment. This review summarizes many known hereditary cancer syndromes with cutaneous findings, their etiology, identification, evaluation, and management. Importantly, this is an ever evolving topic and new findings and syndromes will continue to be recognized. The dermatologist must be always alert to ensure they are detected.
Collapse
Affiliation(s)
- Ryan Ladd
- Department of Dermatology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Matthew Davis
- Department of Dermatology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Jonathan A Dyer
- Department of Dermatology, University of Missouri School of Medicine, Columbia, Missouri, USA.
| |
Collapse
|
40
|
Tangye SG. Genetic susceptibility to EBV infection: insights from inborn errors of immunity. Hum Genet 2020; 139:885-901. [PMID: 32152698 DOI: 10.1007/s00439-020-02145-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 02/27/2020] [Indexed: 02/07/2023]
Abstract
Epstein-Barr virus (EBV) is a ubiquitous human pathogen, infecting > 90% of the adult population. In the vast majority of healthy individuals, infection with EBV runs a relatively benign course. However, EBV is by no means a benign pathogen. Indeed, apart from being associated with at least seven different types of malignancies, EBV infection can cause severe and often fatal diseases-hemophagocytic lymphohistiocytosis, lymphoproliferative disease, B-cell lymphoma-in rare individuals with specific monogenic inborn errors of immunity. The discovery and detailed investigation of inborn errors of immunity characterized by heightened susceptibility to, or increased frequency of, EBV-induced disease have elegantly revealed cell types and signaling pathways that play critical and non-redundant roles in host-defense against EBV. These analyses have revealed not only mechanisms underlying EBV-induced disease in rare genetic conditions, but also identified molecules and pathways that could be targeted to treat severe EBV infection and pathological consequences in immunodeficient hosts, or even potentially enhance the efficacy of an EBV-specific vaccine.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Sydney, NSW, 2010, Australia. .,St. Vincent's Clinical School, University of NSW Sydney, Darlinghurst, NSW, 2010, Australia. .,Clincial Immunogenomics Research Consortium Australasia (CIRCA), Darlinghurst, NSW, Australia.
| |
Collapse
|
41
|
Villanueva JCMM, Chan KW, Ong RC, Andaya AG, Lau YL, van Zelm MC, Kanegane H. Hyper IgE Syndrome Associated With Warts: A First Case of Dedicator of Cytokinesis 8 Deficiency in the Philippines. Front Pediatr 2020; 8:604725. [PMID: 33251169 PMCID: PMC7673426 DOI: 10.3389/fped.2020.604725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 10/09/2020] [Indexed: 11/13/2022] Open
Abstract
Hyper IgE syndrome (HIES) encompasses a group of primary immunodeficiency diseases (PIDs) that is characterized by severe atopy, and recurrent infections and markedly elevated serum IgE levels. The majority of HIES cases suffer from autosomal dominant mutations in the signal transducer and activator of transcription 3 gene. A minority of cases display autosomal recessive inheritance, and one form is caused by mutations in the dedicator of cytokinesis 8 (DOCK8) gene. Here we describe the first recognized and diagnosed case of DOCK8 deficiency in the Philippines. A 14 year-old-girl was referred due to recalcitrant atopic dermatitis, recurrent sinopulmonary infections, with widespread warts on the face, trunk and extremities. She had no coarse facial features or retained primary teeth, whereas she presented with widespread viral skin infections and multiple allergic diseases. Laboratory examinations revealed elevations in eosinophil count and serum IgE. The level of T-cell receptor excision circles was undetectable. The patient was suspected to have HIES with a probable DOCK8 deficiency. Genetic analysis disclosed a large genomic deletion involving exons 2-4 in the DOCK8 gene. A combination of recalcitrant atopic dermatitis, asthma, food allergies, with viral skin infections should increase the physician's consideration of a PID. Patients with HIES accompanied by warts and T-cell deficiency can be strongly suspected to have DOCK8 deficiency.
Collapse
Affiliation(s)
- Jose Carlo Miguel M Villanueva
- Section of Allergy and Clinical Immunology, Department of Pediatrics, University of Santo Tomas Hospital, Manila, Philippines
| | - Koon-Wing Chan
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Remedios C Ong
- Section of Allergy and Clinical Immunology, Department of Pediatrics, University of Santo Tomas Hospital, Manila, Philippines
| | - Agnes G Andaya
- Section of Allergy and Clinical Immunology, Department of Pediatrics, University of Santo Tomas Hospital, Manila, Philippines
| | - Yu-Lung Lau
- Department of Paediatrics and Adolescent Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China
| | - Menno C van Zelm
- Department of Immunology and Pathology, Monash University and Alfred Hospital, Melbourne, VIC, Australia
| | - Hirokazu Kanegane
- Department of Child Health and Development, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
42
|
Lenardo MJ, Holland SM. Introduction: Continuing insights into the healthy and diseased immune system through human genetic investigation. Immunol Rev 2019; 287:5-8. [PMID: 30565248 DOI: 10.1111/imr.12730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Accepted: 11/23/2018] [Indexed: 12/19/2022]
Affiliation(s)
- Michael J Lenardo
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Steven M Holland
- Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
43
|
Kim D, Uner A, Saglam A, Chadburn A, Crane GM. Peripheral eosinophilia in primary immunodeficiencies of actin dysregulation: A case series of Wiskott-Aldrich syndrome, CARMIL2 and DOCK8 deficiency and review of the literature. Ann Diagn Pathol 2019; 43:151413. [DOI: 10.1016/j.anndiagpath.2019.151413] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 09/11/2019] [Indexed: 11/26/2022]
|
44
|
Kuloglu Z, Balcı D, Haskoloğlu ZŞ, Kendirli T, Bingöl-Koloğlu M, Tuna-Kırsaçlıoğlu C, Bal S, Selbuz S, Kırımker O, Savaş B, Altuntaş C, Güner ŞN, Can ÖS, Karayalçın K, Doğu F, Kansu Tanca A, İkincioğulları A. Allogeneic hematopoietic stem cell and liver transplantation in a young girl with dedicator of cytokinesis 8 protein deficiency. Pediatr Transplant 2019; 23:e13545. [PMID: 31297914 DOI: 10.1111/petr.13545] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2019] [Revised: 05/14/2019] [Accepted: 06/08/2019] [Indexed: 11/28/2022]
Abstract
DOCK8 deficiency is a rare inherited combined immunodeficiency, caused by mutations in the DOCK8 gene. We describe a case with DOCK8 deficiency associated with severe CLD in whom orthotopic LT was performed successfully after allogeneic HSCT. A 5 year-old girl with DOCK8 deficiency presented with mild direct hyperbilirubinemia and abnormal GGT level and without a previous history of jaundice. She had severe growth retardation, hepatosplenomegaly and generalized eczema. Progressive worsening of CLD was observed within 4 months. Investigations for etiology of liver disease were negative. Liver biopsy showed bridging necrosis, cholestasis and, cirrhosis. Recurrent immune hemolytic crisis and several viral infections developed in follow-up. She underwent whole cadaveric LT for end-stage liver disease (ESLD) 1 year after allogenic HSCT from a full matched related donor. The postoperative course was uneventful. The patient is alive with normal liver function and moderate skin graft versus host disease for 36 months after LT. In conclusion DOCK8 deficiency can be associated with severe CLD. Successful LT following HSCT is possible in patients with ESLD in DOCK8 deficiency. The timing of LT is challenging in patients requiring both HSCT and LT since conditioning regimens for HSCT can be highly hepatotoxic and the patients with suboptimal liver function can become decompensated during HSCT.
Collapse
Affiliation(s)
- Zarife Kuloglu
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Ankara University School of Medicine, Ankara, Turkey
| | - Deniz Balcı
- Deparment of General Surgery, Ankara University School of Medicine, Ankara, Turkey
| | - Zehra Şule Haskoloğlu
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Tanıl Kendirli
- Division of Pediatric Intensive Care, Ankara University School of Medicine, Ankara, Turkey
| | - Meltem Bingöl-Koloğlu
- Deparment of Pediatric Surgery, Ankara University School of Medicine, Ankara, Turkey
| | - Ceyda Tuna-Kırsaçlıoğlu
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Ankara University School of Medicine, Ankara, Turkey
| | - Sevgi Bal
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Suna Selbuz
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Ankara University School of Medicine, Ankara, Turkey
| | - Onur Kırımker
- Deparment of General Surgery, Ankara University School of Medicine, Ankara, Turkey
| | - Berna Savaş
- Deparment of Pathology, Ankara University School of Medicine, Ankara, Turkey
| | - Cansu Altuntaş
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Ankara University School of Medicine, Ankara, Turkey
| | - Şükrü Nail Güner
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Özlem Selvi Can
- Department of Anesthesiology and Reanimation, Ankara University School of Medicine, Ankara, Turkey
| | - Kaan Karayalçın
- Deparment of General Surgery, Ankara University School of Medicine, Ankara, Turkey
| | - Figen Doğu
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| | - Aydan Kansu Tanca
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, Ankara University School of Medicine, Ankara, Turkey
| | - Aydan İkincioğulları
- Division of Pediatric Immunology and Allergy, Ankara University School of Medicine, Ankara, Turkey
| |
Collapse
|
45
|
Ma CS, Tangye SG. Flow Cytometric-Based Analysis of Defects in Lymphocyte Differentiation and Function Due to Inborn Errors of Immunity. Front Immunol 2019; 10:2108. [PMID: 31552044 PMCID: PMC6737833 DOI: 10.3389/fimmu.2019.02108] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 08/21/2019] [Indexed: 12/13/2022] Open
Abstract
The advent of flow cytometry has revolutionized the way we approach our research and answer specific scientific questions. The flow cytometer has also become a mainstream diagnostic tool in most hospital and pathology laboratories around the world. In particular the application of flow cytometry has been instrumental to the diagnosis of primary immunodeficiencies (PIDs) that result from monogenic mutations in key genes of the hematopoietic, and occasionally non-hematopoietic, systems. The far-reaching applicability of flow cytometry is in part due to the remarkable sensitivity, down to the single-cell level, of flow-based assays and the extremely user-friendly platforms that enable comprehensive analysis, data interpretation, and importantly, robust and rapid methods for diagnosing PIDs. A prime example is the absence of peripheral blood B cells in patients with agammaglobulinemia due to mutations in BTK or related genes in the BCR signaling pathway. Similarly, the development of intracellular staining protocols to detect expression of SAP, XIAP, or DOCK8 expedites the rapid diagnosis of the X-linked lymphoproliferative diseases or an autosomal recessive form of hyper-IgE syndrome (HIES), respectively. It has also become evident that distinct cohorts of PID patients exhibit unique “lymphocyte phenotypic signatures” that are often diagnostic even prior to identifying the genetic lesion. Flow cytometry-based sorting provides a technique for separating specific subsets of immune cells such that they can be studied in isolation. Thus, flow-based assays can be utilized to measure immune cell function in patients with PIDs, such as degranulation by cytotoxic cells, cytokine expression by many immune cells (i.e., CD4+ and CD8+ T cells, macrophages etc.), B-cell differentiation, and phagocyte respiratory burst in vitro. These assays can also be performed using unfractionated PBMCs, provided the caveat that the composition of lymphocytes between healthy donors and the PID patients under investigation is recognized. These functional deficits can assist not only in the clinical diagnosis of PIDs, but also reveal mechanisms of disease pathogenesis. As we move into the next generation of multiparameter flow cytometers, here we review some of our experiences in the use of flow cytometry in the study, diagnosis, and unraveling the pathophysiology of PIDs.
Collapse
Affiliation(s)
- Cindy S Ma
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.,Clincial Immunogenomics Research Consortium Australia, Darlinghurst, NSW, Australia
| | - Stuart G Tangye
- Immunology Division, Garvan Institute of Medical Research, Sydney, NSW, Australia.,Faculty of Medicine, St. Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.,Clincial Immunogenomics Research Consortium Australia, Darlinghurst, NSW, Australia
| |
Collapse
|
46
|
Gowthaman U, Chen JS, Zhang B, Flynn WF, Lu Y, Song W, Joseph J, Gertie JA, Xu L, Collet MA, Grassmann JDS, Simoneau T, Chiang D, Berin MC, Craft JE, Weinstein JS, Williams A, Eisenbarth SC. Identification of a T follicular helper cell subset that drives anaphylactic IgE. Science 2019; 365:science.aaw6433. [PMID: 31371561 PMCID: PMC6901029 DOI: 10.1126/science.aaw6433] [Citation(s) in RCA: 325] [Impact Index Per Article: 54.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 07/18/2019] [Indexed: 12/15/2022]
Abstract
Cross-linking of high-affinity immunoglobulin E (IgE) results in the life-threatening allergic reaction anaphylaxis. Yet the cellular mechanisms that induce B cells to produce IgE in response to allergens remain poorly understood. T follicular helper (TFH) cells direct the affinity and isotype of antibodies produced by B cells. Although TFH cell-derived interleukin-4 (IL-4) is necessary for IgE production, it is not sufficient. We report a rare population of IL-13-producing TFH cells present in mice and humans with IgE to allergens, but not when allergen-specific IgE was absent or only low-affinity. These "TFH13" cells have an unusual cytokine profile (IL-13hiIL-4hiIL-5hiIL-21lo) and coexpress the transcription factors BCL6 and GATA3. TFH13 cells are required for production of high- but not low-affinity IgE and subsequent allergen-induced anaphylaxis. Blocking TFH13 cells may represent an alternative therapeutic target to ameliorate anaphylaxis.
Collapse
Affiliation(s)
- Uthaman Gowthaman
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jennifer S Chen
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Biyan Zhang
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - William F Flynn
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Yisi Lu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Wenzhi Song
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Lan Xu
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA.,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Magalie A Collet
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | | | - Tregony Simoneau
- The Asthma Center, CT Children's Medical Center, Hartford, CT 06106, USA
| | - David Chiang
- Jaffe Food Allergy Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - M Cecilia Berin
- Jaffe Food Allergy Institute and Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Joseph E Craft
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Jason S Weinstein
- Center for Immunity and Inflammation, Rutgers New Jersey Medical School, Newark, NJ 07101, USA
| | - Adam Williams
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA. .,The Department of Genetics and Genome Sciences, University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, Yale University School of Medicine, New Haven, CT 06520, USA. .,Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| |
Collapse
|
47
|
Sacco KA, Milner JD. Gene-environment interactions in primary atopic disorders. Curr Opin Immunol 2019; 60:148-155. [PMID: 31302571 DOI: 10.1016/j.coi.2019.06.002] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Accepted: 06/14/2019] [Indexed: 12/18/2022]
Abstract
Environmental factors modify disease presentation and severity in allergic disorders. Primary atopic disorders (PADs) are a heterogenous group of single gene disorders that lead to significant atopic and allergic disease manifestations. However, a number of these monogenic diseases have variable penetrance suggesting that gene-gene and/or gene-environment interactions could modulate the clinical phenotype. Environmental factors such as diet, the microbiome at the epithelial-environment interface, the presence and/or extent of infection, and psychologic stress can alter disease phenotypic expression of allergic diseases, and PADs provide discrete contexts in which to understand these influences. We outline how gene-environment interactions likely contribute to a variable penetrance and expressivity in PADs. Dietary modifications of both macronutrients and/or micronutrients alter T-cell metabolism and may influence effector T-cell function. The mucosal microbiome may affect local inflammation and may remotely influence regulatory elements, while psychologic stress can affect mast cell and other allergic effector cell function. Understanding gene-environment interactions in PADs can hopefully provide a foundation for interrogating gene-environment interactions to common allergic disorders, and also present opportunities for personalized interventions based on the altered pathways and environmental influences in affected individuals.
Collapse
Affiliation(s)
- Keith A Sacco
- Laboratory of Allergic Diseases, NIAID, NIH, 9000 Rockville Pike, NIH Building 10 Room 11N240A, United States
| | - Joshua D Milner
- Laboratory of Allergic Diseases, NIAID, NIH, 9000 Rockville Pike, NIH Building 10 Room 11N240A, United States.
| |
Collapse
|
48
|
Compound Heterozygous DOCK8 Mutations in a Patient with B Lymphoblastic Leukemia and EBV-Associated Diffuse Large B Cell Lymphoma. J Clin Immunol 2019; 39:592-595. [DOI: 10.1007/s10875-019-00663-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2019] [Accepted: 06/20/2019] [Indexed: 10/26/2022]
|
49
|
Volpi S, Cicalese MP, Tuijnenburg P, Tool ATJ, Cuadrado E, Abu-Halaweh M, Ahanchian H, Alzyoud R, Akdemir ZC, Barzaghi F, Blank A, Boisson B, Bottino C, Brigida I, Caorsi R, Casanova JL, Chiesa S, Chinn IK, Dückers G, Enders A, Erichsen HC, Forbes LR, Gambin T, Gattorno M, Karimiani EG, Giliani S, Gold MS, Jacobsen EM, Jansen MH, King JR, Laxer RM, Lupski JR, Mace E, Marcenaro S, Maroofian R, Meijer AB, Niehues T, Notarangelo LD, Orange J, Pannicke U, Pearson C, Picco P, Quinn PJ, Schulz A, Seeborg F, Stray-Pedersen A, Tawamie H, van Leeuwen EMM, Aiuti A, Yeung R, Schwarz K, Kuijpers TW. A combined immunodeficiency with severe infections, inflammation, and allergy caused by ARPC1B deficiency. J Allergy Clin Immunol 2019; 143:2296-2299. [PMID: 30771411 PMCID: PMC6677392 DOI: 10.1016/j.jaci.2019.02.003] [Citation(s) in RCA: 77] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 01/31/2019] [Accepted: 02/05/2019] [Indexed: 10/27/2022]
Affiliation(s)
- Stefano Volpi
- Clinica Pediatrica e Reumatologia, Centro per le malattie Autoinfiammatorie e Immunodeficienze, Istituto Giannina Gaslini, Genova, Italy; DINOGMI, Università degli Studi di Genova, Genova, Italy.
| | - Maria Pia Cicalese
- Pediatric Immunohematology, San Raffaele Hospital and San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Paul Tuijnenburg
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Anton T J Tool
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Eloy Cuadrado
- Department of Immunopathology, Sanquin Research and Landsteiner Laboratory AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Marwan Abu-Halaweh
- Department of Biotechnology and Genetics Engineering in Philadelphia University, Amman, Jordan
| | - Hamid Ahanchian
- Department of Allergy and Immunology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Raed Alzyoud
- Queen Rania Children's Hospital, Immunology, Allergy and Rheumatology Section, Bone Marrow Transplantation for Primary Immunodeficiency Disorders, Amman, Jordan
| | - Zeynep Coban Akdemir
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas
| | - Federica Barzaghi
- Pediatric Immunohematology, San Raffaele Hospital and San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Alexander Blank
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Bertrand Boisson
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris, France; Imagine Institute, Paris Descartes University, Paris, France
| | - Cristina Bottino
- Department of Experimental Medicine (DIMES), University of Genoa, Genova, Italy; Istituto Giannina Gaslini, Genova, Italy
| | - Immacolata Brigida
- Pediatric Immunohematology, San Raffaele Hospital and San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Roberta Caorsi
- Clinica Pediatrica e Reumatologia, Centro per le malattie Autoinfiammatorie e Immunodeficienze, Istituto Giannina Gaslini, Genova, Italy
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY; Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Paris, France; Imagine Institute, Paris Descartes University, Paris, France; Pediatric Hematology-Immunology and Rheumatology Unit, Necker Hospital for Sick Children, APHP, Paris, France; Howard Hughes Medical Institute, New York, NY
| | - Sabrina Chiesa
- Clinica Pediatrica e Reumatologia, Centro per le malattie Autoinfiammatorie e Immunodeficienze, Istituto Giannina Gaslini, Genova, Italy
| | - Ivan Kingyue Chinn
- Department of Pediatrics, Section of Allergy, Immunology, and Rheumatology & Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas
| | - Gregor Dückers
- Center for Child and Adolescent Medicine, Helios-Clinic, Krefeld, Germany
| | - Anselm Enders
- Department of Immunology and Infectious Disease, John Curtin School of Medical Research and Centre for Personalised Immunology, Australian National University, Canberra, ACT, Australia
| | - Hans Christian Erichsen
- Section of Paediatric Medicine and Transplantation, Division of Paediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Lisa R Forbes
- Department of Pediatrics, Section of Allergy, Immunology, and Rheumatology & Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas
| | - Tomasz Gambin
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas; Institute of Computer Science, Warsaw University of Technology, Warsaw, Poland
| | - Marco Gattorno
- Clinica Pediatrica e Reumatologia, Centro per le malattie Autoinfiammatorie e Immunodeficienze, Istituto Giannina Gaslini, Genova, Italy
| | - Ehsan Ghayoor Karimiani
- Molecular and Clinical Sciences Institute, St George's, University of London, Cranmer Terrace, London, United Kingdom; Innovative Medical Research Center, Mashhad Branch, Islamic Azad University, Mashhad, Iran
| | - Silvia Giliani
- Medical Genetics Unit and "A. Nocivelli" Institute for Molecular Medicine, Spedali Civili Hospital, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Michael S Gold
- Discipline of Pediatrics, School of Medicine, University of Adelaide and Department of Allergy and Clinical Immunology, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | | | - Machiel H Jansen
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Jovanka R King
- Discipline of Pediatrics, School of Medicine, University of Adelaide and Department of Allergy and Clinical Immunology, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Ronald M Laxer
- Division of Rheumatology, Department of Paediatrics and Department of Medicine, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - James R Lupski
- Baylor-Hopkins Center for Mendelian Genomics of the Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, Texas; Department of Pediatrics, Baylor College of Medicine, Houston, Texas; Texas Children's Hospital, Houston, Texas
| | - Emily Mace
- Department of Pediatrics, Section of Allergy, Immunology, and Rheumatology & Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas
| | | | - Reza Maroofian
- Medical Research, RILD Welcome Wolfson Centre, Exeter Medical School, Royal Devon and Exeter NHS Foundation Trust, Exeter and Genetics and Molecular Cell Sciences Research Centre, St George's University of London, London, United Kingdom
| | - Alexander B Meijer
- Department of Plasma Proteins, Sanquin Research and Landsteiner Laboratory AMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Tim Niehues
- Center for Child and Adolescent Medicine, Helios-Clinic, Krefeld, Germany
| | - Luigi D Notarangelo
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, NIH, Bethesda, Md
| | - Jordan Orange
- Department of Pediatrics, Section of Allergy, Immunology, and Rheumatology & Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas
| | - Ulrich Pannicke
- Institute for Transfusion Medicine, University Ulm, Ulm, Germany
| | - Chris Pearson
- Department of General Medicine, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Paolo Picco
- Clinica Pediatrica e Reumatologia, Istituto Giannina Gaslini, Genova, Italy
| | - Patrick J Quinn
- Discipline of Pediatrics, School of Medicine, University of Adelaide and Department of Allergy and Clinical Immunology, Women's and Children's Health Network, Adelaide, South Australia, Australia
| | - Ansgar Schulz
- Department of Pediatrics, University Medical Center Ulm, Ulm, Germany
| | - Filiz Seeborg
- Department of Pediatrics, Section of Allergy, Immunology, and Rheumatology & Center for Human Immunobiology, Texas Children's Hospital, Houston, Texas
| | - Asbjørg Stray-Pedersen
- Norwegian National Unit for Newborn Screening, Division of Pediatric and Adolescent Medicine, Oslo University Hospital, Oslo, Norway
| | - Hasan Tawamie
- Institute of Human Genetics of Leipzig, Leipzig, Germany
| | - Ester M M van Leeuwen
- Department of Experimental Immunology, Amsterdam Infection & Immunity Institute, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
| | - Alessandro Aiuti
- Pediatric Immunohematology, San Raffaele Hospital and San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Milan, Italy
| | - Rae Yeung
- Division of Rheumatology, Department of Paediatrics and Department of Medicine, University of Toronto, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Paediatrics, Institute of Medical Science, University of Toronto, Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Immunology, Institute of Medical Science, University of Toronto, Cell Biology Program, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Klaus Schwarz
- Institute for Transfusion Medicine, University Ulm, Ulm, Germany; the Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Wuerttemberg - Hessen, Ulm, Germany
| | - Taco W Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children's Hospital, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands; Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory AMC, University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
50
|
A combined immunodeficiency with severe infections, inflammation, and allergy caused by ARPC1B deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY 2019. [PMID: 30771411 DOI: 10.1016/j.jaci.2019.02.003)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
|