51
|
Nelson RW, Geha RS, McDonald DR. Inborn Errors of the Immune System Associated With Atopy. Front Immunol 2022; 13:860821. [PMID: 35572516 PMCID: PMC9094424 DOI: 10.3389/fimmu.2022.860821] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 03/28/2022] [Indexed: 11/13/2022] Open
Abstract
Atopic disorders, including atopic dermatitis, food and environmental allergies, and asthma, are increasingly prevalent diseases. Atopic disorders are often associated with eosinophilia, driven by T helper type 2 (Th2) immune responses, and triggered by disrupted barrier function leading to abnormal immune priming in a susceptible host. Immune deficiencies, in contrast, occur with a significantly lower incidence, but are associated with greater morbidity and mortality. A subset of atopic disorders with eosinophilia and elevated IgE are associated with monogenic inborn errors of immunity (IEI). In this review, we discuss current knowledge of IEI that are associated with atopy and the lessons these immunologic disorders provide regarding the fundamental mechanisms that regulate type 2 immunity in humans. We also discuss further mechanistic insights provided by animal models.
Collapse
Affiliation(s)
- Ryan W Nelson
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Raif S Geha
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| | - Douglas R McDonald
- Division of Immunology, Boston Children's Hospital, Harvard Medical School, Boston, MA, United States
| |
Collapse
|
52
|
Dai J, Zhang X, Zhang J, Yang W, Yang X, Bian H, Chen Z. Blockade of mIL‐6R alleviated lipopolysaccharide‐induced systemic inflammatory response syndrome by suppressing NF‐κB‐mediated Ccl2 expression and inflammasome activation. MedComm (Beijing) 2022; 3:e132. [PMID: 35548710 PMCID: PMC9075038 DOI: 10.1002/mco2.132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2021] [Revised: 03/16/2022] [Accepted: 03/18/2022] [Indexed: 12/04/2022] Open
Abstract
Systemic inflammatory response syndrome (SIRS) is characterized by dysregulated cytokine release, immune responses and is associated with organ dysfunction. IL‐6R blockade indicates promising therapeutic effects in cytokine release storm but still remains unknown in SIRS. To address the issue, we generated the human il‐6r knock‐in mice and a defined epitope murine anti‐human membrane‐bound IL‐6R (mIL‐6R) mAb named h‐mIL‐6R mAb. We found that the h‐mIL‐6R and the commercial IL‐6R mAb Tocilizumab significantly improved the survival rate, reduced the levels of TNF‐α, IL‐6, IL‐1β, IFN‐γ, transaminases and blood urea nitrogen of LPS‐induced SIRS mice. Besides, the h‐mIL‐6R mAb could also dramatically reduce the levels of inflammatory cytokines in LPS‐treated THP‐1 cells in vitro. RNA‐seq analysis indicated that the h‐mIL‐6R mAb could regulate LPS‐induced activation of NF‐κB/Ccl2 and NOD‐like receptor signaling pathways. Furthermore, we found that the h‐mIL‐6R mAb could forwardly inhibit Ccl2 expression and NLRP3‐mediated pyroptosis by suppressing NF‐κB in combination with the NF‐κB inhibitor. Collectively, mIL‐6R mAbs suppressed NF‐κB/Ccl2 signaling and inflammasome activation. IL‐6R mAbs are potential alternative therapeutics for suppressing excessive cytokine release, over‐activated inflammatory responses and alleviating organ injuries in SIRS.
Collapse
Affiliation(s)
- Ji‐Min Dai
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
- Faculty of Hepato‐Biliary‐Pancreatic Surgery The First Medical Center of Chinese People's Liberation Army (PLA) General Hospital Beijing P.R. China
| | - Xue‐Qin Zhang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Jia‐Jia Zhang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Wei‐Jie Yang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Xiang‐Min Yang
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Huijie Bian
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| | - Zhi‐Nan Chen
- National Translational Science Center for Molecular Medicine&Department of Cell Biology State Key Laboratory of Cancer Biology the Fourth Military Medical University Xi'an P.R. China
| |
Collapse
|
53
|
Olbrich P, Ortiz Aljaro P, Freeman AF. Eosinophilia Associated With Immune Deficiency. THE JOURNAL OF ALLERGY AND CLINICAL IMMUNOLOGY. IN PRACTICE 2022; 10:1140-1153. [PMID: 35227935 DOI: 10.1016/j.jaip.2022.02.016] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/16/2022] [Accepted: 02/19/2022] [Indexed: 06/14/2023]
Abstract
The differential diagnosis of eosinophilia is broad and includes infections, malignancies, and atopy as well as inborn errors of immunity (IEI). Certain types of IEIs are known to be associated with elevated numbers of eosinophils and frequently elevated serum IgE, whereas for others the degree and frequency of eosinophilia are less established. The molecular defects underlying IEI are heterogeneous and affect different pathways, which highlights the complex regulations of this cell population within the immune system. In this review, we list and discuss clinical manifestations and therapies of immune deficiency or immune dysregulation disorders associated with peripheral blood or tissue eosinophilia with or without raised IgE levels. We present illustrative case vignettes for the most common entities and propose a diagnostic algorithm aiming to help physicians systematically to evaluate patients with eosinophilia and suspicion of an underlying IEI.
Collapse
Affiliation(s)
- Peter Olbrich
- Sección Infectología, Reumatología e Inmunología Pediátrica, UGC de Pediatría, Hospital Universitario Virgen del Rocío, Seville, Spain; Laboratorio de Alteraciones Congénitas de la Inmunidad, Laboratorio 205, Instituto de Biomedicina de Sevilla, Seville, Spain; Departamento de Farmacología, Pediatría y Radiología, Facultad de Medicina, Universidad de Sevilla, Spain.
| | - Pilar Ortiz Aljaro
- Servicio de Inmunología, Hospital Universitario Virgen del Rocío (IBiS, CSIC, US), Seville, Spain
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, Bethesda, Md
| |
Collapse
|
54
|
Conti F, Marzollo A, Moratti M, Lodi L, Ricci S. Inborn Errors of Immunity underlying a susceptibility to pyogenic infections: from innate immune system deficiency to complex phenotypes. Clin Microbiol Infect 2022; 28:1422-1428. [DOI: 10.1016/j.cmi.2022.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 04/29/2022] [Accepted: 05/14/2022] [Indexed: 12/26/2022]
|
55
|
Dowling AR, Luke CE, Cai Q, Pellerito AM, Obi AT, Henke PK. Modulation of interleukin-6 and its effect on late vein wall injury in a stasis mouse model of deep vein thrombosis. JVS Vasc Sci 2022; 3:246-255. [PMID: 35647566 PMCID: PMC9133633 DOI: 10.1016/j.jvssci.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 04/04/2022] [Indexed: 12/02/2022] Open
Abstract
Objective Deep vein thrombosis (DVT) and its sequela, post-thrombotic syndrome (PTS), remain a clinically significant problem. Interleukin-6 (IL-6) is a proinflammatory cytokine that is elevated in patients who develop PTS. We hypothesized that genetic deletion of IL-6 and the use of anti-IL-6 pharmacologic agents would be associated with decreased late vein wall injury. Methods Wild-type C57BL/6J (WT) and IL-6-/- mice underwent induction of stasis venous thrombosis by ligation of the infrarenal IVC. Vein wall inferior vena cava and thrombus were harvested at 21 days after ligation and analyzed by Western blot and immunohistochemistry of the vein wall using monocyte markers CCR2 and arginase 1, the endothelial marker CD31, and fibroblast markers DDR2 and FSP-1. Two anti-IL-6 pharmacologic agents (gp130 [glycoprotein 130] and tocilizumab) were tested and compared with low-molecular-weight heparin (LMWH) as the reference standard in WT mice. Plasma was collected at 4 and 48 hours to confirm the pharmacologic agents' effects. Results Less fibrosis but no increase in luminal endothelialization was found in IL-6-/- mice compared with WT mice at 21 days. The IL-6-/- mice had fewer DDR2- and arginase 1-positive cells in the vein wall compared with the WT mice. However, no difference was found in the CCR2+ cells. Despite documented in vivo activity, exogenous gp130 and tocilizumab were not associated with decreased vein wall fibrosis or increased endothelial luminal coverage at 21 days. LMWH therapy, both before and after treatment, was not associated with decreased vein wall fibrosis at 21 days. Conclusions IL-6 genetic deletion was associated with less fibrotic vein wall injury at a late time point, consistent with the PTS timeframe. However, neither the standard of care LMWH nor two available anti-IL-6 agents showed antifibrotic biologic effects in this model.
Collapse
Affiliation(s)
- Abigail R. Dowling
- Conrad Jobst Vascular Surgery Research Laboratories, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
- Vascular Surgery Section, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
| | - Catherine E. Luke
- Conrad Jobst Vascular Surgery Research Laboratories, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
- Vascular Surgery Section, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
| | - Qing Cai
- Conrad Jobst Vascular Surgery Research Laboratories, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
- Vascular Surgery Section, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
| | - Antonio M. Pellerito
- Conrad Jobst Vascular Surgery Research Laboratories, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
| | - Andrea T. Obi
- Conrad Jobst Vascular Surgery Research Laboratories, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
- Vascular Surgery Section, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
| | - Peter K. Henke
- Conrad Jobst Vascular Surgery Research Laboratories, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
- Vascular Surgery Section, Department of Surgery, College of Medicine, University of Michigan, Ann Arbor, MI
| |
Collapse
|
56
|
Puel A, Bastard P, Bustamante J, Casanova JL. Human autoantibodies underlying infectious diseases. J Exp Med 2022; 219:e20211387. [PMID: 35319722 PMCID: PMC8952682 DOI: 10.1084/jem.20211387] [Citation(s) in RCA: 66] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 03/07/2022] [Accepted: 03/08/2022] [Indexed: 12/14/2022] Open
Abstract
The vast interindividual clinical variability observed in any microbial infection-ranging from silent infection to lethal disease-is increasingly being explained by human genetic and immunological determinants. Autoantibodies neutralizing specific cytokines underlie the same infectious diseases as inborn errors of the corresponding cytokine or response pathway. Autoantibodies against type I IFNs underlie COVID-19 pneumonia and adverse reactions to the live attenuated yellow fever virus vaccine. Autoantibodies against type II IFN underlie severe disease caused by environmental or tuberculous mycobacteria, and other intra-macrophagic microbes. Autoantibodies against IL-17A/F and IL-6 are less common and underlie mucocutaneous candidiasis and staphylococcal diseases, respectively. Inborn errors of and autoantibodies against GM-CSF underlie pulmonary alveolar proteinosis; associated infections are less well characterized. In individual patients, autoantibodies against cytokines preexist infection with the pathogen concerned and underlie the infectious disease. Human antibody-driven autoimmunity can interfere with cytokines that are essential for protective immunity to specific infectious agents but that are otherwise redundant, thereby underlying specific infectious diseases.
Collapse
Affiliation(s)
- Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Paul Bastard
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
| | - Jacinta Bustamante
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique – Hôpitaux de Paris, Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale, Necker Hospital for Sick Children, Paris, France
- Imagine Institute, Paris Cité University, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
- Department of Pediatrics, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, Paris, France
| |
Collapse
|
57
|
Hyper IgE syndromes: A clinical approach. Clin Immunol 2022; 237:108988. [DOI: 10.1016/j.clim.2022.108988] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2021] [Revised: 03/22/2022] [Accepted: 03/23/2022] [Indexed: 12/20/2022]
|
58
|
IL-6 Prevents Lung Macrophage Death and Lung Inflammation Injury by Inhibiting GSDME- and GSDMD-Mediated Pyroptosis during Pneumococcal Pneumosepsis. Microbiol Spectr 2022; 10:e0204921. [PMID: 35297653 PMCID: PMC9045248 DOI: 10.1128/spectrum.02049-21] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Streptococcus pneumoniae is a leading bacterial cause of a wide range of infections, and pneumococcal pneumosepsis causes high mortality in hosts infected with antibiotic-resistant strains and those who cannot resolve ongoing inflammation. The factors which influence the development and outcome of pneumosepsis are currently unclear. IL-6 is critical for maintaining immune homeostasis, and we determined that this cytokine is also essential for resisting pneumosepsis, as it inhibits macrophage pyroptosis and pyroptosis-related inflammation injury in the lung. IL-6 affected infection outcomes in mice and exerted a protective role, primarily via macrophages. We further found that IL-6 deficiency led to increased lung macrophage death and aggravated lung inflammation, and that exogenous administration of IL-6 protein could decrease macrophage death and alleviate lung tissue inflammation. IL-6 also protected Streptococcus pneumoniae-induced lung macrophage death and lung inflammation injury by inhibiting gasdermin E (GSDME)- and gasdermin D (GSDMD)-mediated pyroptosis. Together, these data reveal a novel mechanism for the development of pneumosepsis and the critical protective role of IL-6. These findings may assist in the early identification and treatment of pneumococcal pneumosepsis. IMPORTANCE Pneumococcal pneumonia has been a significant cause of morbidity and mortality throughout human history. Failing to control pneumococcal pneumonia and resolve ongoing inflammation in a host can cause sepsis, namely pneumococcal pneumosepsis, and death ensues. Few theories have suggested an optimally therapeutic option for this infectious disease. The interleukin-6 (IL-6, a cytokine featuring pleiotropic activity) theory, proposed here, implies that IL-6 acts as a protector against pneumococcal pneumosepsis. IL-6 prevents lung macrophage death and lung inflammation injury by inhibiting a caspase-3-GSDME-mediated switch from apoptosis to pyroptosis and inhibiting caspase-1-GSDMD-mediated classic pyroptosis during pneumococcal pneumosepsis. Thus, IL-6 is an important determinant for controlling bacterial invasion and a homeostatic coordinator of pneumococcal pneumosepsis. This study clarifies a novel mechanism of occurrence and development of pneumonia and secondary sepsis following a Streptococcus pneumoniae infection. It is important for the early identification and treatment of pneumococcal pneumosepsis.
Collapse
|
59
|
Yang Y, Xiao J, Tang L, Wang B, Sun X, Xu Z, Liu L, Shi S. Effects of IL-6 Polymorphisms on Individual Susceptibility to Allergic Diseases: A Systematic Review and Meta-Analysis. Front Genet 2022; 13:822091. [PMID: 35368692 PMCID: PMC8966614 DOI: 10.3389/fgene.2022.822091] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Accepted: 01/18/2022] [Indexed: 12/24/2022] Open
Abstract
Background: Many studies have assessed the potential link between interleukin-6 polymorphisms and susceptibility to allergic diseases. However, the results are still conflicting. Therefore, a comprehensive meta-analysis can not only resolve differences but also provide clues for future projects.Methods: A systematic electronic search was conducted on the databases of Web of Science, PubMed, and Cochrane Library to retrieve all published studies. Revman and Stata software were used for statistical analysis.Results: This meta-analysis included 11 studies. The results revealed that there was a statistically significant association between IL-6 rs1800795 polymorphism and the risk of asthma and allergic rhinitis in the general population. Subgroup analyses demonstrated that rs1800795 affected allergic diseases risk in different populations.Conclusion: Our findings suggested that IL-6 rs1800795 was associated with allergic diseases susceptibility among Asians and Caucasians in opposite trends, and it might influence the risk of asthma and allergic rhinitis. None of the IL-6 polymorphisms were shared risk variants of allergic diseases.
Collapse
Affiliation(s)
- Ying Yang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Jingxin Xiao
- Taizhou Hospital of Chinese Medicine, Taizhou, China
| | - Lingling Tang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Bohan Wang
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Xianhong Sun
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhongchi Xu
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
| | - Li Liu
- Laboratory of Molecular Biology, Central Laboratory, Jiangsu Province Hospital of Chinese Medicine, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Suofang Shi, ; Li Liu,
| | - Suofang Shi
- Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- *Correspondence: Suofang Shi, ; Li Liu,
| |
Collapse
|
60
|
Ferenc T, Vilibić-Čavlek T. COMMON VARIABLE IMMUNODEFICIENCY: PREDISPOSING OR PROTECTIVE FACTOR FOR SEVERE COMPLICATIONS OF COVID-19? Acta Clin Croat 2022; 61:107-114. [PMID: 36398083 PMCID: PMC9616023 DOI: 10.20471/acc.2022.61.01.13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 09/06/2021] [Indexed: 08/11/2023] Open
Abstract
Coronavirus disease 2019 (COVID-19) is an emerging infectious disease caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). The usual presentation of the disease is a common cold-like illness but it can present with more severe and sometimes fatal manifestations. Immunocompromised patients such as those with common variable immunodeficiency (CVID) also are among the infected population. A limited number of reports have been published concerning CVID patients with COVID-19. The main reported symptoms were fever, cough, dyspnea and fatigue while the median duration of illness was 19 (interquartile range 14-26.5) days. Total recovery rate was 88.4%. It is still unknown whether primary immunodeficiency interacts as a predisposing or protective factor against the severe forms of COVID-19. Substitute immunoglobulin (IG) therapy is the only treatment option for CVID. Some reports suggest that early administration of intravenous IGs or convalescent plasma infusion may positively influence the outcome of COVID-19 in these patients.
Collapse
Affiliation(s)
- Thomas Ferenc
- Department of Diagnostic and Interventional Radiology, Merkur University Hospital, Zagreb, Croatia
| | - Tatjana Vilibić-Čavlek
- School of Medicine, University of Zagreb, Zagreb, Croatia
- Department of Virology, Croatian Institute of Public Health, Zagreb, Croatia
| |
Collapse
|
61
|
Millrine D, Jenkins RH, Hughes STO, Jones SA. Making sense of IL-6 signalling cues in pathophysiology. FEBS Lett 2022; 596:567-588. [PMID: 34618359 PMCID: PMC9673051 DOI: 10.1002/1873-3468.14201] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 12/15/2022]
Abstract
Unravelling the molecular mechanisms that account for functional pleiotropy is a major challenge for researchers in cytokine biology. Cytokine-receptor cross-reactivity and shared signalling pathways are considered primary drivers of cytokine pleiotropy. However, reports epitomized by studies of Jak-STAT cytokine signalling identify interesting biochemical and epigenetic determinants of transcription factor regulation that affect the delivery of signal-dependent cytokine responses. Here, a regulatory interplay between STAT transcription factors and their convergence to specific genomic enhancers support the fine-tuning of cytokine responses controlling host immunity, functional identity, and tissue homeostasis and repair. In this review, we provide an overview of the signalling networks that shape the way cells sense and interpret cytokine cues. With an emphasis on the biology of interleukin-6, we highlight the importance of these mechanisms to both physiological processes and pathophysiological outcomes.
Collapse
Affiliation(s)
- David Millrine
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
- Present address:
Medical Research Council Protein Phosphorylation and Ubiquitylation UnitSir James Black CentreSchool of Life SciencesUniversity of Dundee3rd FloorDundeeUK
| | - Robert H. Jenkins
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Stuart T. O. Hughes
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| | - Simon A. Jones
- Division of Infection & ImmunitySchool of MedicineCardiff UniversityUK
- Systems Immunity University Research InstituteCardiff UniversityUK
| |
Collapse
|
62
|
Lachover-Roth I, Lagovsky I, Shtorch-Asor A, Confino-Cohen R, Reinstein E, Garty BZ. Hyper IgE Syndrome in an Isolated Population in Israel. Front Immunol 2022; 13:829239. [PMID: 35185921 PMCID: PMC8854367 DOI: 10.3389/fimmu.2022.829239] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2021] [Accepted: 01/20/2022] [Indexed: 11/24/2022] Open
Abstract
Introduction Hyper IgE syndromes (HIES) are a group of rare primary immunodeficiency characterized by high levels of serum IgE, cold abscesses, pulmonary infections, and eczema. ZNF341 deficiency was described in 2018 in 11 patients clinically diagnosed previously with HIES. Eight of those patients, all offspring of consanguineous couples, are from three families who live in a Muslim village in Israel which has approximately 15,000 residents. Objective Our study aimed to evaluate the prevalence of ZNF341 mutation in the population of the village. Methods Three hundred DNA samples of females were included in the study. The samples belong to females that were referred to the Meir Medical Center for prenatal genetic testing before pregnancy, during 2017-2019: 200 samples were from the village, and 100 samples of Muslim females were from other villages. All samples were tested by Sanger sequencing for the ZNF341 mutation (c.904C>T, NM_001282933.1). Results Heterozygous nonsense mutation in ZNF341 was found in ten samples (5%) of the study group compared to zero in the control group (p<0.01). Conclusion The carrier frequency of the mutation in ZNF341 in the studied village population is 1:20. This high frequency is probably due to founder mutation and consanguineous marriages.
Collapse
Affiliation(s)
- Idit Lachover-Roth
- Allergy and Clinical Immunology Unit, Meir Medical Center, Kfar-Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
- *Correspondence: Idit Lachover-Roth,
| | - Irina Lagovsky
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petach-Tikva, Israel
| | | | - Ronit Confino-Cohen
- Allergy and Clinical Immunology Unit, Meir Medical Center, Kfar-Saba, Israel
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
| | - Eyal Reinstein
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Medical Genetics Institute, Meir Medical Center, Kfar-Saba, Israel
| | - Ben-Zion Garty
- Sackler School of Medicine, Tel Aviv University, Tel-Aviv, Israel
- Felsenstein Medical Research Center, Rabin Medical Center, Petach-Tikva, Israel
- Allergy and Clinical Immunology Unit, Schneider Children’s Medical Center, Petach-Tikva, Israel
| |
Collapse
|
63
|
Kishimoto T, Kang S. IL-6 Revisited: From Rheumatoid Arthritis to CAR T Cell Therapy and COVID-19. Annu Rev Immunol 2022; 40:323-348. [PMID: 35113729 DOI: 10.1146/annurev-immunol-101220-023458] [Citation(s) in RCA: 59] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
The diverse biological activity of interleukin-6 (IL-6) contributes to the maintenance of homeostasis. Emergent infection or tissue injury induces rapid production of IL-6 and activates host defense through augmentation of acute-phase proteins and immune responses. However, excessive IL-6 production and uncontrolled IL-6 receptor signaling are critical to pathogenesis. Over the years, therapeutic agents targeting IL-6 signaling, such as tocilizumab, a humanized anti-IL-6 receptor antibody, have shown remarkable efficacy for rheumatoid arthritis, Castleman disease, and juvenile idiopathic arthritis, and their efficacy in other diseases is continually being reported. Emerging evidence has demonstrated the benefit of tocilizumab for several types of acute inflammatory diseases, including cytokine storms induced by chimeric antigen receptor T-cell therapy and coronavirus disease 2019 (COVID-19). Here, we refocus attention on the biology of IL-6 and summarize the distinct pathological roles of IL-6 signaling in several acute and chronic inflammatory diseases. Expected final online publication date for the Annual Review of Immunology, Volume 40 is April 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Tadamitsu Kishimoto
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan; ,
| | - Sujin Kang
- Laboratory of Immune Regulation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan; ,
| |
Collapse
|
64
|
Hsu AP, Holland SM. Host genetics of innate immune system in infection. Curr Opin Immunol 2022; 74:140-149. [DOI: 10.1016/j.coi.2021.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Revised: 10/21/2021] [Accepted: 11/09/2021] [Indexed: 02/06/2023]
|
65
|
Knight V. Immunodeficiency and Autoantibodies to Cytokines. J Appl Lab Med 2022; 7:151-164. [PMID: 34996092 DOI: 10.1093/jalm/jfab139] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Accepted: 10/11/2021] [Indexed: 11/13/2022]
Abstract
BACKGROUND Anti-cytokine autoantibodies (AAbs) associated with an infectious phenotype are now included along with anti-complement AAbs and somatic pathogenic gene variants as a distinct category termed 'phenocopies of primary immunodeficiencies' in the classification of inborn errors of immunity. Anti-cytokine AAbs target specific cytokine pathways, leading to inordinate susceptibility to specific organisms, generally in the setting of immunocompetence. CONTENT Anti-cytokine AAbs are detected in the majority of healthy individuals and may play a regulatory role in limiting exaggerated responses to cytokines. While it is not well understood why some individuals with anti-cytokine AAbs develop increased susceptibility to organisms of low pathogenicity and others do not, it is likely that genetics and environment play a role. To date, AAbs to interferon gamma (IFNγ), interferon alpha (IFNα), interleukins-17 and 22 (IL-17/IL-22), interleukin-6 and granulocyte macrophage colony stimulating factor (GM-CSF) and their association with increased susceptibility to nontuberculous mycobacteria and other intracellular organisms, viral infections, Candida albicans, Staphylococcus aureus and other pyogenic organisms, and fungal infections respectively, have been described. The clinical phenotype of these patients is very similar to that of individuals with pathogenic gene variants in the specific cytokine pathway that the autoantibody targets, hence the term 'phenocopy.' Recognition of anti-cytokine AAbs as a distinct cause of immunodeficiency or immune dysregulation is important for appropriate management of such patients. SUMMARY Understanding the roles that anti-cytokine AAbs play in health and disease continues to be a fascinating area of research. Evaluating generally immunocompetent individuals who present with chronic, treatment refractory, or unusual infections for anti-cytokine AAbs is critical as it may direct therapy and disease management.
Collapse
Affiliation(s)
- Vijaya Knight
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA.,Children's Hospital, Colorado, Aurora, CO, USA
| |
Collapse
|
66
|
Redmond MT, Scherzer R, Prince BT. Novel Genetic Discoveries in Primary Immunodeficiency Disorders. Clin Rev Allergy Immunol 2022; 63:55-74. [PMID: 35020168 PMCID: PMC8753955 DOI: 10.1007/s12016-021-08881-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/24/2021] [Indexed: 01/12/2023]
Abstract
The field of Immunology is one that has undergone great expansion in recent years. With the advent of new diagnostic modalities including a variety of genetic tests (discussed elsewhere in this journal), the ability to diagnose a patient with a primary immunodeficiency disorder (PIDD) has become a more streamlined process. With increased availability of genetic testing for those with suspected or known PIDD, there has been a significant increase in the number of genes associated with this group of disorders. This is of great importance as a misdiagnosis of these rare diseases can lead to a delay in what can be critical treatment options. At times, those options can include life-saving medications or procedures. Presentation of patients with PIDD can vary greatly based on the specific genetic defect and the part(s) of the immune system that is affected by the variation. PIDD disorders lead to varying levels of increased risk of infection ranging from a mild increase such as with selective IgA deficiency to a profound risk with severe combined immunodeficiency. These diseases can also cause a variety of other clinical findings including autoimmunity and gastrointestinal disease.
Collapse
Affiliation(s)
- Margaret T. Redmond
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH USA
| | - Rebecca Scherzer
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH USA
| | - Benjamin T. Prince
- Division of Allergy and Immunology, Department of Pediatrics, Nationwide Children’s Hospital, The Ohio State University College of Medicine, Columbus, OH USA
| |
Collapse
|
67
|
Mentha arvensis Essential Oil Exerts Anti-Inflammatory in LPS-Stimulated Inflammatory Responses via Inhibition of ERK/NF-κB Signaling Pathway and Anti-Atopic Dermatitis-like Effects in 2,4-Dinitrochlorobezene-Induced BALB/c Mice. Antioxidants (Basel) 2021; 10:antiox10121941. [PMID: 34943044 PMCID: PMC8750489 DOI: 10.3390/antiox10121941] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 11/26/2021] [Accepted: 12/01/2021] [Indexed: 01/12/2023] Open
Abstract
The mechanism of atopic dermatitis (AD) is modulated by the release of cytokines and chemokines through the mitogen-activated protein kinase (MAPK)/nuclear factor-kappa B (NF-κB) signaling pathway. Topical steroids are used to treat AD, but some people need safer anti-inflammatory drugs to avoid side effects. Mentha arvensis has been used as a herbal plant with medicinal properties, but its anti-inflammatory effects have not been elucidated in an AD model. In this study, we investigated the anti-inflammatory effects of M. arvensis essential oil (MAEO) and its underlying molecular mechanism in lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages and HaCaT cells (human epidermal keratinocyte). Additionally, we examined the ameliorating effects of the MAEO in a dinitrochlorobenzene (DNCB)-induced murine model of AD. We found, in both RAW 264.7 cells and HaCaT cells, MAEO inhibited LPS-stimulated inflammatory mediators such as nitric oxide (NO) and prostaglandin E2 and proinflammatory cytokines, including IL-1β and IL-6, due to the suppression of COX-2 and iNOS expression. In LPS-stimulated macrophages, we also observed that MAEO inhibited the phosphorylation of ERK and P65. Furthermore, MAEO treatment attenuated AD symptoms, including the dermatitis score, ear thickness, epidermal thickness and infiltration of mast cells, in a DNCB-induced animal model of AD. Overall, our findings suggest that MAEO exerts anti-inflammatory and anti-atopic dermatitis effects via inhibition of the ERK/NF-κB signaling pathway.
Collapse
|
68
|
Abstract
PURPOSE OF REVIEW Primary immunodeficiency diseases (PIDs), also called inborn errors of immunity (IEI), are genetic disorders classically characterized by an increased susceptibility to infection and/or disruption in the regulation of an immunologic pathway. This review summarizes and highlights the new IEI disorders in the IUIS 2019 report and 2020 interim report and discusses the directions for the future management of PIDs. RECENT FINDINGS Since 2017, the International Union of Immunologic Societies (IUIS) IEI committee has updated the IUIS classification of IEIs with 88 new gene defects and 75 new immune disorders. The increased utilization of genetic testing and advances in the strategic evaluation of genetic variants have identified, not only novel IEI disorders, but additional genetic causes for known IEI disorders. Investigation of potential immune susceptibilities during the ongoing COVID-19 pandemic suggests that defects in Type I interferon signalling may underlie more severe disease. SUMMARY The rapid discovery of new IEIs reflects the growing trend of applying genetic testing modalities as part of medical diagnosis and management.In turn, elucidating the pathophysiology of these novel IEIs have enhanced our understanding of how genetic mutations can modulate the immune system and their consequential effect on human health and disease.
Collapse
Affiliation(s)
- Yesim Demirdag
- Division of Basic and Clinical Immunology, Department of Medicine, University of California, Irvine, Irvine, CA, USA
| | - Ramsay Fuleihan
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics
| | - Jordan S Orange
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics
- Division of Immunogenetics, Department of Pediatrics, Columbia University Irving Medical Center, New York, New York, USA
| | - Joyce E Yu
- Division of Allergy, Immunology, and Rheumatology, Department of Pediatrics
| |
Collapse
|
69
|
STAT3 is critical for skeletal development and bone homeostasis by regulating osteogenesis. Nat Commun 2021; 12:6891. [PMID: 34824272 PMCID: PMC8616950 DOI: 10.1038/s41467-021-27273-w] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 10/19/2021] [Indexed: 11/08/2022] Open
Abstract
Skeletal deformities are typical AD-HIES manifestations, which are mainly caused by heterozygous and loss-of-function mutations in Signal transducer and activator of transcription 3 (STAT3). However, the mechanism is still unclear and the treatment strategy is limited. Herein, we reported that the mice with Stat3 deletion in osteoblasts, but not in osteoclasts, induced AD-HIES-like skeletal defects, including craniofacial malformation, osteoporosis, and spontaneous bone fracture. Mechanistic analyses revealed that STAT3 in cooperation with Msh homeobox 1(MSX1) drove osteoblast differentiation by promoting Distal-less homeobox 5(Dlx5) transcription. Furthermore, pharmacological activation of STAT3 partially rescued skeletal deformities in heterozygous knockout mice, while inhibition of STAT3 aggravated bone loss. Taken together, these data show that STAT3 is critical for modulating skeletal development and maintaining bone homeostasis through STAT3-indcued osteogenesis and suggest it may be a potential target for treatments.
Collapse
|
70
|
Molecular and clinical effects of selective TYK2 inhibition with deucravacitinib in psoriasis. J Allergy Clin Immunol 2021; 149:2010-2020.e8. [PMID: 34767869 DOI: 10.1016/j.jaci.2021.11.001] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 09/20/2021] [Accepted: 11/01/2021] [Indexed: 11/24/2022]
Abstract
BACKGROUND Psoriasis, a chronic inflammatory disease dependent on the interleukin (IL)-23/T helper cell 17 (Th17) pathway, is initiated through plasmacytoid dendritic cell activation and type I interferon induction in the skin. Deucravacitinib, a selective tyrosine kinase 2 (TYK2) inhibitor, blocks IL-23, IL-12, and type I interferon signaling in cellular assays. OBJECTIVE Investigate changes in IL-23/Th17- and type I interferon-pathway biomarkers and gene responses, and measures of selectivity for TYK2 over Janus kinases (JAKs) 1-3, in patients with moderate to severe psoriasis receiving deucravacitinib. METHODS Deucravacitinib was evaluated in a randomized, placebo-controlled, dose-ranging trial. Biopsies from non-lesional (Day 1) and lesional skin (Days 1, 15, and 85) were assessed for changes in IL-23/IL-12 and type I interferon pathway biomarkers by quantitative reverse-transcription polymerase chain reaction, RNA sequencing, and immunohistochemistry. Laboratory markers were measured in blood. Percent change from baseline in Psoriasis Area and Severity Index (PASI) score was assessed. RESULTS IL-23 pathway biomarkers in lesional skin returned toward non-lesional levels dose-dependently with deucravacitinib. Interferon and IL-12 pathway genes were normalized. Markers of keratinocyte dysregulation, keratin-16, and β-defensin genes approached non-lesional levels with effective dosages. Select laboratory parameters impacted by JAK1-3 inhibition were unaffected by deucravacitinib. Greater improvements in PASI scores, correlated with biomarker changes, were seen with the highest dosages of deucravacitinib versus lower dosages or placebo. CONCLUSION Robust clinical efficacy with deucravacitinib treatment was associated with decreases in IL-23/Th17 and interferon pathway biomarkers. The lack of effect seen on biomarkers specific to JAK1-3 inhibition support selectivity of deucravacitinib for TYK2; larger studies are needed to further confirm.
Collapse
|
71
|
New insights into IL-6 family cytokines in metabolism, hepatology and gastroenterology. Nat Rev Gastroenterol Hepatol 2021; 18:787-803. [PMID: 34211157 DOI: 10.1038/s41575-021-00473-x] [Citation(s) in RCA: 66] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/20/2021] [Indexed: 02/06/2023]
Abstract
IL-6 family cytokines are defined by the common use of the signal-transducing receptor chain glycoprotein 130 (gp130). Increasing evidence indicates that these cytokines are essential in the regulation of metabolic homeostasis as well as in the pathophysiology of multiple gastrointestinal and liver disorders, thus making them attractive therapeutic targets. Over the past few years, therapies modulating gp130 signalling have grown exponentially in several clinical settings including obesity, cancer and inflammatory bowel disease. A newly engineered gp130 cytokine, IC7Fc, has shown promising preclinical results for the treatment of type 2 diabetes, obesity and liver steatosis. Moreover, drugs that modulate gp130 signalling have shown promise in refractory inflammatory bowel disease in clinical trials. A deeper understanding of the main roles of the IL-6 family of cytokines during homeostatic and pathological conditions, their signalling pathways, sources of production and target cells will be crucial to the development of improved treatments. Here, we review the current state of the role of these cytokines in hepatology and gastroenterology and discuss the progress achieved in translating therapeutics targeting gp130 signalling into clinical practice.
Collapse
|
72
|
Martínez-Pérez C, Kay C, Meehan J, Gray M, Dixon JM, Turnbull AK. The IL6-like Cytokine Family: Role and Biomarker Potential in Breast Cancer. J Pers Med 2021; 11:1073. [PMID: 34834425 PMCID: PMC8624266 DOI: 10.3390/jpm11111073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Revised: 10/20/2021] [Accepted: 10/21/2021] [Indexed: 02/07/2023] Open
Abstract
IL6-like cytokines are a family of regulators with a complex, pleiotropic role in both the healthy organism, where they regulate immunity and homeostasis, and in different diseases, including cancer. Here we summarise how these cytokines exert their effect through the shared signal transducer IL6ST (gp130) and we review the extensive evidence on the role that different members of this family play in breast cancer. Additionally, we discuss how the different cytokines, their related receptors and downstream effectors, as well as specific polymorphisms in these molecules, can serve as predictive or prognostic biomarkers with the potential for clinical application in breast cancer. Lastly, we also discuss how our increasing understanding of this complex signalling axis presents promising opportunities for the development or repurposing of therapeutic strategies against cancer and, specifically, breast neoplasms.
Collapse
Affiliation(s)
- Carlos Martínez-Pérez
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Charlene Kay
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - James Meehan
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - Mark Gray
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| | - J. Michael Dixon
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
| | - Arran K. Turnbull
- Breast Cancer Now Edinburgh Research Team, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH4 2XU, UK; (C.K.); (J.M.D.); (A.K.T.)
- Translational Oncology Research Group, MRC Institute of Genetics and Cancer, Western General Hospital, University of Edinburgh, Edinburgh EH8 9YL, UK; (J.M.); (M.G.)
| |
Collapse
|
73
|
Seth N, Tuano KS, Chinen J. Inborn errors of immunity: Recent progress. J Allergy Clin Immunol 2021; 148:1442-1450. [PMID: 34688776 DOI: 10.1016/j.jaci.2021.10.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 10/16/2021] [Accepted: 10/18/2021] [Indexed: 10/20/2022]
Abstract
Recent advances in the field of inborn errors of immunity (IEIs) have been wide in scope, including progress in mechanisms of disease, diagnosis, and management. New gene defects affecting the immune response continue to be reported, as many as 26 in the year 2020. It was noted that the presentation of IEIs might not include recurrent infections in 9% of cases, and that current diagnostic methods can identify molecular causes in 92% of patients with severe combined immunodeficiency. Progress in immunopathogenesis explained mechanisms leading to symptoms of autosomal-recessive hyper-IgE syndrome. There was an emphasis on research in primary antibody deficiencies. The benefit of antibiotic prophylaxis to reduce the frequency of infections was demonstrated in these patients. The regimen of rituximab and azathioprine or mycophenolate was proven effective for chronic granulocytic interstitial pneumonia. The efficacy and adverse events of hematopoietic stem cell transplant in different IEI conditions were reported, as well as different strategies to improve outcomes, supporting its use in immunodeficiency and immunodysregulatory syndromes. The recent pandemic of coronavirus disease 2019 affected patients with IEIs, in particular those with deficiency in the interferon-mediated activation of the immune response. Initial data suggest that coronavirus disease 2019 vaccines might elicit anti-coronavirus disease 2019-neutralizing antibody responses in some patients with IEI conditions.
Collapse
Affiliation(s)
- Neha Seth
- Division of Immunology, Allergy and Retrovirology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, The Woodlands, Tex
| | - Karen S Tuano
- Division of Immunology, Allergy and Retrovirology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, The Woodlands, Tex
| | - Javier Chinen
- Division of Immunology, Allergy and Retrovirology, Department of Pediatrics, Baylor College of Medicine, Texas Children's Hospital, The Woodlands, Tex.
| |
Collapse
|
74
|
IL-6R/Signal Transducer and Activator of Transcription 3 Signaling in Keratinocytes rather than in T Cells Induces Psoriasis-Like Dermatitis in Mice. J Invest Dermatol 2021; 142:1126-1135.e4. [PMID: 34626614 PMCID: PMC8957489 DOI: 10.1016/j.jid.2021.09.012] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 09/10/2021] [Accepted: 09/22/2021] [Indexed: 12/25/2022]
Abstract
Signal transducer and activator of transcription 3 (STAT3) is important for psoriasis pathogenesis because STAT3 signaling downstream of IL-6, IL-21, IL-22, and IL-23 contributes to T helper type 17 cell development and because transgenic mice with keratinocyte (KC) STAT3 expression (K14-Stat3C mice) develop psoriasis-like dermatitis. In this study, the relative contribution of STAT3 signaling in KCs versus in T cells was evaluated in the imiquimod model of psoriasis-like dermatitis. Mice with STAT3-inducible deletion in KCs (K5-Stat3-/- mice) had decreased psoriasis-like dermatitis and epidermal STAT3 phosphorylation compared with wild-type mice, whereas mice with constitutive deletion of STAT3 in all T cells were similar to wild-type mice. Interestingly, mice with KC-inducible deletion of IL-6Rα had similar findings to those of K5-Stat3-/- mice, identifying IL-6/IL-6R as a predominant upstream signal for KC STAT3-induced psoriasis-like dermatitis. Moreover, psoriasis-like dermatitis inversely associated with type 1 immune gene products, especially CXCL10, whereas CXCL10 limited psoriasis-like dermatitis, suggesting that KC STAT3 signaling promoted psoriasis-like dermatitis by restricting downstream CXCL10 expression. Finally, treatment of mice with the pan-Jak inhibitor, tofacitinib, reduced psoriasis-like dermatitis and epidermal STAT3 phosphorylation. Taken together, STAT3 signaling in KCs rather than in T cells was a more important determinant for psoriasis-like dermatitis in a mechanism that involved upstream KC IL-6R signaling and downstream inhibition of type 1 immunity‒associated CXCL10 responses.
Collapse
|
75
|
Abstract
Clinically and pathologically, the patients with hyper-IgE syndrome present similar skin manifestations to common atopic dermatitis. The original hyper-IgE syndrome is characterized by diminished inflammatory response, in combination with Staphylococcus aureus skin abscess and pneumonia followed by pneumatocele formation. These immunological manifestations are frequently associated with skeletal and connective tissue abnormalities. We previously identified that major causal variants of the hyper-IgE syndrome are dominant negative variants in the STAT3. In addition to the identification of new causative variants for the disorders similar to the original hyper-IgE syndrome, causative variants for new types of hyper-IgE syndrome centered only on atopy, high serum IgE levels, and susceptibility to infection, but not associated with diminished inflammatory response, pneumatocele formation, and connective tissue manifestations, have been identified. Recent discovery identified a novel zinc finger protein that regulates STAT3 transcription. Investigation of IL6ST variants disclosed that IL6ST/IL6R cytokine receptor plays a crucial role for the signal transduction upstream of STAT3 in the pathogenesis of the original hyper-IgE syndrome. Even if the same IL6ST variants are used for the signal transduction of IL-6 family cytokines, the signaling defect is more severe in IL-6/IL-11 and milder in LIF. The fact that the non-immune manifestations of the gain-of-function mutations of TGFBR1 and TGFBR2 are similar to the those of dominant negative mutations of STAT3 provide a clue to elucidate molecular mechanisms of non-immune manifestations of hyper-IgE syndrome. Research on this hereditary atopic syndrome is being actively conducted to elucidate the molecular mechanisms and to develop new therapeutic approaches.
Collapse
|
76
|
Chen YH, Spencer S, Laurence A, Thaventhiran JE, Uhlig HH. Inborn errors of IL-6 family cytokine responses. Curr Opin Immunol 2021; 72:135-145. [PMID: 34044328 PMCID: PMC8591178 DOI: 10.1016/j.coi.2021.04.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 04/15/2021] [Accepted: 04/19/2021] [Indexed: 01/25/2023]
Abstract
The IL-6 family of cytokines mediates functions in host protective immunity, development of multiple organs, tissue regeneration and metabolism. Inborn errors in cytokines or cytokine receptor units highlight specific roles for IL-6, IL-11, LIF, OSM, and CLC signaling whereas incomplete loss-of-function variants in the common receptor chain GP130 encoded by IL6ST or the transcription factor STAT3, as well as genes that affect either GP130 glycosylation (PGM3) or STAT3 transcriptional control (ZNF341) lead to complex phenotypes including features of hyper-IgE syndrome. Gain-of-function variants in the GP130-STAT3 signaling pathway cause immune dysregulation disorders. Insights into IL-6 family cytokine signaling inform on therapeutic application in immune-mediated disorders and potential side effects such as infection susceptibility.
Collapse
Affiliation(s)
- Yin-Huai Chen
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK
| | - Sarah Spencer
- MRC Toxicology Unit, Gleeson Building, Tennis Court Road, Cambridge, UK
| | - Arian Laurence
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK; Department of Haematology, University College Hospital, UCLH Hospitals NHS Trust, UK
| | | | - Holm H Uhlig
- Translational Gastroenterology Unit, University of Oxford, Oxford, UK; Biomedical Research Center, University of Oxford, Oxford, UK; Department of Pediatrics, University of Oxford, Oxford, UK.
| |
Collapse
|
77
|
Shields AM, Faustini SE, Perez-Toledo M, Jossi S, Allen JD, Al-Taei S, Backhouse C, Dunbar LA, Ebanks D, Emmanuel B, Faniyi AA, Garvey M, Grinbergs A, McGinnell G, O'Neill J, Watanabe Y, Crispin M, Wraith DC, Cunningham AF, Drayson MT, Richter AG. Serological responses to SARS-CoV-2 following non-hospitalised infection: clinical and ethnodemographic features associated with the magnitude of the antibody response. BMJ Open Respir Res 2021; 8:e000872. [PMID: 34561239 PMCID: PMC8474079 DOI: 10.1136/bmjresp-2020-000872] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2020] [Accepted: 08/30/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVE To determine clinical and ethnodemographic correlates of serological responses against the SARS-CoV-2 spike glycoprotein following mild-to-moderate COVID-19. DESIGN A retrospective cohort study of healthcare workers who had self-isolated due to COVID-19. SETTING University Hospitals Birmingham NHS Foundation Trust, UK (UHBFT). PARTICIPANTS 956 healthcare workers were recruited by open invitation via UHBFT trust email and social media between 27 April 2020 and the 8 June 2020. INTERVENTION Participants volunteered a venous blood sample that was tested for the presence of anti-SARS-CoV-2 spike glycoprotein antibodies. Results were interpreted in the context of the symptoms of their original illness and ethnodemographic variables. RESULTS Using an assay that simultaneously measures the combined IgG, IgA and IgM response against the spike glycoprotein (IgGAM), the overall seroprevalence within this cohort was 46.2% (n=442/956). The seroprevalence of immunoglobulin isotypes was 36.3%, 18.7% and 8.1% for IgG, IgA and IgM, respectively. IgGAM identified serological responses in 40.6% (n=52/128) of symptomatic individuals who reported a negative SARS-CoV-2 PCR test. Increasing age, non-white ethnicity and obesity were independently associated with greater IgG antibody response against the spike glycoprotein. Self-reported fever and fatigue were associated with greater IgG and IgA responses against the spike glycoprotein. The combination of fever and/or cough and/or anosmia had a positive predictive value of 92.3% for seropositivity in self-isolating individuals a time when Wuhan strain SARS-CoV-2 was predominant. CONCLUSIONS AND RELEVANCE Assays employing combined antibody detection demonstrate enhanced seroepidemiological sensitivity and can detect prior viral exposure even when PCR swabs have been negative. We demonstrate an association between known ethnodemographic risk factors associated with mortality from COVID-19 and the magnitude of serological responses in mild-to-moderate disease.
Collapse
Affiliation(s)
- Adrian M Shields
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Sian E Faustini
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Marisol Perez-Toledo
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Sian Jossi
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Joel D Allen
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - Saly Al-Taei
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Claire Backhouse
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Lynsey A Dunbar
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Daniel Ebanks
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Beena Emmanuel
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Aduragbemi A Faniyi
- Birmingham Acute Care Research Group, Institute of Inflammation and Ageing, University of Birmingham, Birmingham, UK
| | - Mark Garvey
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
- Institute of Microbiology and Infection, University of Birmingham, Birmingham, UK
| | - Annabel Grinbergs
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Golaleh McGinnell
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Joanne O'Neill
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Yasunori Watanabe
- School of Biological Sciences, University of Southampton, Southampton, UK
- Oxford Glycobiology Institute, Department of Biochemistry, University of Oxford, Oxford, UK
| | - Max Crispin
- School of Biological Sciences, University of Southampton, Southampton, UK
| | - David C Wraith
- Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Adam F Cunningham
- MRC Centre for Immune Regulation, University of Birmingham, Birmingham, UK
| | - Mark T Drayson
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| | - Alex G Richter
- Department of Clinical Immunology Service, University of Birmingham College of Medical and Dental Sciences, Birmingham, UK
| |
Collapse
|
78
|
Philippot Q, Casanova JL, Puel A. Candidiasis in patients with APS-1: low IL-17, high IFN-γ, or both? Curr Opin Immunol 2021; 72:318-323. [PMID: 34455138 DOI: 10.1016/j.coi.2021.08.001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Revised: 08/03/2021] [Accepted: 08/05/2021] [Indexed: 12/18/2022]
Abstract
Chronic mucocutaneous candidiasis (CMC) is one of the earliest and most frequent clinical manifestations of autosomal recessive autoimmune polyendocrine syndrome type 1 (APS-1), a monogenic inborn error of immunity caused by deleterious variants of the autoimmune regulator (AIRE) gene. APS-1 patients suffer from various autoimmune diseases, due to the defective thymic deletion of autoreactive T cells, and the development of a large range of autoantibodies (auto-Abs) against various tissue antigens, and some cytokines. The mechanisms underlying CMC remained elusive for many years, until the description in 2010 of high serum titers of neutralizing auto-Abs against IL-17A, IL-17F, and/or IL-22, which are present in almost all APS-1 patients. Excessively high mucosal concentrations of IFN-γ were recently proposed as an alternative mechanism for CMC in APS-1.
Collapse
Affiliation(s)
- Quentin Philippot
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States; Howard Hughes Medical Institute, New York, NY, United States
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, INSERM U1163, Necker Hospital for Sick Children, Paris, France; University of Paris, Imagine Institute, Paris, France; St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY, United States.
| |
Collapse
|
79
|
Jenkins RH, Hughes STO, Figueras AC, Jones SA. Unravelling the broader complexity of IL-6 involvement in health and disease. Cytokine 2021; 148:155684. [PMID: 34411990 DOI: 10.1016/j.cyto.2021.155684] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 07/20/2021] [Accepted: 08/04/2021] [Indexed: 02/07/2023]
Abstract
The classification of interleukin-6 (IL-6) as a pro-inflammatory cytokine undervalues the biological impact of this cytokine in health and disease. With broad activities affecting the immune system, tissue homeostasis and metabolic processes, IL-6 displays complex biology. The significance of these involvements has become increasingly important in clinical settings where IL-6 is identified as a prominent target for therapy. Here, clinical experience with IL-6 antagonists emphasises the need to understand the context-dependent properties of IL-6 within an inflammatory environment and the anticipated or unexpected consequences of IL-6 blockade. In this review, we will describe the immunobiology of IL-6 and explore the gamut of IL-6 bioactivity affecting the clinical response to biological drugs targeting this cytokine pathway.
Collapse
Affiliation(s)
- Robert H Jenkins
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Stuart T O Hughes
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Ana Cardus Figueras
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK
| | - Simon A Jones
- Division of Infection & Immunity, The School of Medicine, Cardiff University, Cardiff, Wales, UK; Systems Immunity Research Institute, The School of Medicine, Cardiff University, Cardiff, Wales, UK.
| |
Collapse
|
80
|
Wey EQ, Bristow C, Nandani A, O'Farrell B, Pang J, Lanzman M, Yang S, Ho S, Mack D, Spiro M, Balakrishnan I, Bhagani S, Pollara G. Preserved C-reactive protein responses to blood stream infections following tocilizumab treatment for COVID-19. J Infect 2021; 83:607-635. [PMID: 34400218 PMCID: PMC8363426 DOI: 10.1016/j.jinf.2021.08.017] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 08/07/2021] [Indexed: 12/22/2022]
Affiliation(s)
- Emmanuel Q Wey
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom; Centre for Clinical Microbiology, Division of Infection & Immunity, UCL, London, United Kingdom
| | - Clare Bristow
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom
| | - Aarti Nandani
- Department of Pharmacy, Royal Free London NHS Trust, London, United Kingdom
| | - Bryan O'Farrell
- Department of Pharmacy, Royal Free London NHS Trust, London, United Kingdom
| | - Jay Pang
- Department of Pharmacy, Royal Free London NHS Trust, London, United Kingdom
| | - Marisa Lanzman
- Department of Pharmacy, Royal Free London NHS Trust, London, United Kingdom
| | - Shuang Yang
- Clinical Practice Group, Analysis Division, Transplant and Specialist Services & Women and Children, Royal Free London NHS Trust, London, United Kingdom
| | - Soo Ho
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom
| | - Damien Mack
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom
| | - Michael Spiro
- Division of Surgery and Interventional Science, University College London, United Kingdom
| | - Indran Balakrishnan
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom
| | - Sanjay Bhagani
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom
| | - Gabriele Pollara
- Department of Infection, Royal Free London NHS Trust, London, United Kingdom; Division of Infection & Immunity, University College London, United Kingdom.
| |
Collapse
|
81
|
Yang R, Weisshaar M, Mele F, Benhsaien I, Dorgham K, Han J, Croft CA, Notarbartolo S, Rosain J, Bastard P, Puel A, Fleckenstein B, Glimcher LH, Di Santo JP, Ma CS, Gorochov G, Bousfiha A, Abel L, Tangye SG, Casanova JL, Bustamante J, Sallusto F. High Th2 cytokine levels and upper airway inflammation in human inherited T-bet deficiency. J Exp Med 2021; 218:e20202726. [PMID: 34160550 PMCID: PMC8225679 DOI: 10.1084/jem.20202726] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 04/16/2021] [Accepted: 05/27/2021] [Indexed: 12/20/2022] Open
Abstract
We have described a child suffering from Mendelian susceptibility to mycobacterial disease (MSMD) due to autosomal recessive, complete T-bet deficiency, which impairs IFN-γ production by innate and innate-like adaptive, but not mycobacterial-reactive purely adaptive, lymphocytes. Here, we explore the persistent upper airway inflammation (UAI) and blood eosinophilia of this patient. Unlike wild-type (WT) T-bet, the mutant form of T-bet from this patient did not inhibit the production of Th2 cytokines, including IL-4, IL-5, IL-9, and IL-13, when overexpressed in T helper 2 (Th2) cells. Moreover, Herpesvirus saimiri-immortalized T cells from the patient produced abnormally large amounts of Th2 cytokines, and the patient had markedly high plasma IL-5 and IL-13 concentrations. Finally, the patient's CD4+ αβ T cells produced most of the Th2 cytokines in response to chronic stimulation, regardless of their antigen specificities, a phenotype reversed by the expression of WT T-bet. T-bet deficiency thus underlies the excessive production of Th2 cytokines, particularly IL-5 and IL-13, by CD4+ αβ T cells, causing blood eosinophilia and UAI. The MSMD of this patient results from defective IFN-γ production by innate and innate-like adaptive lymphocytes, whereas the UAI and eosinophilia result from excessive Th2 cytokine production by adaptive CD4+ αβ T lymphocytes.
Collapse
Affiliation(s)
- Rui Yang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Marc Weisshaar
- Institute of Microbiology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
| | - Federico Mele
- Center of Medical Immunology, Institute for Research in Biomedicine, Faculty of Biomedical Sciences, University of Italian Switzerland, Bellinzona, Switzerland
| | - Ibtihal Benhsaien
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, Casablanca, Morocco
- Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, Centre Hospitalo-Universitaire Averroes, Casablanca, Morocco
| | - Karim Dorgham
- Sorbonne University, Institut national de la santé et de la recherche médicale, Center for Immunology and Microbial Infections-Paris, Paris, France
| | - Jing Han
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
| | - Carys A. Croft
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Institut national de la santé et de la recherche médicale U1223, Paris, France
- University of Paris, Paris, France
| | - Samuele Notarbartolo
- Center of Medical Immunology, Institute for Research in Biomedicine, Faculty of Biomedical Sciences, University of Italian Switzerland, Bellinzona, Switzerland
| | - Jérémie Rosain
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Paul Bastard
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Bernhard Fleckenstein
- Institute for Clinical and Molecular Virology, University Erlangen-Nuremberg, Erlangen, Germany
| | - Laurie H. Glimcher
- Department of Cancer Immunology and Virology, Dana-Farber Cancer Institute, Boston, MA
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
- Department of Immunology, Harvard Medical School, Boston, MA
| | - James P. Di Santo
- Innate Immunity Unit, Institut Pasteur, Paris, France
- Institut national de la santé et de la recherche médicale U1223, Paris, France
| | - Cindy S. Ma
- Garvan Institute of Medical Research, Darlinghurst, Australia
- St. Vincent’s Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Darlinghurst, Australia
| | - Guy Gorochov
- Sorbonne University, Institut national de la santé et de la recherche médicale, Center for Immunology and Microbial Infections-Paris, Paris, France
- Assistance Publique–Hôpitaux de Paris, Department of Immunology, Paris, France
| | - Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation, and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, Casablanca, Morocco
- Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, Centre Hospitalo-Universitaire Averroes, Casablanca, Morocco
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
| | - Stuart G. Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia
- St. Vincent’s Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Darlinghurst, Australia
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Howard Hughes Medical Institute, New York, NY
| | - Jacinta Bustamante
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, Rockefeller University, New York, NY
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut national de la santé et de la recherche médicale Unité Mixte de Recherches 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, Assistance Publique–Hôpitaux de Paris, Paris, France
| | - Federica Sallusto
- Institute of Microbiology, Eidgenössische Technische Hochschule Zurich, Zurich, Switzerland
- Center of Medical Immunology, Institute for Research in Biomedicine, Faculty of Biomedical Sciences, University of Italian Switzerland, Bellinzona, Switzerland
| |
Collapse
|
82
|
Tangye SG, Ma CS. Molecular regulation and dysregulation of T follicular helper cells - learning from inborn errors of immunity. Curr Opin Immunol 2021; 72:249-261. [PMID: 34284230 DOI: 10.1016/j.coi.2021.06.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 06/07/2021] [Accepted: 06/15/2021] [Indexed: 12/21/2022]
Abstract
The production of high-affinity antibodies is a key feature of the vertebrate immune system. Antibodies neutralize and clear pathogens, thereby protecting against infectious diseases. However, dysregulated production of antibodies can cause immune pathologies, such as autoimmunity and immune deficiency. Long-lived humoral immunity depends on B-cell help provided by T follicular helper (Tfh) cells, which support the differentiation of antigen (Ag)-specific B cells into memory and plasma cells. Tfh cells are generated from naïve CD4+ T cells following the receipt of inputs from various cell surface receptors, and can undergo further differentiation into subsets with specialised effector functions to induce and maintain serological memory. While genetically modified mice have provided great understanding of the requirements for generating Tfh cells, it is critical that requirements for human Tfh cell generation and function are also established. Key insights into the molecular requirements for human Tfh cells have been elucidated from the systematic analysis of humans with monogenic germline variants that cause inborn errors of immunity characterised by impaired humoral immunity following infection or vaccination or immune dysregulation and autoimmunity. In this review we will discuss how studying rare 'experiments of nature' has enabled discovery of non-redundant molecules and pathways necessary for Tfh cell generation, differentiation, regulation and function in humans, and how these findings inform us about basic and clinical immunology.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine & Health, UNSW Sydney, Darlinghurst, NSW 2010 Australia; CIRCA (Clinical Immunogenomics Consortium of Australasia), Australia.
| | - Cindy S Ma
- Garvan Institute of Medical Research, Darlinghurst NSW 2010, Australia; St Vincent's Clinical School, Faculty of Medicine & Health, UNSW Sydney, Darlinghurst, NSW 2010 Australia; CIRCA (Clinical Immunogenomics Consortium of Australasia), Australia
| |
Collapse
|
83
|
Tsilifis C, Freeman AF, Gennery AR. STAT3 Hyper-IgE Syndrome-an Update and Unanswered Questions. J Clin Immunol 2021; 41:864-880. [PMID: 33932191 PMCID: PMC8249299 DOI: 10.1007/s10875-021-01051-1] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 04/20/2021] [Indexed: 12/16/2022]
Abstract
The hyper-IgE syndromes (HIES) are a heterogeneous group of inborn errors of immunity sharing manifestations including increased infection susceptibility, eczema, and raised serum IgE. Since the prototypical HIES description 55 years ago, areas of significant progress have included description of key disease-causing genes and differentiation into clinically distinct entities. The first two patients reported had what is now understood to be HIES from dominant-negative mutations in signal transduction and activator of transcription 3 (STAT3-HIES), conferring a broad immune defect across both innate and acquired arms, as well as defects in skeletal, connective tissue, and vascular function, causing a clinical phenotype including eczema, staphylococcal and fungal skin and pulmonary infection, scoliosis and minimal trauma fractures, and vascular tortuosity and aneurysm. Due to the constitutionally expressed nature of STAT3, initial reports at treatment with allogeneic stem cell transplantation were not positive and treatment has hinged on aggressive antimicrobial prophylaxis and treatment to prevent the development of end-organ disease such as pneumatocele. Research into the pathophysiology of STAT3-HIES has driven understanding of the interface of several signaling pathways, including the JAK-STAT pathways, interleukins 6 and 17, and the role of Th17 lymphocytes, and has been expanded by identification of phenocopies such as mutations in IL6ST and ZNF341. In this review we summarize the published literature on STAT3-HIES, present the diverse clinical manifestations of this syndrome with current management strategies, and update on the uncertain role of stem cell transplantation for this disease. We outline key unanswered questions for further study.
Collapse
Affiliation(s)
- Christo Tsilifis
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Andrew R Gennery
- Paediatric Haematopoietic Stem Cell Transplant Unit, Great North Children's Hospital (GNCH), Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK.
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
| |
Collapse
|
84
|
Ma R, Lu T, Li Z, Teng KY, Mansour AG, Yu M, Tian L, Xu B, Ma S, Zhang J, Barr T, Peng Y, Caligiuri MA, Yu J. An Oncolytic Virus Expressing IL15/IL15Rα Combined with Off-the-Shelf EGFR-CAR NK Cells Targets Glioblastoma. Cancer Res 2021; 81:3635-3648. [PMID: 34006525 PMCID: PMC8562586 DOI: 10.1158/0008-5472.can-21-0035] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 03/31/2021] [Accepted: 05/06/2021] [Indexed: 02/05/2023]
Abstract
IL15 is a pleiotropic cytokine with multiple roles that improve immune responses to tumor cells. Oncolytic viruses (OV) specifically lyse tumors and activate immune responses. Systemic administration of IL15 or its complex with the IL15Rα and chimeric antigen receptor (CAR) natural killer (NK) cells are currently being tested in the clinic. Here, we generated a herpes simplex 1-based OV-expressing human IL15/IL15Rα sushi domain fusion protein (named OV-IL15C), as well as off-the-shelf EGFR-CAR NK cells, and studied their monotherapy and combination efficacy in vitro and in multiple glioblastoma (GBM) mouse models. In vitro, soluble IL15/IL15Rα complex was secreted from OV-IL15C-infected GBM cells, which promoted GBM cytotoxicity and improved survival of NK and CD8+ T cells. Frozen, readily available off-the-shelf EGFR-CAR NK cells showed enhanced killing of tumor cells compared with empty vector-transduced NK cells. In vivo, OV-IL15C significantly inhibited tumor growth and prolonged survival of GBM-bearing mice in the presence of CD8+ T cells compared with parental OV. OV-IL15C plus EGFR-CAR NK cells synergistically suppressed tumor growth and significantly improved survival compared with either monotherapy, correlating with increased intracranial infiltration and activation of NK and CD8+ T cells and elevated persistence of CAR NK cells in an immunocompetent model. Collectively, OV-IL15C and off-the-shelf EGFR-CAR NK cells represent promising therapeutic strategies for GBM treatment to improve the clinical management of this devastating disease. SIGNIFICANCE: The combination of an oncolytic virus expressing the IL15/IL15Rα complex and frozen, ready-to-use EGFR-CAR NK cells elicits strong antitumor responses in glioblastoma.
Collapse
Affiliation(s)
- Rui Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, P.R. China
| | - Ting Lu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Zhenlong Li
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Kun-Yu Teng
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Anthony G Mansour
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Melissa Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Lei Tian
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Bo Xu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Shoubao Ma
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Jianying Zhang
- Department of Computational and Quantitative Medicine, City of Hope National Medical Center, Los Angeles, California
| | - Tasha Barr
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California
| | - Yong Peng
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Chengdu, P.R. China.
| | - Michael A Caligiuri
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California.
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Centre, Los Angeles, California
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, California
| | - Jianhua Yu
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, California.
- Department of Immuno-Oncology, Beckman Research Institute, City of Hope Comprehensive Cancer Centre, Los Angeles, California
- Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, California
| |
Collapse
|
85
|
Hackney A, James S, Karanam S. Recurrent severe respiratory infections with low CRP secondary to autoimmune IL-6 deficiency. BMJ Case Rep 2021; 14:14/6/e243334. [PMID: 34155030 DOI: 10.1136/bcr-2021-243334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
C reactive protein (CRP) is an inflammatory protein that is often used in conjunction with the clinical presentation of a patient to help with quantifying infection severity and measuring treatment response. We report the case of a patient who presented with severe pneumonias but had a suboptimal CRP response, later diagnosed as having an underlying immunodeficiency. This case exemplifies the need to consider immunodeficiency as an underlying pathophysiological cause in patients presenting with complicated and severe infections.
Collapse
Affiliation(s)
- Alexander Hackney
- Department of Immunology, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | - Sunil James
- Department of Immunology, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| | - Surendra Karanam
- Department of Immunology, Sandwell and West Birmingham Hospitals NHS Trust, Birmingham, UK
| |
Collapse
|
86
|
Asano T, Khourieh J, Zhang P, Rapaport F, Spaan AN, Li J, Lei WT, Pelham SJ, Hum D, Chrabieh M, Han JE, Guérin A, Mackie J, Gupta S, Saikia B, Baghdadi JEI, Fadil I, Bousfiha A, Habib T, Marr N, Ganeshanandan L, Peake J, Droney L, Williams A, Celmeli F, Hatipoglu N, Ozcelik T, Picard C, Abel L, Tangye SG, Boisson-Dupuis S, Zhang Q, Puel A, Béziat V, Casanova JL, Boisson B. Human STAT3 variants underlie autosomal dominant hyper-IgE syndrome by negative dominance. J Exp Med 2021; 218:212397. [PMID: 34137790 PMCID: PMC8217968 DOI: 10.1084/jem.20202592] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Revised: 03/30/2021] [Accepted: 05/18/2021] [Indexed: 12/18/2022] Open
Abstract
Most patients with autosomal dominant hyper-IgE syndrome (AD-HIES) carry rare heterozygous STAT3 variants. Only six of the 135 in-frame variants reported have been experimentally shown to be dominant negative (DN), and it has been recently suggested that eight out-of-frame variants operate by haploinsufficiency. We experimentally tested these 143 variants, 7 novel out-of-frame variants found in HIES patients, and other STAT3 variants from the general population. Strikingly, all 15 out-of-frame variants were DN via their encoded (1) truncated proteins, (2) neoproteins generated from a translation reinitiation codon, and (3) isoforms from alternative transcripts or a combination thereof. Moreover, 128 of the 135 in-frame variants (95%) were also DN. The patients carrying the seven non-DN STAT3 in-frame variants have not been studied for other genetic etiologies. Finally, none of the variants from the general population tested, including an out-of-frame variant, were DN. Overall, our findings show that heterozygous STAT3 variants, whether in or out of frame, underlie AD-HIES through negative dominance rather than haploinsufficiency.
Collapse
Affiliation(s)
- Takaki Asano
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Joëlle Khourieh
- Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Peng Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Franck Rapaport
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - András N Spaan
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Juan Li
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Wei-Te Lei
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Simon J Pelham
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - David Hum
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Maya Chrabieh
- Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Ji Eun Han
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY
| | - Antoine Guérin
- Garvan Institute of Medical Research, Darlinghurst, Australia.,St. Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Joseph Mackie
- Garvan Institute of Medical Research, Darlinghurst, Australia.,St. Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Sudhir Gupta
- Division of Basic and Clinical Immunology, Department of Medicine, School of Medicine, University of California, Irvine, Irvine, CA
| | - Biman Saikia
- Department of Immunopathology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | | | - Ilham Fadil
- Laboratory of Clinical Immunology, Inflammation and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, Casablanca, Morocco.,Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, Averroes University Hospital Center, Casablanca, Morocco
| | - Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation and Allergy, Faculty of Medicine and Pharmacy of Casablanca, King Hassan II University, Casablanca, Morocco.,Clinical Immunology Unit, Department of Pediatric Infectious Diseases, Children's Hospital, Averroes University Hospital Center, Casablanca, Morocco
| | - Tanwir Habib
- Research Branch, Sidra Medicine, Qatar Foundation, Doha, Qatar
| | - Nico Marr
- Research Branch, Sidra Medicine, Qatar Foundation, Doha, Qatar.,College of Health & Life Sciences, Hamad Bin Khalifa University, Qatar Foundation, Doha, Qatar
| | - Luckshman Ganeshanandan
- Department of Clinical Immunology, PathWest Laboratory Medicine Western Australia, Fiona Stanley Hospital, Perth, Australia
| | - Jane Peake
- Queensland Children's Hospital, South Brisbane, Australia
| | - Luke Droney
- Department of Clinical Immunology, Princess Alexandra Hospital, Brisbane, Australia
| | - Andrew Williams
- Immunology Laboratory, Children's Hospital Westmead, Westmead, Australia
| | - Fatih Celmeli
- Department of Allergy and Immunology, University of Medical Science Antalya Education and Research Hospital, Antalya, Turkey
| | - Nevin Hatipoglu
- Bakirkoy Dr Sadi Konuk Education and Training Hospital, Istanbul, Turkey
| | - Tayfun Ozcelik
- Department of Molecular Biology and Genetics, Bilkent University, Ankara, Turkey
| | - Capucine Picard
- Université de Paris, Paris, France.,Study Center for Primary Immunodeficiencies, Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France.,Laboratory of Lymphocyte Activation and Susceptibility to EBV Infection, Institut National de la Santé et de la Recherche Médicale UMR 1163, Imagine Institute, Paris, France.,Pediatric Immunology-Hematology Unit, Assistance Publique-Hôpitaux de Paris, Necker Hospital for Sick Children, Paris, France
| | - Laurent Abel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Australia.,St. Vincent's Clinical School, Faculty of Medicine and Health, University of New South Wales, Sydney, Australia
| | - Stéphanie Boisson-Dupuis
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Qian Zhang
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Anne Puel
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Vivien Béziat
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| | - Jean-Laurent Casanova
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France.,Howard Hughes Medical Institute, New York, NY
| | - Bertrand Boisson
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, NY.,Paris University, Imagine Institute, Paris, France.,Laboratory of Human Genetics of Infectious Disease, Necker Branch, Institut National de la Santé et de la Recherche Médicale U1163, Paris, France
| |
Collapse
|
87
|
Zhang Y, Xue Z, Hu S, Bai H, Wang J, Wang N. Chrysin Inhibits Pseudo-allergic Reaction by Suppressing Mitochondrial STAT3 Activation via MAS-Related GPR Family Member X2. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2021; 69:6569-6577. [PMID: 34100606 DOI: 10.1021/acs.jafc.1c02565] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Chrysin, one of the most pharmacologically active natural flavonoids, has been extracted from various plants. Mast cells are an important part of innate immunity-mediating anaphylaxis. Pseudo-allergic reactions are currently believed to be associated with the MAS-related GPR family member X2 (MrgX2). In this study, the anti-pseudo allergy effect of chrysin and its underlying mechanisms were studied in vitro and in vivo. Chrysin inhibited passive cutaneous anaphylaxis and systemic pseudo-allergy in vivo. LAD2 cell degranulation, calcium ion (Ca2+) influx, and adenosine 5'-triphosphate (ATP) content were significantly suppressed in a dose-dependent manner. Chrysin suppressed pseudo-allergic reactions through the PLC/IP3/Ca2+ and ERK/STAT3 serine 727 pathways downstream of MrgX2. Therefore, mitochondrial ATP, but not glycolysis, is vital for pseudo-allergic reactions mediated by MrgX2. This study provides new insights for the treatment of pseudo-allergy.
Collapse
Affiliation(s)
- Yongjing Zhang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710049, China
| | - Zhuoyin Xue
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710049, China
| | - Shiling Hu
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710049, China
| | - Haoyun Bai
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710049, China
| | - Jue Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710049, China
| | - Nan Wang
- School of Pharmacy, Xi'an Jiaotong University, Xi'an 710049, China
- Pazhou Lab, Guangzhou 510330, China
| |
Collapse
|
88
|
Schmidt-Arras D, Rose-John S. Endosomes as Signaling Platforms for IL-6 Family Cytokine Receptors. Front Cell Dev Biol 2021; 9:688314. [PMID: 34141712 PMCID: PMC8204807 DOI: 10.3389/fcell.2021.688314] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 04/28/2021] [Indexed: 12/12/2022] Open
Abstract
Interleukin-6 (IL-6) is the name-giving cytokine of a family of eleven members, including IL-6, CNTF, LIF, and IL-27. IL-6 was first recognized as a B-cell stimulating factor but we now know that the cytokine plays a pivotal role in the orchestration of inflammatory processes as well as in inflammation associated cancer. Moreover, IL-6 is involved in metabolic regulation and it has been shown to be involved in major neural activities such as neuroprotection, which can help to repair and to reduce brain damage. Receptor complexes of all members formed at the plasma membrane contain one or two molecules of the signaling receptor subunit GP130 and the mechanisms of signal transduction are well understood. IL-6 type cytokines can also signal from endomembranes, in particular the endosome, and situations have been reported in which endocytosis of receptor complexes are a prerequisite of intracellular signaling. Moreover, pathogenic GP130 variants were shown to interfere with spatial activation of downstream signals. We here summarize the molecular mechanisms underlying spatial regulation of IL-6 family cytokine signaling and discuss its relevance for pathogenic processes.
Collapse
Affiliation(s)
- Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Kiel, Germany
| |
Collapse
|
89
|
Wang J, Zhao Q, Bowden J, Hemani G, Davey Smith G, Small DS, Zhang NR. Causal inference for heritable phenotypic risk factors using heterogeneous genetic instruments. PLoS Genet 2021; 17:e1009575. [PMID: 34157017 PMCID: PMC8301661 DOI: 10.1371/journal.pgen.1009575] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2020] [Revised: 07/23/2021] [Accepted: 05/04/2021] [Indexed: 12/25/2022] Open
Abstract
Over a decade of genome-wide association studies (GWAS) have led to the finding of extreme polygenicity of complex traits. The phenomenon that "all genes affect every complex trait" complicates Mendelian Randomization (MR) studies, where natural genetic variations are used as instruments to infer the causal effect of heritable risk factors. We reexamine the assumptions of existing MR methods and show how they need to be clarified to allow for pervasive horizontal pleiotropy and heterogeneous effect sizes. We propose a comprehensive framework GRAPPLE to analyze the causal effect of target risk factors with heterogeneous genetic instruments and identify possible pleiotropic patterns from data. By using GWAS summary statistics, GRAPPLE can efficiently use both strong and weak genetic instruments, detect the existence of multiple pleiotropic pathways, determine the causal direction and perform multivariable MR to adjust for confounding risk factors. With GRAPPLE, we analyze the effect of blood lipids, body mass index, and systolic blood pressure on 25 disease outcomes, gaining new information on their causal relationships and potential pleiotropic pathways involved.
Collapse
Affiliation(s)
- Jingshu Wang
- Department of Statistics, University of Chicago, Chicago, Illinois, United States of America
| | - Qingyuan Zhao
- Department of Pure Mathematics and Mathematical Statistics, University of Cambridge, Cambridge, United Kingdom
| | - Jack Bowden
- College of Medicine and Health, University of Exeter, Exeter, United Kingdom
| | - Gibran Hemani
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - George Davey Smith
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, United Kingdom
| | - Dylan S. Small
- Department of Statistics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| | - Nancy R. Zhang
- Department of Statistics, University of Pennsylvania, Philadelphia, Pennsylvania, United States of America
| |
Collapse
|
90
|
The two facets of gp130 signalling in liver tumorigenesis. Semin Immunopathol 2021; 43:609-624. [PMID: 34047814 PMCID: PMC8443519 DOI: 10.1007/s00281-021-00861-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 04/28/2021] [Indexed: 02/06/2023]
Abstract
The liver is a vital organ with multiple functions and a large regenerative capacity. Tumours of the liver are the second most frequently cause of cancer-related death and develop in chronically inflamed livers. IL-6-type cytokines are mediators of inflammation and almost all members signal via the receptor subunit gp130 and the downstream signalling molecule STAT3. We here summarize current knowledge on how gp130 signalling and STAT3 in tumour cells and cells of the tumour micro-environment drives hepatic tumorigenesis. We furthermore discuss very recent findings describing also anti-tumorigenic roles of gp130/STAT3 and important considerations for therapeutic interventions.
Collapse
|
91
|
Rose-John S. Therapeutic targeting of IL-6 trans-signaling. Cytokine 2021; 144:155577. [PMID: 34022535 DOI: 10.1016/j.cyto.2021.155577] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/04/2021] [Accepted: 05/06/2021] [Indexed: 02/06/2023]
Abstract
Interleukin-6 (IL-6) is a cytokine, which is involved in innate and acquired immunity, in neural cell maintenance and in metabolism. IL-6 can be synthesized by many different cells including myeloid cells, fibroblasts, endothelial cells and lymphocytes. The synthesis of IL-6 is strongly stimulated by Toll like receptors and by IL-1. Therefore, IL-6 levels in the body are high during infection and inflammatory processes. Moreover, IL-6 is a prominent growth factor of tumor cells and plays a major role in inflammation associated cancer. On target cells, IL-6 binds to an IL-6 receptor, which is not signaling competent. The complex of IL-6 and IL-6 receptor associate with a second receptor subunit, glycoprotein gp130, which dimerizes and initiates intracellular signaling. Cells, which do not express the IL-6 receptor are not responsive to IL-6. They can, however, be stimulated by the complex of IL-6 and a soluble form of the IL-6 receptor, which is generated by limited proteolysis and to a lesser extent by translation from an alternatively spliced mRNA. This process has been named IL-6 trans-signaling. This review article will explain the biology of IL-6 trans-signaling and the specific inhibition of this mode of signaling, which has been recognized to be fundamental in inflammation and cancer.
Collapse
|
92
|
Zandstra J, Jongerius I, Kuijpers TW. Future Biomarkers for Infection and Inflammation in Febrile Children. Front Immunol 2021; 12:631308. [PMID: 34079538 PMCID: PMC8165271 DOI: 10.3389/fimmu.2021.631308] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Accepted: 04/12/2021] [Indexed: 01/08/2023] Open
Abstract
Febrile patients, suffering from an infection, inflammatory disease or autoimmunity may present with similar or overlapping clinical symptoms, which makes early diagnosis difficult. Therefore, biomarkers are needed to help physicians form a correct diagnosis and initiate the right treatment to improve patient outcomes following first presentation or admittance to hospital. Here, we review the landscape of novel biomarkers and approaches of biomarker discovery. We first discuss the use of current plasma parameters and whole blood biomarkers, including results obtained by RNA profiling and mass spectrometry, to discriminate between bacterial and viral infections. Next we expand upon the use of biomarkers to distinguish between infectious and non-infectious disease. Finally, we discuss the strengths as well as the potential pitfalls of current developments. We conclude that the use of combination tests, using either protein markers or transcriptomic analysis, have advanced considerably and should be further explored to improve current diagnostics regarding febrile infections and inflammation. If proven effective when combined, these biomarker signatures will greatly accelerate early and tailored treatment decisions.
Collapse
Affiliation(s)
- Judith Zandstra
- Division Research and Landsteiner Laboratory, Department of Immunopathology, Sanquin Blood Supply, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Ilse Jongerius
- Division Research and Landsteiner Laboratory, Department of Immunopathology, Sanquin Blood Supply, Amsterdam University Medical Center (UMC), Amsterdam, Netherlands
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
| | - Taco W. Kuijpers
- Department of Pediatric Immunology, Rheumatology and Infectious Diseases, Emma Children’s Hospital, Amsterdam UMC, Amsterdam, Netherlands
- Division Research and Landsteiner Laboratory, Department of Blood Cell Research, Sanquin Blood Supply, Amsterdam UMC, Amsterdam, Netherlands
| |
Collapse
|
93
|
Fadil I, Ben-Ali M, Jeddane L, Barbouche MR, Bousfiha AA. The Seven STAT3-Related Hyper-IgE Syndromes. J Clin Immunol 2021; 41:1384-1389. [PMID: 33903995 DOI: 10.1007/s10875-021-01041-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 04/12/2021] [Indexed: 11/24/2022]
Affiliation(s)
- Ilham Fadil
- Laboratory of Clinical Immunology, Inflammation, and Allergy (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Meriem Ben-Ali
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, University Tunis El-Manar, Tunis, Tunisia
| | - Leila Jeddane
- Laboratory of Clinical Immunology, Inflammation, and Allergy (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco.,Immunology-Allergology Unit, National Reference Laboratory, University Mohammed VI of Health Sciences, Casablanca, Morocco
| | - Mohamed-Ridha Barbouche
- Laboratory of Transmission, Control and Immunobiology of Infections, Institut Pasteur de Tunis, University Tunis El-Manar, Tunis, Tunisia.,Faculty of Medicine of Tunis, University Tunis El-Manar, Tunis, Tunisia
| | - Ahmed Aziz Bousfiha
- Laboratory of Clinical Immunology, Inflammation, and Allergy (LICIA), Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco. .,Clinical Immunology Unit, Department of Infectious Diseases, Harouchi Hospital, Ibn Rochd University Hospital, Casablanca, Morocco.
| |
Collapse
|
94
|
Lu HY, Turvey SE. Human MALT1 deficiency and predisposition to infections. Curr Opin Immunol 2021; 72:1-12. [PMID: 33714841 DOI: 10.1016/j.coi.2021.02.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/18/2021] [Accepted: 02/26/2021] [Indexed: 12/20/2022]
Abstract
Human germline MALT1 deficiency is an inborn error of immunity characterized by recurrent bacterial, viral, and fungal infections, periodontal disease, enteropathy, dermatitis, and failure to thrive. The number of identified MALT1-deficient patients have greatly increased in the past two years, which has significantly improved our understanding of the clinical features of this disorder. Patients frequently experience infections affecting the respiratory, skin, gastrointestinal, and blood systems. The most frequently detected pathogens are Staphylococcus aureus, Candida albicans, and cytomegalovirus. Enhanced susceptibility to S. aureus and C. albicans is likely due to impaired Th17 immunity, similar to STAT3 and IL-17 pathway deficiencies.
Collapse
Affiliation(s)
- Henry Y Lu
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada
| | - Stuart E Turvey
- Department of Pediatrics, British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
95
|
Ishitsuka Y, Hanaoka Y, Tanemura A, Fujimoto M. Cutaneous Squamous Cell Carcinoma in the Age of Immunotherapy. Cancers (Basel) 2021; 13:1148. [PMID: 33800195 PMCID: PMC7962464 DOI: 10.3390/cancers13051148] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/02/2021] [Accepted: 03/03/2021] [Indexed: 12/11/2022] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) is the second most prevalent skin cancer globally. Because most cSCC cases are manageable by local excision/radiotherapy and hardly become life-threatening, they are often excluded from cancer registries in most countries. Compared with cutaneous melanoma that originates from the melanin-producing, neural crest-derived epidermal resident, keratinocyte (KC)-derived cancers are influenced by the immune system with regards to their pathogenetic behaviour. Congenital or acquired immunosurveillance impairments compromise tumoricidal activity and raises cSCC incidence rates. Intriguingly, expanded applications of programmed death-1 (PD-1) blockade therapies have revealed cSCC to be one of the most amenable targets, particularly when compared with the mucosal counterparts arisen in the esophagus or the cervix. The clinical observation reminds us that cutaneous tissue has a peculiarly high immunogenicity that can evoke tumoricidal recall responses topically. Here we attempt to redefine cSCC biology and review current knowledge about cSCC from multiple viewpoints that involve epidemiology, clinicopathology, molecular genetics, molecular immunology, and developmental biology. This synthesis not only underscores the primal importance of the immune system, rather than just a mere accumulation of ultraviolet-induced mutations but also reinforces the following hypothesis: PD-1 blockade effectively restores the immunity specially allowed to exist within the fully cornified squamous epithelium, that is, the epidermis.
Collapse
Affiliation(s)
- Yosuke Ishitsuka
- Department of Dermatology Integrated Medicine, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka 565-0871, Japan; (Y.H.); (A.T.); (M.F.)
| | | | | | | |
Collapse
|
96
|
Saikia B, Rawat A, Minz RW, Suri D, Pandiarajan V, Jindal A, Sahu S, Karim A, Desai M, Taur PD, Pandrowala A, Gowri V, Madkaikar M, Dalvi A, Yadav RM, Lashkari HP, Raj R, Uppuluri R, Swaminathan VV, Bhattad S, Cyril G, Kumar H, Shukla A, Kalra M, Govindaraj G, Singh S. Clinical Profile of Hyper-IgE Syndrome in India. Front Immunol 2021; 12:626593. [PMID: 33717144 PMCID: PMC7952512 DOI: 10.3389/fimmu.2021.626593] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 02/05/2021] [Indexed: 11/13/2022] Open
Abstract
Introduction: Hyper-IgE Syndrome (HIES) is a rare inborn error of immunity (IEI) characterized by a constellation of symptoms related to susceptibility to Staphylococcal skin and pulmonary infections, eczema, raised serum IgE (>2,000 IU/ml), craniofacial anomalies, and recurrent bone fractures. Data on HIES from the Indian subcontinent is scarce and restricted to small case series and case reports. This is the first compilation of national data on HIES. Materials and Methods: A total 103 cases clinically diagnosed and treated as HIES were analyzed from nine centers. Cases with clinical and/or molecular diagnosis of DOCK8 deficiency were not included. Patients were divided into two groups: group I for whom a heterozygous rare variant of STAT3 was identified, and group II, with clinical features similar to those of AD STAT3 deficiency, but without any genetic diagnosis. Results: Genetic diagnosis was available in 27 patients (26.2%) and all harbored rare variants in the STAT3 gene. Majority of these STAT3 HIES patients presented with recurrent skin abscesses (77.7%) or pneumonia (62.9%) or both (59.2%). Other features included eczema (37%), candidiasis (55.5%), facial dysmorphism (55.5%), recurrent fractures (11.1%), and retained primary teeth (7.4%). Mycobacterial infections were seen in a significant 18.5%. Mortality was seen in three subjects (11.1%). A similar trend in the clinical presentation was observed when all the 103 patients were analyzed together. Twenty percent of patients without a rare variant in the STAT3 gene had an NIH score of ≥40, whereas, 51.9% of STAT3 HIES subjects had scores below the cut off of ≥40. TH17 cell numbers were low in 10/11 (90.9%) STAT3 HIES tested. Rare variants observed were 8 in exon 21; 8 in exon 13; 3 in exon 10; 2 in exon 15, and one each in exon 6, 16, 17, 19, 22, and splice site downstream of exon 12. Seven variants were novel and included F174S, N567D, L404Sfs*8, G419 =, M329K, T714I, R518X, and a splice site variant downstream of exon 12. Conclusions: The report includes seven novel STAT3 variants, including a rare linker domain nonsense variant and a CC domain variant. Mycobacterial diseases were more frequent, compared to western literature.
Collapse
Affiliation(s)
- Biman Saikia
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Amit Rawat
- Department of Pediatrics, Advanced Pediatric Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ranjana W Minz
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Deepti Suri
- Department of Pediatrics, Advanced Pediatric Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Vignesh Pandiarajan
- Department of Pediatrics, Advanced Pediatric Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Ankur Jindal
- Department of Pediatrics, Advanced Pediatric Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Smrity Sahu
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Adil Karim
- Department of Immunopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - Mukesh Desai
- Department of Immunology, BJ Wadia Hospital for Children, Mumbai, India
| | - Prasad D Taur
- Department of Immunology, BJ Wadia Hospital for Children, Mumbai, India
| | | | - Vijaya Gowri
- Department of Immunology, BJ Wadia Hospital for Children, Mumbai, India
| | | | - Aparna Dalvi
- ICMR-National Institute of Immunohaematology, Mumbai, India
| | | | | | - Revathi Raj
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Ramya Uppuluri
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | - Venkateswaran V Swaminathan
- Department of Pediatric Hematology, Oncology, Blood and Marrow Transplantation, Apollo Hospitals, Chennai, India
| | | | | | | | | | | | | | - Surjit Singh
- Department of Pediatrics, Advanced Pediatric Centre, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
97
|
Inborn errors of immunity with atopic phenotypes: A practical guide for allergists. World Allergy Organ J 2021; 14:100513. [PMID: 33717395 PMCID: PMC7907539 DOI: 10.1016/j.waojou.2021.100513] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 12/22/2020] [Accepted: 01/11/2021] [Indexed: 12/19/2022] Open
Abstract
Inborn errors of immunity (IEI) are a heterogeneous group of disorders, mainly resulting from mutations in genes associated with immunoregulation and immune host defense. These disorders are characterized by different combinations of recurrent infections, autoimmunity, inflammatory manifestations, lymphoproliferation, and malignancy. Interestingly, it has been increasingly observed that common allergic symptoms also can represent the expression of an underlying immunodeficiency and/or immune dysregulation. Very high IgE levels, peripheral or organ-specific hypereosinophilia, usually combined with a variety of atopic symptoms, may sometimes be the epiphenomenon of a monogenic disease. Therefore, allergists should be aware that severe and/or therapy-resistant atopic disorders might be the main clinical phenotype of some IEI. This could pave the way to target therapies, leading to better quality of life and improved survival in affected patients.
Collapse
|
98
|
Tangye SG, Al-Herz W, Bousfiha A, Cunningham-Rundles C, Franco JL, Holland SM, Klein C, Morio T, Oksenhendler E, Picard C, Puel A, Puck J, Seppänen MRJ, Somech R, Su HC, Sullivan KE, Torgerson TR, Meyts I. The Ever-Increasing Array of Novel Inborn Errors of Immunity: an Interim Update by the IUIS Committee. J Clin Immunol 2021; 41:666-679. [PMID: 33598806 PMCID: PMC7889474 DOI: 10.1007/s10875-021-00980-1] [Citation(s) in RCA: 152] [Impact Index Per Article: 50.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 01/20/2021] [Indexed: 12/12/2022]
Abstract
The most recent updated classification of inborn errors of immunity/primary immunodeficiencies, compiled by the International Union of Immunological Societies Expert Committee, was published in January 2020. Within days of completing this report, it was already out of date, evidenced by the frequent publication of genetic variants proposed to cause novel inborn errors of immunity. As the next formal report from the IUIS Expert Committee will not be published until 2022, we felt it important to provide the community with a brief update of recent contributions to the field of inborn errors of immunity. Herein, we highlight studies that have identified 26 additional monogenic gene defects that reach the threshold to represent novel causes of immune defects.
Collapse
Affiliation(s)
- Stuart G Tangye
- Garvan Institute of Medical Research, Darlinghurst, Sydney, New South Wales, 2010, Australia. .,Faculty of Medicine, St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia.
| | - Waleed Al-Herz
- Department of Pediatrics, Faculty of Medicine, Kuwait University, Kuwait City, Kuwait
| | - Aziz Bousfiha
- Laboratoire d'Immunologie Clinique, d'Inflammation et d'Allergy LICIA Clinical Immunology Unit, Casablanca Children's Hospital, Ibn Rochd Medical School, King Hassan II University, Casablanca, Morocco
| | | | - Jose Luis Franco
- Grupo de Inmunodeficiencias Primarias, Facultad de Medicina, Universidad de Antioquia UdeA, Medellin, Colombia
| | - Steven M Holland
- Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Christoph Klein
- Dr von Hauner Childrens Hospital, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Tomohiro Morio
- Department of Pediatrics and Developmental Biology, Tokyo Medical and Dental University, Tokyo, Japan
| | - Eric Oksenhendler
- Department of Clinical Immunology, Hôpital Saint-Louis, APHP, University Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Capucine Picard
- Study Center for Primary Immunodeficiencies, Necker Hospital for Sick Children, APHP, Paris, France.,Laboratory of Lymphocyte Activation and Susceptibility to EBV, INSERM UMR1163, Imagine Institute, Necker Hospital for Sick Children, Paris University, Paris, France
| | - Anne Puel
- Laboratory of Human Genetics of Infectious Diseases, INSERM U1163, Necker Hospital, 75015, Paris, France.,Imagine Institute, University of Paris, 75015, Paris, France
| | - Jennifer Puck
- Department of Pediatrics, University of California San Francisco and UCSF Benioff Children's Hospital, San Francisco, CA, USA
| | - Mikko R J Seppänen
- Adult Immunodeficiency Unit, Infectious Diseases, Inflammation Center and Rare Diseases Center, Childrens Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Raz Somech
- Pediatric Department and Immunology Unit, Sheba Medical Center, Tel Aviv, Israel
| | - Helen C Su
- Laboratory of Clinical Immunology & Microbiology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Kathleen E Sullivan
- Division of Allergy Immunology, Department of Pediatrics, Childrens Hospital of Philadelphia, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | | | - Isabelle Meyts
- Department of Immunology and Microbiology, Laboratory for Inborn Errors of Immunity, Department of Pediatrics, University Hospitals Leuven and KU Leuven, 3000, Leuven, Belgium
| |
Collapse
|
99
|
Dmitrieva NI, Walts AD, Nguyen DP, Grubb A, Zhang X, Wang X, Ping X, Jin H, Yu Z, Yu ZX, Yang D, Schwartzbeck R, Dalgard CL, Kozel BA, Levin MD, Knutsen RH, Liu D, Milner JD, López DB, O'Connell MP, Lee CCR, Myles IA, Hsu AP, Freeman AF, Holland SM, Chen G, Boehm M. Impaired angiogenesis and extracellular matrix metabolism in autosomal-dominant hyper-IgE syndrome. J Clin Invest 2021; 130:4167-4181. [PMID: 32369445 DOI: 10.1172/jci135490] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Accepted: 04/29/2020] [Indexed: 12/21/2022] Open
Abstract
There are more than 7000 described rare diseases, most lacking specific treatment. Autosomal-dominant hyper-IgE syndrome (AD-HIES, also known as Job's syndrome) is caused by mutations in STAT3. These patients present with immunodeficiency accompanied by severe nonimmunological features, including skeletal, connective tissue, and vascular abnormalities, poor postinfection lung healing, and subsequent pulmonary failure. No specific therapies are available for these abnormalities. Here, we investigated underlying mechanisms in order to identify therapeutic targets. Histological analysis of skin wounds demonstrated delayed granulation tissue formation and vascularization during skin-wound healing in AD-HIES patients. Global gene expression analysis in AD-HIES patient skin fibroblasts identified deficiencies in a STAT3-controlled transcriptional network regulating extracellular matrix (ECM) remodeling and angiogenesis, with hypoxia-inducible factor 1α (HIF-1α) being a major contributor. Consistent with this, histological analysis of skin wounds and coronary arteries from AD-HIES patients showed decreased HIF-1α expression and revealed abnormal organization of the ECM and altered formation of the coronary vasa vasorum. Disease modeling using cell culture and mouse models of angiogenesis and wound healing confirmed these predicted deficiencies and demonstrated therapeutic benefit of HIF-1α-stabilizing drugs. The study provides mechanistic insights into AD-HIES pathophysiology and suggests potential treatment options for this rare disease.
Collapse
Affiliation(s)
- Natalia I Dmitrieva
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Avram D Walts
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Dai Phuong Nguyen
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Alex Grubb
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Xue Zhang
- Bioinformatics and Systems Biology Core, and
| | - Xujing Wang
- Bioinformatics and Systems Biology Core, and
| | - Xianfeng Ping
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Hui Jin
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Zhen Yu
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Zu-Xi Yu
- Pathology Core, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Dan Yang
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Robin Schwartzbeck
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Clifton L Dalgard
- Department of Anatomy, Physiology & Genetics.,The American Genome Center, and.,Collaborative Health Initiative Research Program, Uniformed Services University of the Health Sciences, Bethesda, Maryland, USA
| | - Beth A Kozel
- Laboratory of Vascular and Matrix Genetics, NHLBI
| | - Mark D Levin
- Laboratory of Vascular and Matrix Genetics, NHLBI
| | | | - Delong Liu
- Laboratory of Vascular and Matrix Genetics, NHLBI
| | - Joshua D Milner
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Diego B López
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Michael P O'Connell
- Laboratory of Allergic Diseases, National Institute of Allergy and Infectious Diseases (NIAID)
| | - Chyi-Chia Richard Lee
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute (NCI), and
| | - Ian A Myles
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, USA
| | - Amy P Hsu
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, USA
| | - Alexandra F Freeman
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, USA
| | - Steven M Holland
- Laboratory of Clinical Immunology and Microbiology, NIAID, NIH, Bethesda, Maryland, USA
| | - Guibin Chen
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| | - Manfred Boehm
- Laboratory of Cardiovascular Regenerative Medicine, Translational Vascular Medicine Branch
| |
Collapse
|
100
|
Schumacher N, Yan K, Gandraß M, Müller M, Krisp C, Häsler R, Carambia A, Nofer JR, Bernardes JP, Khouja M, Thomsen I, Chalupsky K, Bolik J, Hölscher C, Wunderlich T, Herkel J, Rosenstiel P, Schramm C, Schlüter H, Renné T, Mittrücker HW, Rose-John S, Schmidt-Arras D. Cell-autonomous hepatocyte-specific GP130 signaling is sufficient to trigger a robust innate immune response in mice. J Hepatol 2021; 74:407-418. [PMID: 32987028 DOI: 10.1016/j.jhep.2020.09.021] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2020] [Revised: 09/11/2020] [Accepted: 09/14/2020] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Interleukin (IL)-6 cytokine family members contribute to inflammatory and regenerative processes. Engagement of the signaling receptor subunit gp130 is common to almost all members of the family. In the liver, all major cell types respond to IL-6-type cytokines, making it difficult to delineate cell type-specific effects. We therefore generated mouse models for liver cell type-specific analysis of IL-6 signaling. METHODS We produced mice with a Cre-inducible expression cassette encoding a designed pre-dimerized constitutive active gp130 variant. We bred these mice to different Cre-drivers to induce transgenic gp130 signaling in distinct liver cell types: hepatic stellate cells, cholangiocytes/liver progenitor cells or hepatocytes. We phenotyped these mice using multi-omics approaches, immunophenotyping and a bacterial infection model. RESULTS Hepatocyte-specific gp130 activation led to the upregulation of innate immune system components, including acute-phase proteins. Consequently, we observed peripheral mobilization and recruitment of myeloid cells to the liver. Hepatic myeloid cells, including liver-resident Kupffer cells were instructed to adopt a bactericidal phenotype which ultimately conferred enhanced resistance to bacterial infection in these mice. We demonstrate that persistent hepatocyte-specific gp130 activation resulted in amyloid A amyloidosis in aged mice. In contrast, we did not observe overt effects of hepatic stellate cell- or cholangiocyte/liver progenitor cell-specific transgenic gp130 signaling. CONCLUSIONS Hepatocyte-specific gp130 activation alone is sufficient to trigger a robust innate immune response in the absence of NF-κB activation. We therefore conclude that gp130 engagement, e.g. by IL-6 trans-signaling, represents a safe-guard mechanism in innate immunity. LAY SUMMARY Members of the interleukin-6 cytokine family signal via the receptor subunit gp130 and are involved in multiple processes in the liver. However, as several liver cell types respond to interleukin-6 family cytokines, it is difficult to delineate cell type-specific effects. Using a novel mouse model, we provide evidence that hepatocyte-specific gp130 activation is sufficient to trigger a robust systemic innate immune response.
Collapse
Affiliation(s)
- Neele Schumacher
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Karsten Yan
- Institute of Immunology, University Medical Center Hamburg-Eppendorf, Germany
| | - Monja Gandraß
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Miryam Müller
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Christoph Krisp
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Robert Häsler
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Germany
| | - Antonella Carambia
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Germany
| | - Jerzy-Roch Nofer
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Joanna P Bernardes
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Germany
| | - Mouhamad Khouja
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Ilka Thomsen
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Karel Chalupsky
- Laboratory of Transgenic Models of Diseases, Institute of Molecular Genetics of the ASCR, Prague, Czech Republic
| | - Julia Bolik
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Christoph Hölscher
- Infection Immunology, Research Center Borstel, Leibniz Lung Center, Germany
| | | | - Johannes Herkel
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Germany
| | - Philip Rosenstiel
- Institute of Clinical Molecular Biology, Christian-Albrechts-University Kiel, Germany
| | - Christoph Schramm
- Department of Medicine I, University Medical Center Hamburg-Eppendorf, Germany; Martin Zeitz Center for Rare Diseases
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Germany
| | | | - Stefan Rose-John
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany
| | - Dirk Schmidt-Arras
- Institute of Biochemistry, Christian-Albrechts-University Kiel, Germany.
| |
Collapse
|