1
|
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
|
2
|
Attias M, Alvarez F, Al-Aubodah TA, Istomine R, McCallum P, Huang F, Sleiman A, Nishimura T, Del Rincón SV, Riazalhosseini Y, Piccirillo CA. Anti-PD-1 amplifies costimulation in melanoma-infiltrating T h1-like Foxp3 + regulatory T cells to alleviate local immunosuppression. J Immunother Cancer 2025; 13:e009435. [PMID: 39762077 PMCID: PMC11748786 DOI: 10.1136/jitc-2024-009435] [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: 04/12/2024] [Accepted: 11/14/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors targeting programmed cell death protein-1 (PD-1) are the first line of treatment for many solid tumors including melanoma. PD-1 blockade enhances the effector functions of melanoma-infiltrating CD8+ T cells, leading to durable tumor remissions. However, 55% of patients with melanoma do not respond to treatment. As Foxp3+ regulatory T (Treg) cells play an important role in tumor-induced immunosuppression and express PD-1, we hypothesized that anti-PD-1 also increases the functions of melanoma-infiltrating Treg cells, which could be detrimental to treatment efficacy. METHODS The cellular and functional dynamics of Treg cells were evaluated in C57Bl/6 Foxp3-reporter mice bearing highly immunogenic and PD-1 blockade-sensitive Yale University Mouse Melanoma Exposed to Radiation 1.7 (YUMMER1.7) tumors. Treg cell responses in tumors and lymphoid compartments were examined throughout tumor growth or therapy and were assessed ex vivo by multiparametric flow cytometry analysis, with in vitro suppression assays using tumor-infiltrating lymphocytes isolated by fluorescence-activated cell sorting (FACS) and in situ through spatial proteomic and transcriptomic profiling. RESULTS In this highly immunogenic melanoma model, anti-PD-1 monotherapy yielded high responders (HRs) and low responders (LRs). We show that the potent CD8+ T cell responses characteristic of HR tumors paradoxically coincide with the expansion of highly-activated, Helios-expressing Treg cells. In both HRs and LRs, Treg cells co-localize with CD8+ T cells in immunogenic regions of the tumor and display potent suppressive capacity in vitro. Further characterization revealed that melanoma-infiltrating Treg cells progressively acquire T-bet and interferon gamma expression, exclusively in HRs, and induction of this T helper cell 1 (Th1)-like phenotype in vitro led to CD8+ T cell evasion from Treg cell-mediated suppression. Using spatial proteomic and transcriptomic profiling, we demonstrate that Treg cells display an increased activity of PI3K/Akt signaling in regions of HR tumors with an elevated CD8:Treg cell ratio. CONCLUSIONS PD-1 blockade promotes the expansion of a subset of highly-activated Treg cells coexpressing PD-1 and Helios. While these cells are potently suppressive outside tumor environments, costimulatory and inflammatory signals present in the tumor microenvironment lead to their local acquisition of Th1-like characteristics and loss of suppression of effector T cells.
Collapse
Affiliation(s)
- Mikhaël Attias
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Centre for Translation Biology (CTB), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montreal, Quebec, Canada
| | - Fernando Alvarez
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Centre for Translation Biology (CTB), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montreal, Quebec, Canada
| | - Tho-Alfakar Al-Aubodah
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Centre for Translation Biology (CTB), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montreal, Quebec, Canada
| | - Roman Istomine
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Centre for Translation Biology (CTB), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montreal, Quebec, Canada
| | - Paige McCallum
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Fan Huang
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Abrahim Sleiman
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Centre for Translation Biology (CTB), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montreal, Quebec, Canada
| | - Tamiko Nishimura
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
| | - Sonia V Del Rincón
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Segal Cancer Centre, Lady Davis Institute and Jewish General Hospital, McGill University, Montreal, Quebec, Canada
| | - Yasser Riazalhosseini
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
- Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | - Ciriaco A Piccirillo
- Department of Microbiology and Immunology, McGill University, Montreal, Quebec, Canada
- Infectious Diseases and Immunity in Global Health (IDIGH) Program, Centre for Translation Biology (CTB), The Research Institute of the McGill University Health Centre (RI-MUHC), Montreal, Quebec, Canada
- Centre of Excellence in Translational Immunology (CETI), McGill University, Montreal, Quebec, Canada
- Division of Experimental Medicine, McGill University, Montreal, Quebec, Canada
- Victor Philip Dahdaleh Institute of Genomic Medicine at McGill University, Montreal, Quebec, Canada
| |
Collapse
|
3
|
Azizoglu ZB, Babayeva R, Haskologlu ZS, Acar MB, Ayaz-Guner S, Okus FZ, Alsavaf MB, Can S, Basaran KE, Canatan MF, Ozcan A, Erkmen H, Leblebici CB, Yilmaz E, Karakukcu M, Kose M, Canoz O, Özen A, Karakoc-Aydiner E, Ceylaner S, Gümüş G, Per H, Gumus H, Canatan H, Ozcan S, Dogu F, Ikinciogullari A, Unal E, Baris S, Eken A. DIAPH1-Deficiency is Associated with Major T, NK and ILC Defects in Humans. J Clin Immunol 2024; 44:175. [PMID: 39120629 PMCID: PMC11315734 DOI: 10.1007/s10875-024-01777-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024]
Abstract
Loss of function mutations in Diaphanous related formin 1 (DIAPH1) are associated with seizures, cortical blindness, and microcephaly syndrome (SCBMS) and are recently linked to combined immunodeficiency. However, the extent of defects in T and innate lymphoid cells (ILCs) remain unexplored. Herein, we characterized the primary T, natural killer (NK) and helper ILCs of six patients carrying two novel loss of function mutation in DIAPH1 and Jurkat cells after DIAPH1 knockdown. Mutations were identified by whole exome sequencing. T-cell immunophenotyping, proliferation, migration, cytokine signaling, survival, and NK cell cytotoxicity were studied via flow cytometry-based assays, confocal microscopy, and real-time qPCR. CD4+ T cell proteome was analyzed by mass spectrometry. p.R351* and p.R322*variants led to a significant reduction in the DIAPH1 mRNA and protein levels. DIAPH1-deficient T cells showed proliferation, activation, as well as TCR-mediated signaling defects. DIAPH1-deficient PBMCs also displayed impaired transwell migration, defective STAT5 phosphorylation in response to IL-2, IL-7 and IL-15. In vitro generation/expansion of Treg cells from naïve T cells was significantly reduced. shRNA-mediated silencing of DIAPH1 in Jurkat cells reduced DIAPH1 protein level and inhibited T cell proliferation and IL-2/STAT5 axis. Additionally, NK cells from patients had diminished cytotoxic activity, function and IL-2/STAT5 axis. Lastly, DIAPH1-deficient patients' peripheral blood contained dramatically reduced numbers of all helper ILC subsets. DIAPH1 deficiency results in major functional defects in T, NK cells and helper ILCs underlining the critical role of formin DIAPH1 in the biology of those cell subsets.
Collapse
Affiliation(s)
- Zehra Busra Azizoglu
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
| | - Royala Babayeva
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Zehra Sule Haskologlu
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | | | - Serife Ayaz-Guner
- Department of Molecular Biology and Genetics, Izmir Institute of Technology, Izmir, Türkiye
| | - Fatma Zehra Okus
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | | | - Salim Can
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Kemal Erdem Basaran
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
- Department of Physiology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | | | - Alper Ozcan
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Hasret Erkmen
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Can Berk Leblebici
- Department of Medical Genetics, Ankara University Faculty of Medicine, Ankara, Türkiye
| | - Ebru Yilmaz
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Musa Karakukcu
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey
| | - Mehmet Kose
- Division of Pediatric Pulmonology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Ozlem Canoz
- Department of Pathology, Faculty of Medicine, Erciyes University, 38039, Kayseri, Türkiye
| | - Ahmet Özen
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Elif Karakoc-Aydiner
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye
| | - Serdar Ceylaner
- Intergen, Genetic, Rare and Undiagnosed Diseases, Diagnosis and Research Center, Ankara, Türkiye
| | - Gülsüm Gümüş
- Division of Pediatric Radiology, Department of Radiology, Erciyes University Faculty of Medicine, Kayseri, Türkiye
| | - Huseyin Per
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Hakan Gumus
- Division of Pediatric Neurology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
| | - Halit Canatan
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
| | - Servet Ozcan
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye
- Department of Biology, Faculty of Science, Erciyes University, Kayseri, 38039, Türkiye
| | - Figen Dogu
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Aydan Ikinciogullari
- Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Ankara University, Ankara, Türkiye
| | - Ekrem Unal
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Faculty of Medicine, Erciyes University, Kayseri, 38039, Turkey.
- School of Health Sciences, Hasan Kalyoncu University, Gaziantep, Türkiye.
- Medical Point Hospital, Pediatric Hematology Oncology and BMT Unit, Gaziantep, Türkiye.
| | - Safa Baris
- The Istanbul Jeffrey Modell Diagnostic Center for Primary Immunodeficiency Diseases, The Isil Berat Barlan Center for Translational Medicine, Division of Pediatric Allergy and Immunology, Department of Pediatrics, Faculty of Medicine, Marmara University, Istanbul, Türkiye.
| | - Ahmet Eken
- Department of Medical Biology, Faculty of Medicine, Erciyes University, Kayseri, 38039, Türkiye.
- Genome and Stem Cell Center, Kayseri, 38039, Türkiye.
| |
Collapse
|
4
|
Zhang B, Chen S, Yin X, McBride CD, Gertie JA, Yurieva M, Bielecka AA, Hoffmann B, Travis Hinson J, Grassmann J, Xu L, Siniscalco ER, Soldatenko A, Hoyt L, Joseph J, Norton EB, Uthaman G, Palm NW, Liu E, Eisenbarth SC, Williams A. Metabolic fitness of IgA + plasma cells in the gut requires DOCK8. Mucosal Immunol 2024; 17:431-449. [PMID: 38159726 PMCID: PMC11571232 DOI: 10.1016/j.mucimm.2023.12.001] [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: 07/10/2023] [Revised: 11/16/2023] [Accepted: 12/01/2023] [Indexed: 01/03/2024]
Abstract
Dedicator of cytokinesis 8 (DOCK8) mutations lead to a primary immunodeficiency associated with recurrent gastrointestinal infections and poor antibody responses but, paradoxically, heightened IgE to food antigens, suggesting that DOCK8 is central to immune homeostasis in the gut. Using Dock8-deficient mice, we found that DOCK8 was necessary for mucosal IgA production to multiple T cell-dependent antigens, including peanut and cholera toxin. Yet DOCK8 was not necessary in T cells for this phenotype. Instead, B cell-intrinsic DOCK8 was required for maintenance of antigen-specific IgA-secreting plasma cells (PCs) in the gut lamina propria. Unexpectedly, DOCK8 was not required for early B cell activation, migration, or IgA class switching. An unbiased interactome screen revealed novel protein partners involved in metabolism and apoptosis. Dock8-deficient IgA+ B cells had impaired cellular respiration and failed to engage glycolysis appropriately. These results demonstrate that maintenance of the IgA+ PC compartment requires DOCK8 and suggest that gut IgA+ PCs have unique metabolic requirements for long-term survival in the lamina propria.
Collapse
Affiliation(s)
- Biyan Zhang
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A*STAR), 8A Biomedical Grove, Immunos, Singapore 138648, Singapore
| | - Shuting Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Xiangyun Yin
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Caleb D McBride
- The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Jake A Gertie
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Agata A Bielecka
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Microbial Immunoregulation, Helmholtz Center for Infection Research, 38124 Braunschweig, Germany
| | - Brian Hoffmann
- Mass Spectrometry and Protein Chemistry, The Jackson Laboratory for Genomic Medicine, Bar Harbor, ME 04609, USA
| | - J Travis Hinson
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Cardiology center, Department of Medicine, UConn Health, Farmington, CT, USA
| | - Jessica Grassmann
- The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA
| | - Lan Xu
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Emily R Siniscalco
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Arielle Soldatenko
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Laura Hoyt
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Julie Joseph
- Department of Laboratory Medicine, USA; Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, PA 19102, USA
| | - Elizabeth B Norton
- Department of Microbiology & Immunology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Gowthaman Uthaman
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Department of Pathology, University of Massachusetts Medical School, Worcester, MA 01655, USA
| | - Noah W Palm
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Elise Liu
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; Section of Rheumatology, Allergy & Immunology, Yale University School of Medicine, New Haven, CT 06520, USA
| | - Stephanie C Eisenbarth
- Department of Laboratory Medicine, USA; Department of Immunobiology, Yale University School of Medicine, New Haven, CT 06520, USA; The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| | - Adam Williams
- The Department Medicine, Division of Allergy and Immunology, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, USA; The Jackson Laboratory for Genomic Medicine, Farmington, CT 06030, USA; Center for Human Immunobiology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
5
|
Kennedy-Batalla R, Acevedo D, Luo Y, Esteve-Solé A, Vlagea A, Correa-Rocha R, Seoane-Reula ME, Alsina L. Treg in inborn errors of immunity: gaps, knowns and future perspectives. Front Immunol 2024; 14:1278759. [PMID: 38259469 PMCID: PMC10800401 DOI: 10.3389/fimmu.2023.1278759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Regulatory T cells (Treg) are essential for immune balance, preventing overreactive responses and autoimmunity. Although traditionally characterized as CD4+CD25+CD127lowFoxP3hi, recent research has revealed diverse Treg subsets such as Tr1, Tr1-like, and CD8 Treg. Treg dysfunction leads to severe autoimmune diseases and immune-mediated inflammatory disorders. Inborn errors of immunity (IEI) are a group of disorders that affect correct functioning of the immune system. IEI include Tregopathies caused by genetic mutations affecting Treg development or function. In addition, Treg dysfunction is also observed in other IEIs, whose underlying mechanisms are largely unknown, thus requiring further research. This review provides a comprehensive overview and discussion of Treg in IEI focused on: A) advances and controversies in the evaluation of Treg extended subphenotypes and function; B) current knowledge and gaps in Treg disturbances in Tregopathies and other IEI including Treg subpopulation changes, genotype-phenotype correlation, Treg changes with disease activity, and available therapies, and C) the potential of Treg cell-based therapies for IEI with immune dysregulation. The aim is to improve both the diagnostic and the therapeutic approaches to IEI when there is involvement of Treg. We performed a non-systematic targeted literature review with a knowledgeable selection of current, high-quality original and review articles on Treg and IEI available since 2003 (with 58% of the articles within the last 6 years) in the PubMed database.
Collapse
Affiliation(s)
- Rebeca Kennedy-Batalla
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Daniel Acevedo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Yiyi Luo
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Ana Esteve-Solé
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Alexandru Vlagea
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Immunology Department, Biomedic Diagnostic Center (CDB), Hospital Clínic of Barcelona, Clinical Immunology Unit Hospital Sant Joan de Déu-Hospital Clínic de Barcelona, Barcelona, Spain
| | - Rafael Correa-Rocha
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
| | - Ma Elena Seoane-Reula
- Laboratory of Immune-Regulation, Gregorio Marañón Health Research Institute (IISGM), Madrid, Spain
- Pediatric Immuno-Allergy Unit, Allergy Department, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Primary Immunodeficiencies Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain
| | - Laia Alsina
- Clinical Immunology and Primary Immunodeficiencies Unit, Allergy and Clinical Immunology Department, Hospital Sant Joan de Déu, Barcelona, Spain
- Clinical Immunology Unit, Hospital Sant Joan de Déu-Hospital Clínic, Barcelona, Spain
- Study Group for Immune Dysfunction Diseases in Children (GEMDIP), Institut de Recerca Sant Joan de Déu, Barcelona, Spain
- Department of Surgery and Surgical Specializations, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
6
|
Mandato E, Yan Q, Ouyang J, Paczkowska J, Qin Y, Hao Y, Bojarczuk K, Hansen J, Chapuy B, Rodig SJ, Khan SJ, Redd RA, Shipp MA. MYD88L265P augments proximal B-cell receptor signaling in large B-cell lymphomas via an interaction with DOCK8. Blood 2023; 142:1219-1232. [PMID: 37467575 DOI: 10.1182/blood.2023019865] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/31/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023] Open
Abstract
Diffuse large B-cell lymphoma (DLBCL) is a clinically and genetically heterogeneous disease with at least 5 recognized molecular subtypes. Cluster 5 (C5)/MCD tumors frequently exhibit concurrent alterations in the toll-like receptor (TLR) and B-cell receptor (BCR) pathway members, MYD88L265P and CD79B, and have a less favorable prognosis. In healthy B cells, the synergy between TLR and BCR signaling pathways integrates innate and adaptive immune responses and augments downstream NF-κB activation. In addition, physiologic TLR9 pathway engagement via MYD88, protein tyrosine kinase 2 (PYK2), and dedicator of cytokinesis 8 (DOCK8) increases proximal BCR signaling in healthy murine B cells. Although C5/MCD DLBCLs are selectively sensitive to Bruton tyrosine kinase (BTK) inhibition in in vitro studies and certain clinical trials, the role of mutated MYD88 in proximal BCR signaling remains undefined. Using engineered DLBCL cell line models, we found that concurrent MYD88L265P and CD79B alterations significantly increased the magnitude and duration of proximal BCR signaling, at the level of spleen tyrosine kinase and BTK, and augmented PYK2-dependent DOCK8 phosphorylation. MYD88L265P DLBCLs have significantly increased colocalization of DOCK8 with both MYD88 and the proximal BCR-associated Src kinase, LYN, in comparison with MYD88WT DLBCLs, implicating DOCK8 in MYD88L265P/proximal BCR cross talk. Additionally, DOCK8 depletion selectively decreased proximal BCR signaling, cellular proliferation, and viability of DLBCLs with endogenous MYD88L265P/CD79BY196F alterations and increased the efficacy of BTK blockade in these lymphomas. Therefore, MYD88L265P/DOCK8-enhanced proximal BCR signaling is a likely mechanism for the increased sensitivity of C5/MCD DLBCLs to BTK blockade.
Collapse
Affiliation(s)
- Elisa Mandato
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Qingsheng Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jing Ouyang
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Bristol Myers Squibb, Cambridge, MA
| | - Julia Paczkowska
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Yan Qin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Parthenon Therapeutics, Boston, MA
| | - Yansheng Hao
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Pathology, University of Rochester Medical Center, Rochester, NY
| | - Kamil Bojarczuk
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Julia Hansen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Björn Chapuy
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
- Department of Hematology, Oncology, and Tumor Immunology, Charité - University Medical Center Berlin, Campus Benjamin Franklin, Berlin, Germany
| | - Scott J Rodig
- Department of Pathology, Brigham and Women's Hospital, Boston, MA
| | - Sumbul Jawed Khan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Robert A Redd
- Department of Data Science, Dana-Farber Cancer Institute, Boston, MA
| | - Margaret A Shipp
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
7
|
Ma CS. T-helper-2 cells and atopic disease: lessons learnt from inborn errors of immunity. Curr Opin Immunol 2023; 81:102298. [PMID: 36870225 DOI: 10.1016/j.coi.2023.102298] [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: 11/17/2022] [Revised: 01/30/2023] [Accepted: 02/02/2023] [Indexed: 03/06/2023]
Abstract
Inborn errors of immunity (IEI) are caused by monogenic variants that affect the host response to bacterial, viral, and fungal pathogens. As such, individuals with IEI often present with severe, recurrent, and life-threatening infections. However, the spectrum of disease due to IEI is very broad and extends to include autoimmunity, malignancy, and atopic diseases such as eczema, atopic dermatitis, and food and environmental allergies. Here, I review IEI that affect cytokine signaling pathways that dysregulate CD4+ T-cell differentiation, resulting in increased T-helper-2 (Th2) cell development, function, and pathogenicity. These are elegant examples of how rare IEI can provide unique insights into more common pathologies such as allergic disease that are impacting the general population at increased frequency.
Collapse
Affiliation(s)
- Cindy S Ma
- Garvan Institute of Medical Research, Sydney, NSW, Australia; School of Clinical Medicine, Faculty of Medicine and Health, UNSW Sydney, Sydney, NSW, Australia; Clinical Immunogenomics Research Consortium of Australasia (CIRCA), Australia.
| |
Collapse
|
8
|
Hewavisenti RV, Arena J, Ahlenstiel CL, Sasson SC. Human papillomavirus in the setting of immunodeficiency: Pathogenesis and the emergence of next-generation therapies to reduce the high associated cancer risk. Front Immunol 2023; 14:1112513. [PMID: 36960048 PMCID: PMC10027931 DOI: 10.3389/fimmu.2023.1112513] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 02/03/2023] [Indexed: 03/09/2023] Open
Abstract
Human papillomavirus (HPV), a common sexually transmitted virus infecting mucosal or cutaneous stratified epithelia, is implicated in the rising of associated cancers worldwide. While HPV infection can be cleared by an adequate immune response, immunocompromised individuals can develop persistent, treatment-refractory, and progressive disease. Primary immunodeficiencies (PIDs) associated with HPV-related disease include inborn errors of GATA, EVER1/2, and CXCR4 mutations, resulting in defective cellular function. People living with secondary immunodeficiency (e.g. solid-organ transplants recipients of immunosuppression) and acquired immunodeficiency (e.g. concurrent human immunodeficiency virus (HIV) infection) are also at significant risk of HPV-related disease. Immunocompromised people are highly susceptible to the development of cutaneous and mucosal warts, and cervical, anogenital and oropharyngeal carcinomas. The specific mechanisms underlying high-risk HPV-driven cancer development in immunocompromised hosts are not well understood. Current treatments for HPV-related cancers include surgery with adjuvant chemotherapy and/or radiotherapy, with clinical trials underway to investigate the use of anti-PD-1 therapy. In the setting of HIV co-infection, persistent high-grade anal intraepithelial neoplasia can occur despite suppressive antiretroviral therapy, resulting in an ongoing risk for transformation to overt malignancy. Although therapeutic vaccines against HPV are under development, the efficacy of these in the setting of PID, secondary- or acquired- immunodeficiencies remains unclear. RNA-based therapeutic targeting of the HPV genome or mRNA transcript has become a promising next-generation therapeutic avenue. In this review, we summarise the current understanding of HPV pathogenesis, immune evasion, and malignant transformation, with a focus on key PIDs, secondary immunodeficiencies, and HIV infection. Current management and vaccine regimes are outlined in relation to HPV-driven cancer, and specifically, the need for more effective therapeutic strategies for immunocompromised hosts. The recent advances in RNA-based gene targeting including CRISPR and short interfering RNA (siRNA), and the potential application to HPV infection are of great interest. An increased understanding of both the dysregulated immune responses in immunocompromised hosts and of viral persistence is essential for the design of next-generation therapies to eliminate HPV persistence and cancer development in the most at-risk populations.
Collapse
Affiliation(s)
- Rehana V. Hewavisenti
- Immunovirology and Pathogenesis Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Joshua Arena
- Immunovirology and Pathogenesis Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
- UNSW RNA Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Chantelle L. Ahlenstiel
- Immunovirology and Pathogenesis Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
- UNSW RNA Institute, The University of New South Wales, Sydney, NSW, Australia
| | - Sarah C. Sasson
- Immunovirology and Pathogenesis Program, The Kirby Institute, The University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
9
|
Su HC. Insights into the pathogenesis of allergic disease from dedicator of cytokinesis 8 deficiency. Curr Opin Immunol 2023; 80:102277. [PMID: 36508760 PMCID: PMC9972721 DOI: 10.1016/j.coi.2022.102277] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 11/01/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022]
Abstract
Clinical observations and mechanistic studies in dedicator of cytokinesis 8 (DOCK8)-deficient patients and mice have revealed multiple mechanisms that could contribute to their unusually prevalent and severe allergic disease manifestations. Physical interactions of DOCK8 with STAT3 in B cells and T cells may contribute to increased IgE isotype switching or defective immune synapse formation that decreases T-cell receptor signal strength. A newly discovered TFH13 cell type promotes the development of life-threatening allergy via production of IL-13 and is increased in DOCK8 deficiency. Cytoskeletal derangements and cytothripsis, which were previously shown to account for the increased susceptibility to viral skin infection in DOCK8 deficiency, can lead to interplay between myeloid cells and T cells to ultimately increase production of IL-4, IL-5, and IL-13. Finally, the effects on type-2 innate lymphoid cells may also contribute to allergic disease.
Collapse
Affiliation(s)
- Helen C Su
- Human Immunological Diseases Section, Laboratory of Clinical Immunology and Microbiology, Intramural Research Program, National Institute of Allergy and Infectious Diseases, National Institutes of Health, United States.
| |
Collapse
|
10
|
Pan C, Zhao A, Li M. Atopic Dermatitis-like Genodermatosis: Disease Diagnosis and Management. Diagnostics (Basel) 2022; 12:diagnostics12092177. [PMID: 36140582 PMCID: PMC9498295 DOI: 10.3390/diagnostics12092177] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 07/23/2022] [Accepted: 08/15/2022] [Indexed: 11/29/2022] Open
Abstract
Eczema is a classical characteristic not only in atopic dermatitis but also in various genodermatosis. Patients suffering from primary immunodeficiency diseases such as hyper-immunoglobulin E syndromes, Wiskott-Aldrich syndrome, immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome, STAT5B deficiency, Omenn syndrome, atypical complete DiGeorge syndrome; metabolic disorders such as acrodermatitis enteropathy, multiple carboxylase deficiency, prolidase deficiency; and other rare syndromes like severe dermatitis, multiple allergies and metabolic wasting syndrome, Netherton syndrome, and peeling skin syndrome frequently perform with eczema-like lesions. These genodermatosis may be misguided in the context of eczematous phenotype. Misdiagnosis of severe disorders unavoidably affects appropriate treatment and leads to irreversible outcomes for patients, which underlines the importance of molecular diagnosis and genetic analysis. Here we conclude clinical manifestations, molecular mechanism, diagnosis and management of several eczema-related genodermatosis and provide accessible advice to physicians.
Collapse
Affiliation(s)
- Chaolan Pan
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Anqi Zhao
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
| | - Ming Li
- Department of Dermatology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Institute of Dermatology, Shanghai Jiaotong University School of Medicine, Shanghai 200092, China
- Department of Dermatology, The Children’s Hospital of Fudan University, Shanghai 200092, China
- Correspondence: ; Tel.: +86-2125078571
| |
Collapse
|
11
|
Thangavelu G, Andrejeva G, Bolivar-Wagers S, Jin S, Zaiken MC, Loschi M, Aguilar EG, Furlan SN, Brown CC, Lee YC, Hyman CM, Feser CJ, Panoskaltsis-Mortari A, Hippen KL, MacDonald KP, Murphy WJ, Maillard I, Hill GR, Munn DH, Zeiser R, Kean LS, Rathmell JC, Chi H, Noelle RJ, Blazar BR. Retinoic acid signaling acts as a rheostat to balance Treg function. Cell Mol Immunol 2022; 19:820-833. [PMID: 35581350 PMCID: PMC9243059 DOI: 10.1038/s41423-022-00869-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 04/14/2022] [Indexed: 02/03/2023] Open
Abstract
Regulatory T cells (Tregs) promote immune homeostasis by maintaining self-tolerance and regulating inflammatory responses. Under certain inflammatory conditions, Tregs can lose their lineage stability and function. Previous studies have reported that ex vivo exposure to retinoic acid (RA) enhances Treg function and stability. However, it is unknown how RA receptor signaling in Tregs influences these processes in vivo. Herein, we employed mouse models in which RA signaling is silenced by the expression of the dominant negative receptor (DN) RARα in all T cells. Despite the fact that DNRARα conventional T cells are hypofunctional, Tregs had increased CD25 expression, STAT5 pathway activation, mTORC1 signaling and supersuppressor function. Furthermore, DNRARα Tregs had increased inhibitory molecule expression, amino acid transporter expression, and metabolic fitness and decreased antiapoptotic proteins. Supersuppressor function was observed when wild-type mice were treated with a pharmacologic pan-RAR antagonist. Unexpectedly, Treg-specific expression of DNRARα resulted in distinct phenotypes, such that a single allele of DNRARα in Tregs heightened their suppressive function, and biallelic expression led to loss of suppression and autoimmunity. The loss of Treg function was not cell intrinsic, as Tregs that developed in a noninflammatory milieu in chimeric mice reconstituted with DNRARα and wild-type bone marrow maintained the enhanced suppressive capacity. Fate mapping suggested that maintaining Treg stability in an inflammatory milieu requires RA signaling. Our findings indicate that RA signaling acts as a rheostat to balance Treg function in inflammatory and noninflammatory conditions in a dose-dependent manner.
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA.
| | - Gabriela Andrejeva
- Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sara Bolivar-Wagers
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Sujeong Jin
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Michael C Zaiken
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Michael Loschi
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Ethan G Aguilar
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Scott N Furlan
- Department of Pediatrics, University of Washington, Seattle, WA, USA
- Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Chrysothemis C Brown
- Howard Hughes Medical Institute, Immunology Program, and Ludwig Center, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yu-Chi Lee
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Lebanon, USA
| | - Cameron McDonald Hyman
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Colby J Feser
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | | | - Keli L Hippen
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| | - Kelli P MacDonald
- Department of Immunology, Queensland Institute of Medical Research (QIMR) Berghofer Medical Research Institute and School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - William J Murphy
- Department of Dermatology, School of Medicine, University of California, Davis, Sacramento, CA, USA
| | - Ivan Maillard
- Division of Hematology/Oncology, Department of Medicine, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - David H Munn
- Georgia Cancer Center, Augusta University, Augusta, GA, USA
| | - Robert Zeiser
- Department of Haematology, Oncology and Stem Cell Transplantation, Faculty of Medicine, Freiburg University Medical Centre, Freiburg, Germany
| | - Leslie S Kean
- Boston Children's Hospital and the Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jeffrey C Rathmell
- Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Randolph J Noelle
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Norris Cotton Cancer Center, Lebanon, USA
| | - Bruce R Blazar
- Department of Pediatrics, Center for Immunology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
12
|
Vaseghi-Shanjani M, Snow AL, Margolis DJ, Latrous M, Milner JD, Turvey SE, Biggs CM. Atopy as Immune Dysregulation: Offender Genes and Targets. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1737-1756. [PMID: 35680527 DOI: 10.1016/j.jaip.2022.04.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 04/06/2022] [Accepted: 04/06/2022] [Indexed: 06/15/2023]
Abstract
Allergic diseases are a heterogeneous group of disorders resulting from exaggerated type 2 inflammation. Although typically viewed as polygenic multifactorial disorders caused by the interaction of several genes with the environment, we have come to appreciate that allergic diseases can also be caused by monogenic variants affecting the immune system and the skin epithelial barrier. Through a myriad of genetic association studies and high-throughput sequencing tools, many monogenic and polygenic culprits of allergic diseases have been described. Identifying the genetic causes of atopy has shaped our understanding of how these conditions occur and how they may be treated and even prevented. Precision diagnostic tools and therapies that address the specific molecular pathways implicated in allergic inflammation provide exciting opportunities to improve our care for patients across the field of allergy and immunology. Here, we highlight offender genes implicated in polygenic and monogenic allergic diseases and list targeted therapeutic approaches that address these disrupted pathways.
Collapse
Affiliation(s)
- Maryam Vaseghi-Shanjani
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Andrew L Snow
- Department of Pharmacology and Molecular Therapeutics, Uniformed Services University of the Health Sciences, Bethesda, Md
| | - David J Margolis
- Department of Dermatology and Dermatologic Surgery, University of Pennsylvania Medical Center, Philadelphia, Pa; Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Medical Center, Philadelphia, Pa
| | - Meriem Latrous
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Joshua D Milner
- Department of Pediatrics, Columbia University Irving Medical Center, New York, NY
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Experimental Medicine Program, Faculty of Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Catherine M Biggs
- Department of Pediatrics, British Columbia Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; St Paul's Hospital, Vancouver, British Columbia, Canada.
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Weliwitigoda A, Palle P, Gessner M, Hubbard NW, Oukka M, Bettelli E. Cutting Edge: DOCK8 Regulates a Subset of Dendritic Cells That Is Critical for the Development of Experimental Autoimmune Encephalomyelitis. THE JOURNAL OF IMMUNOLOGY 2021; 207:2417-2422. [PMID: 34663621 DOI: 10.4049/jimmunol.2001294] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Accepted: 09/22/2021] [Indexed: 01/03/2023]
Abstract
Dedicator of cytokinesis 8 (DOCK8) is a guanine nucleotide exchange factor with an essential role in cytoskeletal rearrangement, cell migration, and survival of various immune cells. Interestingly, DOCK8-deficient mice are resistant to the development of experimental autoimmune encephalomyelitis (EAE). To understand if EAE resistance in these mice results from an alteration in dendritic cell (DC) functions, we generated mice with conditional deletion of DOCK8 in DCs and observed attenuated EAE in these mice compared with control mice. Additionally, we demonstrated that DOCK8 is important for the existence of splenic conventional DC2 and lymph node migratory DCs and further established that migratory DC, rather than resident DC, are essential for the generation and proliferation of pathogenic T cell populations upon immunization with myelin Ag in adjuvant. Therefore, our data suggest that limiting migratory DCs through DOCK8 deletion and possibly other mechanisms could limit the development of CNS autoimmunity.
Collapse
Affiliation(s)
- Asanga Weliwitigoda
- Immunology Program, Benaroya Research Institute, Seattle, WA; and.,Department of Immunology, University of Washington, Seattle, WA
| | - Pushpalatha Palle
- Immunology Program, Benaroya Research Institute, Seattle, WA; and.,Department of Immunology, University of Washington, Seattle, WA
| | - Melissa Gessner
- Immunology Program, Benaroya Research Institute, Seattle, WA; and.,Department of Immunology, University of Washington, Seattle, WA
| | | | - Mohamed Oukka
- Department of Immunology, University of Washington, Seattle, WA
| | - Estelle Bettelli
- Immunology Program, Benaroya Research Institute, Seattle, WA; and .,Department of Immunology, University of Washington, Seattle, WA
| |
Collapse
|
16
|
Hashim IF, Ahmad Mokhtar AM. Small Rho GTPases and their associated RhoGEFs mutations promote immunological defects in primary immunodeficiencies. Int J Biochem Cell Biol 2021; 137:106034. [PMID: 34216756 DOI: 10.1016/j.biocel.2021.106034] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/14/2021] [Accepted: 06/28/2021] [Indexed: 01/10/2023]
Abstract
Primary immunodeficiencies (PIDs) are associated with deleterious mutations of genes that encode proteins involved in actin cytoskeleton reorganisation. This deficiency affects haematopoietic cells. PID results in the defective function of immune cells, such as impaired chemokine-induced motility, receptor signalling, development and maturation. Some of the genes mutated in PIDs are related to small Ras homologous (Rho) guanosine triphosphatase (GTPase), one of the families of the Ras superfamily. Most of these genes act as molecular switches by cycling between active guanosine triphosphate-bound and inactive guanosine diphosphate-bound forms to control multiple cellular functions. They are best studied for their role in promoting cytoskeleton reorganisation, cell adhesion and motility. Currently, only three small Rho GTPases, namely, Rac2, Cdc42 and RhoH, have been identified in PIDs. However, several other Rho small G proteins might also contribute to the deregulation and phenotype observed in PIDs. Their contribution in PIDs may involve their main regulator, Rho guanine nucleotide exchange factors such as DOCK2 and DOCK8, wherein mutations may result in the impairment of small Rho GTPase activation. Thus, this review outlines the potential contribution of several small Rho GTPases to the promotion of PIDs.
Collapse
Affiliation(s)
- Ilie Fadzilah Hashim
- Primary Immunodeficiency Diseases Group, Regenerative Medicine Cluster, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Penang, 13200, Malaysia.
| | - Ana Masara Ahmad Mokhtar
- Bioprocess Technology Division, School of Industrial Technology, Universiti Sains Malaysia, Gelugor, Penang, 11800, Malaysia.
| |
Collapse
|
17
|
Zhang Z, Bao Y, Zhou L, Ye Y, Fu W, Sun C. DOCK8 Serves as a Prognostic Biomarker and Is Related to Immune Infiltration in Patients With HPV Positive Head and Neck Squamous Cell Carcinoma. Cancer Control 2021; 28:10732748211011951. [PMID: 33910393 PMCID: PMC8482706 DOI: 10.1177/10732748211011951] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Purpose: Dedicator of cytokinesis 8 (DOCK8) was reported to have a vital link to immunoregulation. However, the mechanisms by which it drives immune infiltration in cancer remain uncertain. We tried to assess the role of DOCK8 in patients with cancer, especially human papillomavirus (HPV)-positive head and neck squamous cell carcinoma (HNSCC). Methods: Data on the expression and survival of DOCK8 in patients with various cancers were analyzed using the Oncomine and TIMER databases. The TIMER database assessed the relationship of DOCK8 with immune infiltration levels and various markers of multiple immune cells. Gene set enrichment analysis revealed tumor-associated biological processes related to DOCK8. ENCODE database was used to explore relevant transcription factors of DOCK8, and a PPI network was constructed using GENEMINIA. The expression and survival role of DOCK8 was confirmed in patients from independent GEO datasets. Results: We determined that DOCK8 expression was upregulated or downregulated in various cancers unlike in healthy tissues. A high expression of DOCK8 was significantly correlated with a favorable prognosis in HPV-positive HNSCC and lung adenocarcinoma (LUAD). Furthermore, multivariate Cox regression analysis revealed that DOCK8 was an independent prognostic factor of HPV-positive HNSCC. Additionally, elevated DOCK8 expression was positively correlated with multiple immune cell infiltration levels and immune marker expression associated with particular immune cell subsets. Also, 14 pathways involved in immune activities and carcinogenesis, 22 potential TFs, and co-expression proteins of DOCK8 indicated DOCK8 to be related to tumor-associated biological processes. Ultimately, we verified that DOCK8 is upregulated and confers a favorable overall survival and progression-free survival status in patients with HPV-positive HNSCC. Conclusion: These results elucidate that high expression of DOCK8 indicates a favorable prognosis in patients with HPV-positive HNSCC as well as increased microenvironmental immune infiltration levels. It would provide new insights into the prognosis predicting and clinical regimen decision making in patients with HPV-positive HNSCC.
Collapse
Affiliation(s)
- Zeying Zhang
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China.,Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, Liaoning, China.,Department of Medical Genetics, China Medical University, Shenyang, China
| | - Yandong Bao
- Department of Cardiology, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, China
| | - Lu Zhou
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China.,Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, Liaoning, China
| | - Yanling Ye
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China.,Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, Liaoning, China
| | - Weineng Fu
- Department of Medical Genetics, China Medical University, Shenyang, China
| | - Changfu Sun
- Department of Oromaxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Shenyang, Liaoning, China.,Liaoning Provincial Key Laboratory of Oral Diseases, China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
18
|
Seo E, Lee BH, Lee JH, Park YS, Im HJ, Lee J. Hematopoietic stem cell transplantation in an infant with dedicator of cytokinesis 8 (DOCK8) deficiency associated with systemic lupus erythematosus: A case report. Medicine (Baltimore) 2021; 100:e20866. [PMID: 33787566 PMCID: PMC8021304 DOI: 10.1097/md.0000000000020866] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 05/21/2020] [Indexed: 01/04/2023] Open
Abstract
INTRODUCTION DOCK8 deficiency is a primary immunodeficiency characterized by recurrent infections, severe allergic disease, and autoimmunity. Here, we report a patient with DOCK8 deficiency that was initially presented as systemic lupus erythematosus (SLE) without recurrent infections and treated with hematopoietic stem cell transplantation (HSCT). PATIENT CONCERNS A 16-month-old boy with a previous history of eczema developed high fever and hand and foot swelling. Over time, multiple purpura, oral ulcers, and oliguria developed with a persistent fever. His laboratory findings showed anemia, thrombocytopenia, and coagulopathy with a high level of C-reactive protein (CRP). No definite pathogens were identified. The complement fractions C3, C4, and CH50 were low. Autoantibodies including antinuclear antibody (ANA) and anti-ds DNA antibody were positive. He definitively satisfied the 2015 ACR/SLICC revised criteria for the diagnosis of SLE (7 points out of 16); therefore, he was treated with a steroid. Lupus nephritis was confirmed by renal biopsy later. Considering the early-onset SLE, partial exome sequencing was performed. DIAGNOSIS One heterozygous missense variant, c.5536A>G (p.Lys1846Glu), which was inherited from his father, and heterozygous deletion of exon 1 to 8 inherited from his mother were found. Through the results of the genetic testing, the patient was confirmed to have DOCK8 deficiency. INTERVENTIONS At the age of 28 months, he received haploidentical HSCT from his mother as a donor. OUTCOMES Laboratory findings including complement fractions C3, C4, CH50, anti-ds DNA antibody, and the ANA became normal after HSCT. Currently, at 12 months post-HSCT, he is doing well, without any autoimmune features or infections. CONCLUSIONS DOCK8 deficiency can be presented as autoimmune disease such as SLE. Encountering a child diagnosed with SLE at a very young age, pediatricians should consider immunodeficiency syndrome including DOCK8 deficiency.
Collapse
Affiliation(s)
- Euri Seo
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
- Department of Pediatrics, Dongguk University Ilsan Hospital, Goyang, Korea
| | - Beom Hee Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Joo Hoon Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Young Seo Park
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Ho Joon Im
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| | - Jina Lee
- Department of Pediatrics, Asan Medical Center Children's Hospital, University of Ulsan College of Medicine, Seoul
| |
Collapse
|
19
|
Capkova Z, Capkova P, Srovnal J, Adamova K, Prochazka M, Hajduch M. Duplication of 9p24.3 in three unrelated patients and their phenotypes, considering affected genes, and similar recurrent variants. Mol Genet Genomic Med 2021; 9:e1592. [PMID: 33455084 PMCID: PMC8104183 DOI: 10.1002/mgg3.1592] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Revised: 11/22/2020] [Accepted: 12/15/2020] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Recent studies suggest that duplication of the 9p24.3 chromosomal locus, which includes the DOCK8 and KANK1 genes, is associated with autism spectrum disorders (ASD), intellectual disability/developmental delay (ID/DD), learning problems, language disorders, hyperactivity, and epilepsy. Correlation between this duplication and the carrier phenotype needs further discussion. METHODS In this study, three unrelated patients with ID/DD and ASD underwent SNP aCGH and MLPA testing. Similarities in the phenotypes of patients with 9p24.3, 15q11.2, and 16p11.2 duplications were also observed. RESULTS All patients with ID/DD and ASD carried the 9p24.3 duplication and showed intragenic duplication of DOCK8. Additionally, two patients had ADHD, one was hearing impaired and obese, and one had macrocephaly. Inheritance of the 9p24.3 duplication was confirmed in one patient and his sibling. In one patient KANK1 was duplicated along with DOCK8. Carriers of 9p24.3, 15q11.2, and 16p11.2 duplications showed several phenotypic similarities, with ID/DD more strongly associated with duplication of 9p24.3 than of 15q11.2 and 16p11.2. CONCLUSION We concluded that 9p24.3 is a likely cause of ASD and ID/DD, especially in cases of DOCK8 intragenic duplication. DOCK8 is a likely causative gene, and KANK1 aberrations a modulator, of the clinical phenotype observed. Other modulators were not excluded.
Collapse
Affiliation(s)
- Zuzana Capkova
- Department of Medical GeneticsUniversity Hospital OlomoucOlomoucCzech Republic
- Department of Medical GeneticsFaculty of Medicine and DentistryPalacký University OlomoucOlomoucCzech Republic
| | - Pavlina Capkova
- Department of Medical GeneticsUniversity Hospital OlomoucOlomoucCzech Republic
- Department of Medical GeneticsFaculty of Medicine and DentistryPalacký University OlomoucOlomoucCzech Republic
| | - Josef Srovnal
- Department of Medical GeneticsUniversity Hospital OlomoucOlomoucCzech Republic
- Institute of Molecular and Translational MedicineFaculty of Medicine and DentistryPalacký University OlomoucOlomoucCzech Republic
| | - Katerina Adamova
- Department of Medical GeneticsUniversity Hospital OlomoucOlomoucCzech Republic
- Department of Medical GeneticsFaculty of Medicine and DentistryPalacký University OlomoucOlomoucCzech Republic
| | - Martin Prochazka
- Department of Medical GeneticsUniversity Hospital OlomoucOlomoucCzech Republic
- Department of Medical GeneticsFaculty of Medicine and DentistryPalacký University OlomoucOlomoucCzech Republic
| | - Marian Hajduch
- Institute of Molecular and Translational MedicineFaculty of Medicine and DentistryPalacký University OlomoucOlomoucCzech Republic
| |
Collapse
|
20
|
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
|
21
|
Wang K, Fu W. Transcriptional regulation of Treg homeostasis and functional specification. Cell Mol Life Sci 2020; 77:4269-4287. [PMID: 32350553 PMCID: PMC7606275 DOI: 10.1007/s00018-020-03534-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 04/19/2020] [Accepted: 04/20/2020] [Indexed: 12/15/2022]
Abstract
CD4+Foxp3+ regulatory T (Treg) cells are key players in keeping excessive inflammation in check. Mounting evidence has shown that Treg cells exert much more diverse functions in both immunological and non-immunological processes. The development, maintenance and functional specification of Treg cells are regulated by multilayered factors, including antigens and TCR signaling, cytokines, epigenetic modifiers and transcription factors (TFs). In the review, we will focus on TFs by summarizing their unique and redundant roles in Treg cells under physiological and pathophysiological conditions. We will also discuss the recent advances of Treg trajectories between lymphoid organs and non-lymphoid tissues. This review will provide an updated view of the newly identified TFs and new functions of known TFs in Treg biology.
Collapse
Affiliation(s)
- Ke Wang
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA
| | - Wenxian Fu
- Pediatric Diabetes Research Center, Department of Pediatrics, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
- Moores Cancer Center, University of California San Diego, 9500 Gilman Drive, La Jolla, CA, 92093, USA.
| |
Collapse
|
22
|
Eken A, Cansever M, Okus FZ, Erdem S, Nain E, Azizoglu ZB, Haliloglu Y, Karakukcu M, Ozcan A, Devecioglu O, Aksu G, Arikan Ayyildiz Z, Topal E, Karakoc Aydiner E, Kiykim A, Metin A, Cipe F, Kaya A, Artac H, Reisli I, Guner SN, Uygun V, Karasu G, Dönmez Altuntas H, Canatan H, Oukka M, Ozen A, Chatila TA, Keles S, Baris S, Unal E, Patiroglu T. ILC3 deficiency and generalized ILC abnormalities in DOCK8-deficient patients. Allergy 2020; 75:921-932. [PMID: 31596517 DOI: 10.1111/all.14081] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Revised: 09/10/2019] [Accepted: 09/20/2019] [Indexed: 12/15/2022]
Abstract
BACKGROUND Dedicator of cytokinesis 8 (DOCK8) deficiency is the main cause of the autosomal recessive hyper-IgE syndrome (HIES). We previously reported the selective loss of group 3 innate lymphoid cell (ILC) number and function in a Dock8-deficient mouse model. In this study, we sought to test whether DOCK8 is required for the function and maintenance of ILC subsets in humans. METHODS Peripheral blood ILC1-3 subsets of 16 DOCK8-deficient patients recruited at the pretransplant stage, and seven patients with autosomal dominant (AD) HIES due to STAT3 mutations, were compared with those of healthy controls or post-transplant DOCK8-deficient patients (n = 12) by flow cytometry and real-time qPCR. Sorted total ILCs from DOCK8- or STAT3-mutant patients and healthy controls were assayed for survival, apoptosis, proliferation, and activation by IL-7, IL-23, and IL-12 by cell culture, flow cytometry, and phospho-flow assays. RESULTS DOCK8-deficient but not STAT3-mutant patients exhibited a profound depletion of ILC3s, and to a lesser extent ILC2s, in their peripheral blood. DOCK8-deficient ILC1-3 subsets had defective proliferation, expressed lower levels of IL-7R, responded less to IL-7, IL-12, or IL-23 cytokines, and were more prone to apoptosis compared with those of healthy controls. CONCLUSION DOCK8 regulates human ILC3 expansion and survival, and more globally ILC cytokine signaling and proliferation. DOCK8 deficiency leads to loss of ILC3 from peripheral blood. ILC3 deficiency may contribute to the susceptibility of DOCK8-deficient patients to infections.
Collapse
|
23
|
Janssen E, Geha RS. Primary immunodeficiencies caused by mutations in actin regulatory proteins. Immunol Rev 2019; 287:121-134. [PMID: 30565251 DOI: 10.1111/imr.12716] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 08/31/2018] [Indexed: 12/31/2022]
Abstract
The identification of patients with monogenic gene defects have illuminated the function of different proteins in the immune system, including proteins that regulate the actin cytoskeleton. Many of these actin regulatory proteins are exclusively expressed in leukocytes and regulate the formation and branching of actin filaments. Their absence or abnormal function leads to defects in immune cell shape, cellular projections, migration, and signaling. Through the study of patients' mutations and generation of mouse models that recapitulate the patients' phenotypes, our laboratory and others have gained a better understanding of the role these proteins play in cell biology and the underlying pathogenesis of immunodeficiencies and immune dysregulatory syndromes.
Collapse
Affiliation(s)
- Erin Janssen
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| | - Raif S Geha
- Division of Immunology, Department of Pediatrics, Boston Children's Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
24
|
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
|
25
|
Su HC, Jing H, Angelus P, Freeman AF. Insights into immunity from clinical and basic science studies of DOCK8 immunodeficiency syndrome. Immunol Rev 2019; 287:9-19. [PMID: 30565250 PMCID: PMC6350515 DOI: 10.1111/imr.12723] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 12/29/2022]
Abstract
DOCK8 immunodeficiency syndrome (DIDS) is a progressive combined immunodeficiency that can be distinguished from other combined immunodeficiencies or hyperimmunoglobulinemia E syndromes in featuring (a) profound susceptibility to virus infections of the skin, with associated skin cancers, and (b) severe food allergies. The DOCK8 locus has many repetitive sequence elements that predispose to the generation of large germline deletions as well as recombination-mediated somatic DNA repair. Residual DOCK8 protein contributes to the variable disease phenotype. The severe virus infections of the skin, and probably also VZV-associated vasculopathy, reflect an important function of DOCK8, which is normally required to maintain lymphocyte shape integrity as the cells migrate through dense tissues. Loss of DOCK8 also causes immune deficits through other mechanisms including a milder generalized cell survival defect and skewing of T helper cell subsets. Recent work has uncovered the roles for DOCK8 in dendritic cell responses that can also help explain the virus susceptibility, as well as in regulatory T cells that might help explain autoimmunity in a minority of patients. Fortunately, hematopoietic stem cell transplantation cures the eczema and infection susceptibility of DIDS, but not necessarily the other disease manifestations including food allergies.
Collapse
Affiliation(s)
- Helen C. Su
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Huie Jing
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| | - Pam Angelus
- Clinical Monitoring Research Program Directorate, Frederick National Laboratory for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Alexandra F. Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health
| |
Collapse
|
26
|
Al-Shaikhly T, Ochs HD. Hyper IgE syndromes: clinical and molecular characteristics. Immunol Cell Biol 2018; 97:368-379. [PMID: 30264496 DOI: 10.1111/imcb.12209] [Citation(s) in RCA: 79] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 09/20/2018] [Accepted: 09/24/2018] [Indexed: 01/18/2023]
Abstract
Hyper IgE syndromes comprise a group of rare primary immunodeficiency disorders characterized by a triad of atopic dermatitis, recurrent skin and lung infections along with elevated IgE levels. Job syndrome or autosomal dominant hyper IgE syndrome because of heterozygous loss-of-function mutations with dominant negative effect in signal transducer and activator of transcription-3 is the prototype of these disorders. However, several other genetically characterized immunodeficiency disorders have been identified over the past decade and joined the umbrella of hyper IgE syndromes including autosomal recessive mutations in the DOCK8, ZNF431 and PGM3 genes and heterozygous mutations with dominant negative effect in the CARD11 gene. Moreover, a number of phenotypically distinct immunodeficiency disorders can mimic hyper IgE syndromes, adding to the diagnostic challenge. Herein, we will concisely review these disorders, their molecular bases, highlighting key distinguishing clinical and laboratory findings and therapeutic options.
Collapse
Affiliation(s)
- Taha Al-Shaikhly
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Hans D Ochs
- Seattle Children's Research Institute, Seattle, Washington, USA.,Department of Pediatrics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
27
|
Interaction of septin 7 and DOCK8 in equine lymphocytes reveals novel insights into signaling pathways associated with autoimmunity. Sci Rep 2018; 8:12332. [PMID: 30120291 PMCID: PMC6098150 DOI: 10.1038/s41598-018-30753-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Accepted: 06/22/2018] [Indexed: 01/21/2023] Open
Abstract
The GTP-binding protein septin 7 is involved in various cellular processes, including cytoskeleton organization, migration and the regulation of cell shape. Septin 7 function in lymphocytes, however, is poorly characterized. Since the intracellular signaling role of septin 7 is dependent on its interaction network, interaction proteomics was applied to attain novel knowledge about septin 7 function in hematopoietic cells. Our previous finding of decreased septin 7 expression in blood-derived lymphocytes in ERU, a spontaneous animal model for autoimmune uveitis in man, extended the role of septin 7 to a potential key player in autoimmunity. Here, we revealed novel insights into septin 7 function by identification of DOCK8 as an interaction partner in primary blood-derived lymphocytes. Since DOCK8 is associated with important immune functions, our finding of significantly decreased DOCK8 expression and altered DOCK8 interaction network in ERU might explain changes in immune response and shows the contribution of DOCK8 in pathomechanisms of spontaneous autoimmune diseases. Moreover, our analyses revealed insights in DOCK8 function, by identifying the signal transducer ILK as a DOCK8 interactor in lymphocytes. Our finding of the enhanced enrichment of ILK in ERU cases indicates a deviant influence of DOCK8 on inter- and intracellular signaling in autoimmune disease.
Collapse
|
28
|
Zhang Q, Boisson B, Béziat V, Puel A, Casanova JL. Human hyper-IgE syndrome: singular or plural? Mamm Genome 2018; 29:603-617. [PMID: 30094507 PMCID: PMC6317873 DOI: 10.1007/s00335-018-9767-2] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/02/2018] [Indexed: 12/15/2022]
Abstract
Spectacular progress has been made in the characterization of human hyper-IgE syndrome (HIES) over the last 50 years. HIES is a primary immunodeficiency defined as an association of atopy in a context of very high serum IgE levels, characteristic bacterial and fungal diseases, low-level clinical and biological inflammation, and various non-hematopoietic developmental manifestations. Somewhat arbitrarily, three disorders were successively put forward as the underlying cause of HIES: autosomal dominant (AD) STAT3 deficiency, the only disorder corresponding to the original definition of HIES, and autosomal recessive (AR) DOCK8 and PGM3 deficiencies, in which atopy and high serum IgE levels occur in a context of manifestations not seen in patients with typical HIES. Indeed, these three disorders disrupt different molecular pathways, affect different cell types, and underlie different clinical phenotypes. Surprisingly, several other inherited inborn errors of immunity in which serum IgE levels are high, sometimes almost as high as those in HIES patients, are not considered to belong to the HIES group of diseases. Studies of HIES have been further complicated by the lack of a high serum IgE phenotype in all mouse models of the disease other than two Stat3 mutant strains. The study of infections in mutant mice has helped elucidate only some forms of HIES and infection. Mouse models of these conditions have also been used to study non-hematopoietic phenotypes for STAT3 deficiency, tissue-specific immunity for DOCK8 deficiency, and cell lineage maturation for PGM3 deficiency. We review here the history of the field of HIES since the first clinical description of this condition in 1966, together with the three disorders commonly referred to as HIES, focusing, in particular, on their mouse models. We propose the restriction of the term "HIES" to patients with an AD STAT3-deficiency phenotype, including the most recently described AR ZNF341 deficiency, thus excluding AR DOCK8 and PGM3 deficiencies from the definition of this disease.
Collapse
Affiliation(s)
- Qian Zhang
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA.
| | - Bertrand Boisson
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Descartes University, Imagine Institute, Paris, France
| | - Vivien Béziat
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Descartes University, Imagine Institute, Paris, France
| | - Anne Puel
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Descartes University, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- St Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY, USA
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Paris, France
- Paris Descartes University, Imagine Institute, Paris, France
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, AP-HP, 75015, Paris, France
- Howard Hughes Medical Institute, New York, NY, USA
| |
Collapse
|
29
|
Milner JD. TCR Signaling Abnormalities in Human Th2-Associated Atopic Disease. Front Immunol 2018; 9:719. [PMID: 29713322 PMCID: PMC5911486 DOI: 10.3389/fimmu.2018.00719] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2018] [Accepted: 03/22/2018] [Indexed: 11/15/2022] Open
Abstract
Stimulation of naïve CD4 T cells with weak T cell receptor agonists even in the absence of T helper-skewing cytokines can result in IL-4 production which can drive a Th2 response. Evidence for the in vivo consequences of such a phenomenon can be found in a number of mouse models and, importantly, a series of monogenic human diseases associated with significant atopy which are caused by mutations in the T cell receptor signaling cascade. Such diseases can help understand how Th2 responses evolve in humans, and potentially provide insight into therapeutic interventions.
Collapse
Affiliation(s)
- Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID), National Institutes of Health (NIH), Bethesda, MD, United States
| |
Collapse
|
30
|
Lyons JJ, Milner JD. Primary atopic disorders. J Exp Med 2018; 215:1009-1022. [PMID: 29549114 PMCID: PMC5881472 DOI: 10.1084/jem.20172306] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 02/21/2018] [Accepted: 03/01/2018] [Indexed: 12/19/2022] Open
Abstract
Important insights from monogenic disorders into the immunopathogenesis of allergic diseases and reactions are discussed. Monogenic disorders have provided fundamental insights into human immunity and the pathogenesis of allergic diseases. The pathways identified as critical in the development of atopy range from focal defects in immune cells and epithelial barrier function to global changes in metabolism. A major goal of studying heritable single-gene disorders that lead to severe clinical allergic diseases is to identify fundamental pathways leading to hypersensitivity that can be targeted to provide novel therapeutic strategies for patients with allergic diseases, syndromic and nonsyndromic alike. Here, we review known single-gene disorders leading to severe allergic phenotypes in humans, discuss how the revealed pathways fit within our current understanding of the atopic diathesis, and propose how some pathways might be targeted for therapeutic benefit.
Collapse
Affiliation(s)
- Jonathan J Lyons
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD
| |
Collapse
|